1
|
Yasarbas SS, Inal E, Yildirim MA, Dubrac S, Lamartine J, Mese G. Connexins in epidermal health and diseases: insights into their mutations, implications, and therapeutic solutions. Front Physiol 2024; 15:1346971. [PMID: 38827992 PMCID: PMC11140265 DOI: 10.3389/fphys.2024.1346971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/15/2024] [Indexed: 06/05/2024] Open
Abstract
The epidermis, the outermost layer of the skin, serves as a protective barrier against external factors. Epidermal differentiation, a tightly regulated process essential for epidermal homeostasis, epidermal barrier formation and skin integrity maintenance, is orchestrated by several players, including signaling molecules, calcium gradient and junctional complexes such as gap junctions (GJs). GJ proteins, known as connexins facilitate cell-to-cell communication between adjacent keratinocytes. Connexins can function as either hemichannels or GJs, depending on their interaction with other connexons from neighboring keratinocytes. These channels enable the transport of metabolites, cAMP, microRNAs, and ions, including Ca2+, across cell membranes. At least ten distinct connexins are expressed within the epidermis and mutations in at least five of them has been linked to various skin disorders. Connexin mutations may cause aberrant channel activity by altering their synthesis, their gating properties, their intracellular trafficking, and the assembly of hemichannels and GJ channels. In addition to mutations, connexin expression is dysregulated in other skin conditions including psoriasis, chronic wound and skin cancers, indicating the crucial role of connexins in skin homeostasis. Current treatment options for conditions with mutant or altered connexins are limited and primarily focus on symptom management. Several therapeutics, including non-peptide chemicals, antibodies, mimetic peptides and allele-specific small interfering RNAs are promising in treating connexin-related skin disorders. Since connexins play crucial roles in maintaining epidermal homeostasis as shown with linkage to a range of skin disorders and cancer, further investigations are warranted to decipher the molecular and cellular alterations within cells due to mutations or altered expression, leading to abnormal proliferation and differentiation. This would also help characterize the roles of each isoform in skin homeostasis, in addition to the development of innovative therapeutic interventions. This review highlights the critical functions of connexins in the epidermis and the association between connexins and skin disorders, and discusses potential therapeutic options.
Collapse
Affiliation(s)
- S. Suheda Yasarbas
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Izmir, Turkiye
| | - Ece Inal
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Izmir, Turkiye
| | - M. Azra Yildirim
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Izmir, Turkiye
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jérôme Lamartine
- Skin Functional Integrity Group, Laboratory for Tissue Biology and Therapeutics Engineering (LBTI) CNRS UMR5305, University of Lyon, Lyon, France
| | - Gulistan Mese
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Izmir, Turkiye
| |
Collapse
|
2
|
Jang DG, Kwon KY, Kweon YC, Kim BG, Myung K, Lee HS, Young Park C, Kwon T, Park TJ. GJA1 depletion causes ciliary defects by affecting Rab11 trafficking to the ciliary base. eLife 2022; 11:81016. [PMID: 36004726 PMCID: PMC9448326 DOI: 10.7554/elife.81016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
The gap junction complex functions as a transport channel across the membrane. Among gap junction subunits, gap junction protein α1 (GJA1) is the most commonly expressed subunit. A recent study showed that GJA1 is necessary for the maintenance of motile cilia; however, the molecular mechanism and function of GJA1 in ciliogenesis remain unknown. Here, we examined the functions of GJA1 during ciliogenesis in human retinal pigment epithelium-1 and Xenopus laevis embryonic multiciliated-cells. GJA1 localizes to the motile ciliary axonemes or pericentriolar regions beneath the primary cilium. GJA1 depletion caused malformation of both the primary cilium and motile cilia. Further study revealed that GJA1 depletion affected several ciliary proteins such as BBS4, CP110, and Rab11 in the pericentriolar region and basal body. Interestingly, CP110 removal from the mother centriole was significantly reduced by GJA1 depletion. Importantly, Rab11, a key regulator during ciliogenesis, was immunoprecipitated with GJA1, and GJA1 knockdown caused the mislocalization of Rab11. These findings suggest that GJA1 regulates ciliogenesis by interacting with the Rab11-Rab8 ciliary trafficking pathway.
Collapse
Affiliation(s)
- Dong Gil Jang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Keun Yeong Kwon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yeong Cheon Kweon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Byung-Gyu Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Chan Young Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Taejoon Kwon
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Tae Joo Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
3
|
Novielli-Kuntz NM, Press ER, Barr K, Prado MAM, Laird DW. Mutant Cx30-A88V mice exhibit hydrocephaly and sex-dependent behavioral abnormalities, implicating a functional role for Cx30 in the brain. Dis Model Mech 2021; 14:14/1/dmm046235. [PMID: 33735099 PMCID: PMC7859702 DOI: 10.1242/dmm.046235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/03/2020] [Indexed: 01/01/2023] Open
Abstract
Connexin 30 (Cx30; also known as Gjb6 when referring to the mouse gene) is expressed in ependymal cells of the brain ventricles, in leptomeningeal cells and in astrocytes rich in connexin 43 (Cx43), leading us to question whether patients harboring GJB6 mutations exhibit any brain anomalies. Here, we used mice harboring the human disease-associated A88V Cx30 mutation to address this gap in knowledge. Brain Cx30 levels were lower in male and female Cx30A88V/A88V mice compared with Cx30A88V/+ and Cx30+/+ mice, whereas Cx43 levels were lower only in female Cx30 mutant mice. Characterization of brain morphology revealed a disrupted ependymal cell layer, significant hydrocephalus and enlarged ventricles in 3- to 6-month-old adult male and female Cx30A88V/A88V mice compared with Cx30A88V/+ or Cx30+/+ sex-matched littermate mice. To determine the functional significance of these molecular and morphological changes, we investigated a number of behavioral activities in these mice. Interestingly, only female Cx30A88V/A88V mice exhibited abnormal behavior compared with all other groups. Cx30A88V/A88V female mice demonstrated increased locomotor and exploratory activity in both the open field and the elevated plus maze. They also exhibited dramatically reduced ability to learn the location of the escape platform during Morris water maze training, although they were able to swim as well as other genotypes. Our findings suggest that the homozygous A88V mutation in Cx30 causes major morphological changes in the brain of aging mice, possibly attributable to an abnormal ependymal cell layer. Remarkably, these changes had a more pronounced consequence for cognitive function in female mice, which is likely to be linked to the dysregulation of both Cx30 and Cx43 levels in the brain.
Collapse
Affiliation(s)
- Nicole M Novielli-Kuntz
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON, Canada N6A 5C1
| | - Eric R Press
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON, Canada N6A 5C1.,Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada N6A 5C1
| | - Kevin Barr
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON, Canada N6A 5C1
| | - Marco A M Prado
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON, Canada N6A 5C1.,Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada N6A 5C1.,Robarts Research Institute, The University of Western Ontario, London, ON, Canada N6A 5K8
| | - Dale W Laird
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON, Canada N6A 5C1 .,Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada N6A 5C1
| |
Collapse
|
4
|
Au A, Shao Q, White KK, Lucaciu SA, Esseltine JL, Barr K, Laird DW. Comparative Analysis of Cx31 and Cx43 in Differentiation-Competent Rodent Keratinocytes. Biomolecules 2020; 10:biom10101443. [PMID: 33066499 PMCID: PMC7602205 DOI: 10.3390/biom10101443] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/30/2020] [Accepted: 10/08/2020] [Indexed: 12/25/2022] Open
Abstract
When considering connexin expression and regulation, the epidermis of the skin is one of the most complex tissues found in mammals even though it largely contains a single cell type, the keratinocyte. In the rodent epidermis, up to 9 connexin family members have been detected at the mRNA level. Many of these connexins are temporally and spatially regulated in coordination with keratinocyte progenitor cell differentiation and migration from the stratum basale to form the stratum spinosum and stratum granulosum layers before finally forming the stratum corneum. Cx43 is the principal connexin found in basal keratinocytes and to a lesser degree found in keratinocytes that have begun to differentiate where Cx26, Cx30 and Cx31 become prevalent. Here we show that the CRISPR-Cas9 ablation of Cx43 reduces overall gap junction coupling in monolayer cultures of rat epidermal keratinocytes (REKs) and dysregulates the differentiation of REKs when grown in organotypic cultures. Natively found in differentiated keratinocytes, Cx31 readily assembles into gap junctions when expressed in REKs where it can extensively co-assemble into the same gap junctions with co-expressed Cx30. Time-lapse imaging indicated that many Cx31 gap junctions are mobile within the plasma membrane undergoing both fusion and fission events. Finally, the persistence of pre-existing Cx31 gap junctions in the presence of the protein trafficking blocker, brefeldin A, is longer than that found for Cx43 gap junctions indicating that it has a distinctly different life expectancy in REKs. Collectively, this study highlights the importance of Cx43 in rodent keratinocyte differentiation and suggests that Cx31 acquires life-cycle properties that are distinct from Cx43.
Collapse
Affiliation(s)
- Akina Au
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON N6A 5C1, Canada; (A.A.); (S.A.L.)
| | - Qing Shao
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON N6A 5C1, Canada; (Q.S.); (K.K.W.); (K.B.)
| | - Kyra K. White
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON N6A 5C1, Canada; (Q.S.); (K.K.W.); (K.B.)
| | - Sergiu A. Lucaciu
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON N6A 5C1, Canada; (A.A.); (S.A.L.)
| | - Jessica L. Esseltine
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada;
| | - Kevin Barr
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON N6A 5C1, Canada; (Q.S.); (K.K.W.); (K.B.)
| | - Dale W. Laird
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON N6A 5C1, Canada; (A.A.); (S.A.L.)
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, ON N6A 5C1, Canada; (Q.S.); (K.K.W.); (K.B.)
- Correspondence: ; Tel.: +1-519-661-2111 (ext. 86827)
| |
Collapse
|
5
|
Brecker M, Khakhina S, Schubert TJ, Thompson Z, Rubenstein RC. The Probable, Possible, and Novel Functions of ERp29. Front Physiol 2020; 11:574339. [PMID: 33013490 PMCID: PMC7506106 DOI: 10.3389/fphys.2020.574339] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022] Open
Abstract
The luminal endoplasmic reticulum (ER) protein of 29 kDa (ERp29) is a ubiquitously expressed cellular agent with multiple critical roles. ERp29 regulates the biosynthesis and trafficking of several transmembrane and secretory proteins, including the cystic fibrosis transmembrane conductance regulator (CFTR), the epithelial sodium channel (ENaC), thyroglobulin, connexin 43 hemichannels, and proinsulin. ERp29 is hypothesized to promote ER to cis-Golgi cargo protein transport via COP II machinery through its interactions with the KDEL receptor; this interaction may facilitate the loading of ERp29 clients into COP II vesicles. ERp29 also plays a role in ER stress (ERS) and the unfolded protein response (UPR) and is implicated in oncogenesis. Here, we review the vast array of ERp29’s clients, its role as an ER to Golgi escort protein, and further suggest ERp29 as a potential target for therapies related to diseases of protein misfolding and mistrafficking.
Collapse
Affiliation(s)
- Margaret Brecker
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Svetlana Khakhina
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Tyler J. Schubert
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Zachary Thompson
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ronald C. Rubenstein
- Cystic Fibrosis Center, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
- *Correspondence: Ronald C. Rubenstein, ;
| |
Collapse
|
6
|
Beach R, Abitbol JM, Allman BL, Esseltine JL, Shao Q, Laird DW. GJB2 Mutations Linked to Hearing Loss Exhibit Differential Trafficking and Functional Defects as Revealed in Cochlear-Relevant Cells. Front Cell Dev Biol 2020; 8:215. [PMID: 32300592 PMCID: PMC7142214 DOI: 10.3389/fcell.2020.00215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/12/2020] [Indexed: 11/13/2022] Open
Abstract
GJB2 gene (that encodes Cx26) mutations are causal of hearing loss highlighting the importance of Cx26-based channel signaling amongst the supporting cells in the organ of Corti. While the majority of these GJB2 mutations are inherited in an autosomal recessive manner, others are inherited in an autosomal dominant manner and lead to syndromic hearing loss as well as skin diseases. To assess if common or divergent mechanisms are at the root of GJB2-linked hearing loss, we expressed several mutants in cochlear-relevant HEI-OC1 cells derived from the developing organ of Corti. Since supporting cells of the mature mammalian organ of Corti have negligible Cx43, but HEI-OC1 cells are rich in Cx43, we first used CRISPR-Cas9 to ablate endogenous Cx43, thus establishing a connexin-deficient platform for controlled reintroduction of hearing-relevant connexins and Cx26 mutants. We found three distinct outcomes and cellular phenotypes when hearing loss-linked Cx26 mutants were expressed in cochlear-relevant cells. The dominant syndromic Cx26 mutant N54K had trafficking defects and did not fully prevent wild-type Cx26 gap junction plaque formation but surprisingly formed gap junctions when co-expressed with Cx30. In contrast, the dominant syndromic S183F mutant formed gap junctions incapable of transferring dye and, as expected, co-localized in the same gap junctions as wild-type Cx26 and Cx30, but also gained the capacity to intermix with Cx43 within gap junctions. Both recessive non-syndromic Cx26 mutants (R32H and R184P) were retained in intracellular vesicles including early endosomes and did not co-localize with Cx30. As might be predicted, none of the Cx26 mutants prevented Cx43 gap junction plaque formation in Cx43-rich HEI-OC1 cells while Cx43-ablation had little effect on the expression of reference genes linked to auditory cell differentiation. We conclude from our studies in cochlear-relevant cells that the selected Cx26 mutants likely evoke hearing loss via three unique connexin defects that are independent of Cx43 status.
Collapse
Affiliation(s)
- Rianne Beach
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Julia M. Abitbol
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Brian L. Allman
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Jessica L. Esseltine
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Qing Shao
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Dale W. Laird
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
7
|
Cohen-Barak E, Godsel LM, Koetsier JL, Hegazy M, Kushnir-Grinbaum D, Hammad H, Danial-Farran N, Harmon R, Khayat M, Bochner R, Peled A, Rozenblat M, Krausz J, Sarig O, Johnson JL, Ziv M, Shalev SA, Sprecher E, Green KJ. The Role of Desmoglein 1 in Gap Junction Turnover Revealed through the Study of SAM Syndrome. J Invest Dermatol 2019; 140:556-567.e9. [PMID: 31465738 DOI: 10.1016/j.jid.2019.08.433] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/15/2019] [Accepted: 08/02/2019] [Indexed: 11/18/2022]
Abstract
An effective epidermal barrier requires structural and functional integration of adherens junctions, tight junctions, gap junctions (GJ), and desmosomes. Desmosomes govern epidermal integrity while GJs facilitate small molecule transfer across cell membranes. Some patients with severe dermatitis, multiple allergies, and metabolic wasting (SAM) syndrome, caused by biallelic desmoglein 1 (DSG1) mutations, exhibit skin lesions reminiscent of erythrokeratodermia variabilis, caused by mutations in connexin (Cx) genes. We, therefore, examined whether SAM syndrome-causing DSG1 mutations interfere with Cx expression and GJ function. Lesional skin biopsies from SAM syndrome patients (n = 7) revealed decreased Dsg1 and Cx43 plasma membrane localization compared with control and nonlesional skin. Cultured keratinocytes and organotypic skin equivalents depleted of Dsg1 exhibited reduced Cx43 expression, rescued upon re-introduction of wild-type Dsg1, but not Dsg1 constructs modeling SAM syndrome-causing mutations. Ectopic Dsg1 expression increased cell-cell dye transfer, which Cx43 silencing inhibited, suggesting that Dsg1 promotes GJ function through Cx43. As GJA1 gene expression was not decreased upon Dsg1 loss, we hypothesized that Cx43 reduction was due to enhanced protein degradation. Supporting this, PKC-dependent Cx43 S368 phosphorylation, which signals Cx43 turnover, increased after Dsg1 depletion, while lysosomal inhibition restored Cx43 levels. These data reveal a role for Dsg1 in regulating epidermal Cx43 turnover.
Collapse
Affiliation(s)
- Eran Cohen-Barak
- Departments of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Dermatology, "Emek" Medical Center, Afula, Israel; Bruce and Ruth Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Lisa M Godsel
- Departments of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jennifer L Koetsier
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Marihan Hegazy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Helwe Hammad
- Department of Dermatology, "Emek" Medical Center, Afula, Israel
| | | | - Robert Harmon
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Morad Khayat
- The Genetic Institute, "Emek" Medical Center, Afula, Israel
| | - Ron Bochner
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Alon Peled
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel; Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mati Rozenblat
- Department of Dermatology, "Emek" Medical Center, Afula, Israel
| | - Judit Krausz
- Department of Pathology, "Emek" Medical Center, Afula, Israel
| | - Ofer Sarig
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Jodi L Johnson
- Departments of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michael Ziv
- Department of Dermatology, "Emek" Medical Center, Afula, Israel
| | - Stavit A Shalev
- Bruce and Ruth Rappaport Faculty of Medicine, Technion, Haifa, Israel; The Genetic Institute, "Emek" Medical Center, Afula, Israel
| | - Eli Sprecher
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel; Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Kathleen J Green
- Departments of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
8
|
Abstract
The connexin family of channel-forming proteins is present in every tissue type in the human anatomy. Connexins are best known for forming clustered intercellular channels, structurally known as gap junctions, where they serve to exchange members of the metabolome between adjacent cells. In their single-membrane hemichannel form, connexins can act as conduits for the passage of small molecules in autocrine and paracrine signalling. Here, we review the roles of connexins in health and disease, focusing on the potential of connexins as therapeutic targets in acquired and inherited diseases as well as wound repair, while highlighting the associated clinical challenges.
Collapse
|
9
|
Zhu S, Ehnert S, Rouß M, Häussling V, Aspera-Werz RH, Chen T, Nussler AK. From the Clinical Problem to the Basic Research-Co-Culture Models of Osteoblasts and Osteoclasts. Int J Mol Sci 2018; 19:ijms19082284. [PMID: 30081523 PMCID: PMC6121694 DOI: 10.3390/ijms19082284] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Bone tissue undergoes constant remodeling and healing when fracture happens, in order to ensure its structural integrity. In order to better understand open biological and clinical questions linked to various bone diseases, bone cell co-culture technology is believed to shed some light into the dark. Osteoblasts/osteocytes and osteoclasts dominate the metabolism of bone by a multitude of connections. Therefore, it is widely accepted that a constant improvement of co-culture models with both cell types cultured on a 3D scaffold, is aimed to mimic an in vivo environment as closely as possible. Although in recent years a considerable knowledge of bone co-culture models has been accumulated, there are still many open questions. We here try to summarize the actual knowledge and address open questions.
Collapse
Affiliation(s)
- Sheng Zhu
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Marc Rouß
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Victor Häussling
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Romina H Aspera-Werz
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Tao Chen
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Andreas K Nussler
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| |
Collapse
|
10
|
DeMille D, Carlston CM, Tam OH, Palumbos JC, Stalker HJ, Mao R, Zori RT, Viskochil DH, Park AH, Carey JC. Three novel
GJB2
(connexin 26) variants associated with autosomal dominant syndromic and nonsyndromic hearing loss. Am J Med Genet A 2018; 176:945-950. [DOI: 10.1002/ajmg.a.38648] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 01/27/2018] [Accepted: 01/29/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Desiree DeMille
- ARUP Institute for Clinical and Experimental PathologySalt Lake City Utah
| | - Colleen M. Carlston
- ARUP Institute for Clinical and Experimental PathologySalt Lake City Utah
- Department of PathologyUniversity of UtahSalt Lake City Utah
| | - Oliver H. Tam
- ARUP Institute for Clinical and Experimental PathologySalt Lake City Utah
| | - Janice C. Palumbos
- Department of Pediatrics, Division of Medical GeneticsUniversity of UtahSalt Lake City Utah
| | - Heather J. Stalker
- Division of Pediatric Genetics and MetabolismUniversity of FloridaGainesville Florida
| | - Rong Mao
- ARUP Institute for Clinical and Experimental PathologySalt Lake City Utah
- Department of PathologyUniversity of UtahSalt Lake City Utah
| | - Roberto T. Zori
- Division of Pediatric Genetics and MetabolismUniversity of FloridaGainesville Florida
| | - David H. Viskochil
- Department of Pediatrics, Division of Medical GeneticsUniversity of UtahSalt Lake City Utah
| | - Albert H. Park
- Division of Otolaryngology – Head and Neck SurgeryUniversity of UtahSalt Lake City Utah
| | - John C. Carey
- Department of Pediatrics, Division of Medical GeneticsUniversity of UtahSalt Lake City Utah
| |
Collapse
|
11
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
12
|
Esseltine JL, Shao Q, Brooks C, Sampson J, Betts DH, Séguin CA, Laird DW. Connexin43 Mutant Patient-Derived Induced Pluripotent Stem Cells Exhibit Altered Differentiation Potential. J Bone Miner Res 2017; 32:1368-1385. [PMID: 28177159 DOI: 10.1002/jbmr.3098] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 01/26/2017] [Accepted: 02/01/2017] [Indexed: 01/06/2023]
Abstract
We present for the first time the generation of induced pluripotent stem cells (iPSCs) from a patient with a connexin-linked disease. The importance of gap junctional intercellular communication in bone homeostasis is exemplified by the autosomal dominant developmental disorder oculodentodigital dysplasia (ODDD), which is linked to mutations in the GJA1 (Cx43) gene. ODDD is characterized by craniofacial malformations, ophthalmic deficits, enamel hypoplasia, and syndactyly. In addition to harboring a Cx43 p.V216L mutation, ODDD iPSCs exhibit reduced Cx43 mRNA and protein abundance when compared to control iPSCs and display impaired channel function. Osteogenic differentiation involved an early, and dramatic downregulation of Cx43 followed by a slight upregulation during the final stages of differentiation. Interestingly, osteoblast differentiation was delayed in ODDD iPSCs. Moreover, Cx43 subcellular localization was altered during chondrogenic differentiation of ODDD iPSCs compared to controls and this may have contributed to the more compact cartilage pellet morphology found in differentiated ODDD iPSCs. These studies highlight the importance of Cx43 expression and function during osteoblast and chondrocyte differentiation, and establish a potential mechanism for how ODDD-associated Cx43 mutations may have altered cell lineages involved in bone and cartilage development. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jessica L Esseltine
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario. London, ON, Canada
| | - Qing Shao
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario. London, ON, Canada
| | - Courtney Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Jacinda Sampson
- Department of Neurology, Stanford University Medical Center, Palo Alto, CA, USA
| | - Dean H Betts
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Cheryle A Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Dale W Laird
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario. London, ON, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
13
|
Connexin30.3 is expressed in mouse embryonic stem cells and is responsive to leukemia inhibitory factor. Sci Rep 2017; 7:42403. [PMID: 28205646 PMCID: PMC5304323 DOI: 10.1038/srep42403] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/09/2017] [Indexed: 01/08/2023] Open
Abstract
The expression of 19 connexin (Cx) isoforms was observed in the mouse embryonic stem (ES) cell line, EB3. Their expression patterns could be classified into either pluripotent state-specific, differentiating stage-specific, or non-specific Cxs. We focused on Cx30.3 as typical of the first category. Cx30.3 was pluripotent state-specific and upregulated by leukemia inhibitory factor (LIF), a specific cytokine that maintains the pluripotent state of ES cell, via a Jak signaling pathway. Cx30.3 protein was localized to both the cell membrane and cytosol. The dynamic movement of Cx30.3 in the cell membrane was suggested by the imaging analysis by means of overexpressed Cx30.3-EGFP fusion protein. The cytosolic portion was postulated to be a ready-to-use Cx pool. The Cx30.3 expression level in ES cell colonies dramatically decreased immediately after their separation into single cells. It was suggested that mRNA for Cx30.3 and Cx30.3 protein might be decomposed more rapidly than mRNA for Cx43 and Cx43 protein, respectively. These indicate possible involvement of Cx30.3 in the rapid formation and/or decomposition of gap junctions; implying a functional relay between Cx30.3 and other systems such as adhesion proteins.
Collapse
|
14
|
Merrifield PA, Laird DW. Connexins in skeletal muscle development and disease. Semin Cell Dev Biol 2016; 50:67-73. [DOI: 10.1016/j.semcdb.2015.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 11/30/2022]
|
15
|
Manipulating Cx43 expression triggers gene reprogramming events in dermal fibroblasts from oculodentodigital dysplasia patients. Biochem J 2015; 472:55-69. [PMID: 26349540 DOI: 10.1042/bj20150652] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/08/2015] [Indexed: 02/06/2023]
Abstract
Oculodentodigital dysplasia (ODDD) is primarily an autosomal dominant disorder linked to over 70 GJA1 gene [connexin43 (Cx43)] mutations. For nearly a decade, our laboratory has been investigating the relationship between Cx43 and ODDD by expressing disease-linked mutants in reference cells, tissue-relevant cell lines, 3D organ cultures and by using genetically modified mouse models of human disease. Although salient features of Cx43 mutants have been revealed, these models do not necessarily reflect the complexity of the human context. To further overcome these limitations, we have acquired dermal fibroblasts from two ODDD-affected individuals harbouring D3N and V216L mutations in Cx43, along with familial controls. Using these ODDD patient dermal fibroblasts, which naturally produce less GJA1 gene product, along with RNAi and RNA activation (RNAa) approaches, we show that manipulating Cx43 expression triggers cellular gene reprogramming. Quantitative RT-PCR, Western blot and immunofluorescent analysis of ODDD patient fibroblasts show unusually high levels of extracellular matrix (ECM)-interacting proteins, including integrin α5β1, matrix metalloproteinases as well as secreted ECM proteins collagen-I and laminin. Cx43 knockdown in familial control cells produces similar effects on ECM expression, whereas Cx43 transcriptional up-regulation using RNAa decreases production of collagen-I. Interestingly, the enhanced levels of ECM-associated proteins in ODDD V216L fibroblasts is not only a consequence of increased ECM gene expression, but also due to an apparent deficit in collagen-I secretion which may further contribute to impaired collagen gel contraction in ODDD fibroblasts. These findings further illuminate the altered function of Cx43 in ODDD-affected individuals and highlight the impact of manipulating Cx43 expression in human cells.
Collapse
|
16
|
M Kidder G, Winterhager E. Physiological roles of connexins in labour and lactation. Reproduction 2015; 150:R129-36. [PMID: 26150552 DOI: 10.1530/rep-15-0134] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/06/2015] [Indexed: 01/08/2023]
Abstract
The connexin family of proteins are best known as oligomerizing to form intercellular membrane channels (gap junctions) that metabolically and ionically couple cells to allow for coordinated cellular function. Nowhere in the body is this role better illustrated than in the uterine smooth muscle during parturition, where gap junctions conduct the contraction wave throughout the tissue to deliver the baby. Parturition is followed by the onset of lactation with connexins contributing to both the dramatic reorganization of mammary gland tissue leading up to lactation and the smooth muscle contraction of the myoepithelial cells which extrudes the milk. This review summarizes what is known about the expression and roles of individual connexin family members in the uterus during labour and in the mammary glands during development and lactation. Connexin loss or malfunction in mammary glands and the uterus can have serious implications for the health of both the mother and the newborn baby.
Collapse
Affiliation(s)
- Gerald M Kidder
- Department of Physiology and PharmacologySchulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, CanadaInstitute of Molecular BiologyUniversity of Duisburg-Essen, University Clinics, 45211 Essen, Germany
| | - Elke Winterhager
- Department of Physiology and PharmacologySchulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, CanadaInstitute of Molecular BiologyUniversity of Duisburg-Essen, University Clinics, 45211 Essen, Germany
| |
Collapse
|
17
|
Zappitelli T, Aubin JE. The "connexin" between bone cells and skeletal functions. J Cell Biochem 2015; 115:1646-58. [PMID: 24818806 DOI: 10.1002/jcb.24836] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 05/08/2014] [Indexed: 11/11/2022]
Abstract
The processes of bone modeling and remodeling are crucial in the skeleton's functions as a supportive and protective structure, a mineral reservoir, and an endocrine organ. The coordination between bone cell activities (bone formation and bone resorption), necessary to maintain the integrity of the skeleton during these processes, is mediated at least in part by cell-cell and cell-environment interactions across gap junctions and hemichannels. The increasing number of genetically engineered Connexin 43 (Cx43) knockout and missense mouse models have provided insight into the complex and critical roles of Cx43-containing gap junctions and hemichannels in the development and turnover of the skeleton, in differentiation, activity and survival of the bone cell lineages, and in the cellular and molecular mechanisms by which Cx43 functions and assists in mediating cellular responses to stimuli in bone. Cx43 may be an important potential therapeutic target, making it crucial that we continue to gain understanding of the multiple and complex roles of Cx43 in bone.
Collapse
Affiliation(s)
- Tanya Zappitelli
- Department of Medical Biophysics, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
| | | |
Collapse
|
18
|
CIP75 (connexin43-interacting protein of 75 kDa) mediates the endoplasmic reticulum dislocation of connexin43. Biochem J 2014; 458:57-67. [DOI: 10.1042/bj20131247] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The proteasomal degradation of the gap junction protein connexin43 is facilitated by its interaction with the connexin43-interacting protein of 75 kDa at the endoplasmic reticulum membrane.
Collapse
|
19
|
Connexins: mechanisms regulating protein levels and intercellular communication. FEBS Lett 2014; 588:1212-20. [PMID: 24457202 DOI: 10.1016/j.febslet.2014.01.013] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/10/2014] [Accepted: 01/13/2014] [Indexed: 11/21/2022]
Abstract
Intercellular communication can occur through gap junction channels, which are comprised of connexin proteins. Therefore, levels of connexins can directly correlate with gap junctional intercellular communication. Because gap junctions have a critical role in maintaining cellular homeostasis, the regulation of connexin protein levels is important. In the connexin life cycle, connexin protein levels can be modified through differential gene transcription or altered through trafficking and degradation mechanisms. More recently, significant attention has been directed to the pathways that cells utilize to increase or decrease connexin levels and thus indirectly, gap junctional communication. Here, we review the studies revealing the mechanisms that affect connexin protein levels and gap junctional intercellular communication.
Collapse
|
20
|
Zou J, Salarian M, Chen Y, Veenstra R, Louis CF, Yang JJ. Gap junction regulation by calmodulin. FEBS Lett 2014; 588:1430-8. [PMID: 24440348 DOI: 10.1016/j.febslet.2014.01.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 01/09/2014] [Accepted: 01/10/2014] [Indexed: 10/25/2022]
Abstract
Intracellular Ca(2+) activated calmodulin (CaM) inhibits gap junction channels in the low nanomolar to high micromolar range of [Ca(2+)]i. This regulation plays an essential role in numerous cellular processes that include hearing, lens transparency, and synchronized contractions of the heart. Previous studies have indicated that gap junction mediated cell-to-cell communication was inhibited by CaM antagonists. More recent evidence indicates a direct role of CaM in regulating several members of the connexin family. Since the intracellular loop and carboxyl termini of connexins are largely "invisible" in electron microscopy and X-ray crystallographic structures due to disorder in these domains, peptide models encompassing the putative CaM binding sites of several intracellular domains of connexins have been used to identify the Ca(2+)-dependent CaM binding sites of these proteins. This approach has been used to determine the CaM binding affinities of peptides derived from a number of different connexin-subfamilies.
Collapse
Affiliation(s)
- Juan Zou
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, United States
| | - Mani Salarian
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, United States
| | - Yanyi Chen
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, United States
| | - Richard Veenstra
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Charles F Louis
- Department of Cell Biology and Neuroscience, University of California, Riverside, CA 92521, United States.
| | - Jenny J Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, United States.
| |
Collapse
|
21
|
Patel D, Zhang X, Veenstra RD. Connexin hemichannel and pannexin channel electrophysiology: how do they differ? FEBS Lett 2014; 588:1372-8. [PMID: 24434538 DOI: 10.1016/j.febslet.2013.12.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 12/20/2013] [Accepted: 12/30/2013] [Indexed: 12/21/2022]
Abstract
Connexin hemichannels are postulated to form a cell permeabilization pore for the uptake of fluorescent dyes and release of cellular ATP. Connexin hemichannel activity is enhanced by low external [Ca(2+)]o, membrane depolarization, metabolic inhibition, and some disease-causing gain-of-function connexin mutations. This paper briefly reviews the electrophysiological channel conductance, permeability, and pharmacology properties of connexin hemichannels, pannexin 1 channels, and purinergic P2X7 receptor channels as studied in exogenous expression systems including Xenopus oocytes and mammalian cell lines such as HEK293 cells. Overlapping pharmacological inhibitory and channel conductance and permeability profiles makes distinguishing between these channel types sometimes difficult. Selective pharmacology for Cx43 hemichannels (Gap19 peptide), probenecid or FD&C Blue #1 (Brilliant Blue FCF, BB FCF) for Panx1, and A740003, A438079, or oxidized ATP (oATP) for P2X7 channels may be the best way to distinguish between these three cell permeabilizing channel types. Endogenous connexin, pannexin, and P2X7 expression should be considered when performing exogenous cellular expression channel studies. Cell pair electrophysiological assays permit the relative assessment of the connexin hemichannel/gap junction channel ratio not often considered when performing isolated cell hemichannel studies.
Collapse
Affiliation(s)
- Dakshesh Patel
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Xian Zhang
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Richard D Veenstra
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, United States.
| |
Collapse
|
22
|
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: 9.6] [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
|
23
|
Sedaghat AR, Cunningham MJ, Busaba NY. Connexin 32 and 43 mutations: do they play a role in chronic rhinosinusitis? Am J Otolaryngol 2014; 35:33-6. [PMID: 24119489 DOI: 10.1016/j.amjoto.2013.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/06/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Dysfunction of the sinonasal epithelium may contribute to the pathogenesis of chronic rhinosinusitis (CRS) including recurrent acute rhinosinusitis (RARS). Mutations in connexin 32 and 43 proteins have been associated with a number of human diseases. The objective of this study is to investigate the role of mutations in connexin 32 or connexin 43 genes in CRS and RARS. METHODS Prospective case series of 19 patients with CRS and /or RARS. Clinical and demographic factors were noted and buccal swabs were collected for DNA sequencing of connexin 32 and connexin 43 genes. RESULTS One patient was found to have a conservative V193I mutation in the connexin 32 gene. Connexin 43 mutations were found in two patients - a silent R239R mutation and an AAA insertion after the stop codon in the 3' UTR. None of these mutations are associated with any known diseases or predicted to lead to protein dysfunction. CONCLUSION Mutations in connexin 32 or 43 genes in patients with CRS, including RARS, appear to be rare. The etiologic role of connexin mutations in chromic rhinosinusitis is suspect, and routine sequencing for connexin mutations in patients with RARS or CRS is not cost effective.
Collapse
Affiliation(s)
- Ahmad R Sedaghat
- Department of Otolaryngology - Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, MA, USA; Department of Otology and Laryngology, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
24
|
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: 51] [Impact Index Per Article: 4.3] [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
|
25
|
Grosely R, Kopanic JL, Nabors S, Kieken F, Spagnol G, Al-Mugotir M, Zach S, Sorgen PL. Effects of phosphorylation on the structure and backbone dynamics of the intrinsically disordered connexin43 C-terminal domain. J Biol Chem 2013; 288:24857-70. [PMID: 23828237 DOI: 10.1074/jbc.m113.454389] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphorylation of the connexin43 C-terminal (Cx43CT) domain regulates gap junction intercellular communication. However, an understanding of the mechanisms by which phosphorylation exerts its effects is lacking. Here, we test the hypothesis that phosphorylation regulates Cx43 gap junction intercellular communication by mediating structural changes in the C-terminal domain. Circular dichroism and nuclear magnetic resonance were used to characterize the effects of phosphorylation on the secondary structure and backbone dynamics of soluble and membrane-tethered Cx43CT domains. Cx43CT phospho-mimetic isoforms, which have Asp substitutions at specific Ser/Tyr sites, revealed phosphorylation alters the α-helical content of the Cx43CT domain only when attached to the membrane. The changes in secondary structure are due to variations in the conformational preference and backbone flexibility of residues adjacent and distal to the site(s) of modification. In addition to the known direct effects of phosphorylation on molecular partner interactions, the data presented here suggest phosphorylation may also indirectly regulate binding affinity by altering the conformational preference of the Cx43CT domain.
Collapse
Affiliation(s)
- Rosslyn Grosely
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Kim AH, Nahm E, Sollas A, Mattiace L, Rozental R. Connexin 43 and hearing: possible implications for retrocochlear auditory processing. Laryngoscope 2013; 123:3185-93. [PMID: 23817980 DOI: 10.1002/lary.24249] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 05/10/2013] [Accepted: 05/22/2013] [Indexed: 11/09/2022]
Abstract
OBJECTIVES/HYPOTHESIS To examine the relationship between hearing and connexin 43, a dominant gap junctional protein in the central nervous system. STUDY DESIGN Original research. METHODS Connexin 43 heterozygous mice are used to assess its mutational effect on hearing. Results are compared to controls consisting of connexin 43, wild type and CBA/J mice. Hearing is assessed using auditory brainstem response and distortion product otoacoustic emissions tests. Distribution of connexin 43 in the organ of Corti and the retrocochlear auditory centers (eight nerve, cochlear nucleus, olivary complex, lateral lemniscus, inferior colliculus, respectively) is examined. Fluorescent markers are used to elucidate cell types. RESULTS Mean click auditory brainstem response threshold for the young connexin 43 heterozygous mice (3-4 months) was 36.7 ± 12.6 dB compared to 25 ± 0 dB for control mice (P < 0.05). Mean threshold difference became more pronounced (68 ± 7.5 dB vs. 31 ± 2.2 dB) at 10 months (P < 0.05). Tonal auditory brainstem response testing showed elevated thresholds (>60 dB) at all frequencies (4-32 kHz) compared to the controls. Distortion product otoacoustic emissions (DPOAE) were present in all the mice, although the older connexin 43 heterozygous mice responded at higher thresholds. The pattern of connexin 43 immunoreactivity was distinctive from connexin 26 and 30, showing minimal presence in the organ of Corti but robustly present in the retrocochlear centers. CONCLUSION Connexin 43 heterozygous mice demonstrated greater degree of hearing loss compared to age-matched controls. It is abundantly found in the retrocochlear auditory centers. The mechanism of hearing loss in these mice does not appear to be related to hair cell loss.
Collapse
Affiliation(s)
- Ana H Kim
- Department of Otolaryngology, New York Eye and Ear Infirmary, New York; New York Medical College, Valhalla, New York
| | | | | | | | | |
Collapse
|
27
|
Ma W, Zhang X, Shi S, Zhang Y. Neuropeptides stimulate human osteoblast activity and promote gap junctional intercellular communication. Neuropeptides 2013; 47:179-86. [PMID: 23726661 DOI: 10.1016/j.npep.2012.12.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 11/22/2012] [Accepted: 12/20/2012] [Indexed: 10/27/2022]
Abstract
Neuropeptides released from the skeletal nerve fibers have neurotransmitter and immunoregulatory roles; they exert paracrine biological effects on bone cells present close to the nerve endings expressing these signaling molecules. The aims of this study were a systematic investigation of the effects of the neuropeptides substance P (SP), calcitonin gene-related peptide (CGRP), vasoactive intestinal polypeptide (VIP), Neuropeptide Y (NPY) and tyrosine hydroxylase (TH) on the cell viability and function of the human osteoblasts, and comparing their difference in the role of regulating bone formation. Cultures of normal human osteoblasts were treated with SP, CGRP, VIP, NPY or TH at three concentrations. We found that each of the five neuropeptides induced increases in cell viability of human osteoblasts. The stimulatory action of NPY was the highest, followed by VIP, SP and TH, while CGRP had the lowest stimulatory effect. The viability index of osteoblasts was inversely associated with the concentration of neuropeptides, and positively with the time of exposure. Moreover, the five neuropeptides increased the ALP activity and osteocalcin to different extents in a dose-dependent manner. The GJIC of osteoblasts was significantly promoted by neuropeptides. The results demonstrated that neuropeptides released from skeletal nerve endings after a stimulus appeared to be able to induce the proliferation and activity of osteoblasts via enhancing GJIC between cells, and further influence the bone formation. These findings may contribute toward a better understanding of the neural influence on bone remodeling and improving treatments related to bone diseases.
Collapse
Affiliation(s)
- Wenhui Ma
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051 Hebei Province, China.
| | | | | | | |
Collapse
|
28
|
Avshalumova L, Fabrikant J, Koriakos A. Overview of skin diseases linked to connexin gene mutations. Int J Dermatol 2013; 53:192-205. [DOI: 10.1111/ijd.12062] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | - Jordan Fabrikant
- Texas Division; Department of Dermatology; Larkin Community Hospital; Miami FL USA
| | - Angie Koriakos
- Department of Dermatology; University of North Texas Health Science Center/Texas College of Osteopathic Medicine; Houston TX USA
| |
Collapse
|
29
|
Huang T, Shao Q, MacDonald A, Xin L, Lorentz R, Bai D, Laird DW. Autosomal recessive GJA1 (Cx43) gene mutations cause oculodentodigital dysplasia by distinct mechanisms. J Cell Sci 2013; 126:2857-66. [PMID: 23606748 DOI: 10.1242/jcs.123315] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oculodentodigital dysplasia (ODDD) is mainly an autosomal dominant human disease caused by mutations in the GJA1 gene, which encodes the gap junction protein connexin43 (Cx43). Surprisingly, there have been two autosomal recessive mutations reported that cause ODDD: a single amino acid substitution (R76H) and a premature truncation mutation (R33X). When expressed in either gap junctional intercellular communication (GJIC)-deficient HeLa cells or Cx43-expressing NRK cells, the R76H mutant trafficked to the plasma membrane to form gap junction-like plaques, whereas the R33X mutant remained diffusely localized throughout the cell, including the nucleus. As expected, the R33X mutant failed to form functional channels. In the case of the R76H mutant, dye transfer studies in HeLa cells and electrical conductance analysis in GJIC-deficient N2a cells revealed that this mutant could form functional gap junction channels, albeit with reduced macroscopic and single channel conductance. Alexa 350 dye transfer studies further revealed that the R76H mutant had no detectable negative effect on the function of co-expressed Cx26, Cx32, Cx37 or Cx40, whereas the R33X mutant exhibited significant dominant or trans-dominant effects on Cx43 and Cx40 as manifested by a reduction in wild-type connexin gap junction plaques. Taken together, our results suggest that the trans-dominant effect of R33X together with its complete inability to form a functional channel may explain why patients harboring this autosomal recessive R33X mutant exhibit greater disease burden than patients harboring the R76H mutant.
Collapse
Affiliation(s)
- Tao Huang
- Department of Anatomy and Cell Biology, University of Western Ontario, London ON N6A-5C1, Canada
| | | | | | | | | | | | | |
Collapse
|
30
|
Meda P. Protein-mediated interactions of pancreatic islet cells. SCIENTIFICA 2013; 2013:621249. [PMID: 24278783 PMCID: PMC3820362 DOI: 10.1155/2013/621249] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 12/10/2012] [Indexed: 05/29/2023]
Abstract
The islets of Langerhans collectively form the endocrine pancreas, the organ that is soley responsible for insulin secretion in mammals, and which plays a prominent role in the control of circulating glucose and metabolism. Normal function of these islets implies the coordination of different types of endocrine cells, noticeably of the beta cells which produce insulin. Given that an appropriate secretion of this hormone is vital to the organism, a number of mechanisms have been selected during evolution, which now converge to coordinate beta cell functions. Among these, several mechanisms depend on different families of integral membrane proteins, which ensure direct (cadherins, N-CAM, occludin, and claudins) and paracrine communications (pannexins) between beta cells, and between these cells and the other islet cell types. Also, other proteins (integrins) provide communication of the different islet cell types with the materials that form the islet basal laminae and extracellular matrix. Here, we review what is known about these proteins and their signaling in pancreatic β -cells, with particular emphasis on the signaling provided by Cx36, given that this is the integral membrane protein involved in cell-to-cell communication, which has so far been mostly investigated for effects on beta cell functions.
Collapse
Affiliation(s)
- Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland
| |
Collapse
|
31
|
Paraguassú-Braga FH, Alves APG, Andrade Santos IMA, Bonamino M, Bonomo A. An Ectopic Stromal Implant Model for Hematopoietic Reconstitution and in Vivo Evaluation of Bone Marrow Niches. Cell Transplant 2012; 21:2677-88. [DOI: 10.3727/096368912x636993] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In adults, hematopoiesis takes places in the bone marrow, where specialized niches containing mesenchymal nonhematopoietic cells (stroma) harbor the hematopoietic stem cell (HSC). These niches are responsible and essential for the maintenance of HSCs. Attempts to expand HSCs fail to keep the general properties of stem cells, which depend on several niche components difficult to reproduce in in vitro culture systems. Here, we describe a methodology for in vivo study of hematopoietic stroma. We use stroma-loaded macroporous microcarriers implanted in the subcutaneous tissue of experimental animals and show that the ectopic stroma implant (ESI) is able to support hematopoiesis. Moreover, lethally irradiated mice can be rescued by ESI preloaded with HSCs, showing that they function as an ectopic bone marrow. ESI is also shown as a good system to study the role of different niche components. As an example, we used stromas lacking connexin 43 (Cx43) and confirm the importance of this molecule in the maintenance of the HSC niche in vivo. We believe ESI can work as an ectopic bone marrow allowing in vivo testing of different niches components and opening new avenues for the treatment of a variety of hematologic conditions particularly when stromal cell defects are the main cause of disease.
Collapse
Affiliation(s)
- Flávio Henrique Paraguassú-Braga
- Banco de Sangue de Cordão Umbilical e Placentário, Centro de Transplante de Medula Óssea, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Ana Paula G. Alves
- Programa de Medicina Experimental, Coordenação Geral Técnico-Científica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | - Martin Bonamino
- Programa de Medicina Experimental, Coordenação Geral Técnico-Científica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Adriana Bonomo
- Programa de Medicina Experimental, Coordenação Geral Técnico-Científica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Shao Q, Liu Q, Lorentz R, Gong XQ, Bai D, Shaw GS, Laird DW. Structure and functional studies of N-terminal Cx43 mutants linked to oculodentodigital dysplasia. Mol Biol Cell 2012; 23:3312-21. [PMID: 22809623 PMCID: PMC3431933 DOI: 10.1091/mbc.e12-02-0128] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mutations in the connexin-43 gap junction protein cause the developmental disease known as oculodentodigital dysplasia. Structure and function approaches are used to demonstrate that the nature of the missense mutation in the amino-terminal domain of connexin-43 governs the mechanism that leads to loss of connexin-43 function. Mutations in the gene encoding connexin-43 (Cx43) cause the human development disorder known as oculodentodigital dysplasia (ODDD). In this study, ODDD-linked Cx43 N-terminal mutants formed nonfunctional gap junction–like plaques and exhibited dominant-negative effects on the coupling conductance of coexpressed endogenous Cx43 in reference cell models. Nuclear magnetic resonance (NMR) protein structure determination of an N-terminal 23–amino acid polypeptide of wild-type Cx43 revealed that it folded in to a kinked α-helical structure. This finding predicted that W4 might be critically important in intramolecular and intermolecular interactions. Thus we engineered and characterized a W4A mutant and found that this mutant formed a regular, nonkinked α-helix but did not form functional gap junctions. Furthermore, a G2V variant peptide of Cx43 showed a kinked helix that now included V2 interactions with W4, resulting in the G2V mutant forming nonfunctional gap junctions. Also predicted from the NMR structures, a G2S mutant was found to relieve these interactions and allowed the protein to form functional gap junctions. Collectively, these studies suggest that the nature of the mutation conveys loss of Cx43 function by distinctly different mechanisms that are rooted in the structure of the N-terminal region.
Collapse
Affiliation(s)
- Qing Shao
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | | | | | | | | | | | | |
Collapse
|
33
|
Degradation of connexins through the proteasomal, endolysosomal and phagolysosomal pathways. J Membr Biol 2012; 245:389-400. [PMID: 22772442 DOI: 10.1007/s00232-012-9461-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 06/20/2012] [Indexed: 01/23/2023]
Abstract
Connexins comprise gap junction channels, which create a direct conduit between the cytoplasms of adjacent cells and provide for intercellular communication. Therefore, the level of total cellular connexin protein can have a direct influence on the level of intercellular communication. Control of connexin protein levels can occur through different mechanisms during the connexin life cycle, such as by regulation of connexin gene expression and turnover of existing protein. The degradation of connexins has been extensively studied, revealing proteasomal, endolysosomal and more recently autophagosomal degradation mechanisms that modulate connexin turnover and, subsequently, affect intercellular communication. Here, we review the current knowledge of connexin degradation pathways.
Collapse
|
34
|
Shen DW, Pouliot LM, Gillet JP, Ma W, Johnson AC, Hall MD, Gottesman MM. The transcription factor GCF2 is an upstream repressor of the small GTPAse RhoA, regulating membrane protein trafficking, sensitivity to doxorubicin, and resistance to cisplatin. Mol Pharm 2012; 9:1822-33. [PMID: 22571463 PMCID: PMC3367311 DOI: 10.1021/mp300153z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Our aim was to explore the involvement of the transcriptional suppressor GCF2 in silencing RhoA, disorganization of the cytoskeleton, mislocalization of MRP1, and sensitivity to anticancer agents as an upstream gene target in cancer therapy. Increased expression of GCF2 was found in human cisplatin-resistant cells, and overexpression in GCF2-transfected cells results in loss of RhoA expression and disruption of the actin/filamin network. In consequence, the membrane transporter MRP1 was internalized from the cell surface into the cytoplasm, rendering cells sensitive to doxorubicin by more than 10-fold due to increased accumulation of doxorubicin in the cells. The GCF2 transfectants also showed reduced accumulation of cisplatin and increased resistance. siRNA targeted to GCF2 suppressed the expression of GCF2 in cisplatin-resistant cells, reactivated RhoA expression, and restored the fine structure of actin microfilaments. MRP1 was also relocated to the cell surface. siRNA targeted to RhoA increased resistance 3-fold in KB-3-1 and KB-CP.5 cells. These data for the first time demonstrate a novel complex regulatory pathway downstream from GCF2 involving the small GTPase RhoA, actin/filamin dynamics, and membrane protein trafficking. This pathway mediates diverse responses to cytotoxic compounds, and also provides a molecular basis for further investigation into the pleiotropic resistance mechanism at play in cisplatin-resistant cells.
Collapse
Affiliation(s)
- Ding-Wu Shen
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Lynn M. Pouliot
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jean-Pierre Gillet
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Wenting Ma
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Alfred C. Johnson
- Office of the Director, National Institutes of Health, Bethesda, MD 20892
| | - Matthew D. Hall
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michael M. Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
35
|
Finegold DN, Baty CJ, Knickelbein KZ, Perschke S, Noon SE, Campbell D, Karlsson JM, Huang D, Kimak MA, Lawrence EC, Feingold E, Meriney SD, Brufsky AM, Ferrell RE. Connexin 47 mutations increase risk for secondary lymphedema following breast cancer treatment. Clin Cancer Res 2012; 18:2382-90. [PMID: 22351697 DOI: 10.1158/1078-0432.ccr-11-2303] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Secondary lymphedema is a frequent complication of breast cancer associated with surgery, chemotherapy, or radiation following breast cancer treatment. The potential contribution of genetic susceptibility to risk of developing secondary lymphedema following surgical trauma, radiation, and other tissue insults has not been studied. EXPERIMENTAL DESIGN To determine whether women with breast cancer and secondary lymphedema had mutations in candidate lymphedema genes, we undertook a case-control study of 188 women diagnosed with breast cancer recruited from the University of Pittsburgh Breast Cancer Program (http://www.upmccancercenter.com/breast/index.cfm) between 2000 and 2010. Candidate lymphedema genes, GJC2 (encoding connexin 47 [Cx47]), FOXC2, HGF, MET, and FLT4 (encoding VEGFR3), were sequenced for mutation. Bioinformatics analysis and in vitro functional assays were used to confirm significance of novel mutations. RESULTS Cx47 mutations were identified in individuals having secondary lymphedema following breast cancer treatment but not in breast cancer controls or normal women without breast cancer. These novel mutations are dysfunctional as assessed through in vitro assays and bioinformatics analysis and provide evidence that altered gap junction function leads to lymphedema. CONCLUSIONS Our findings challenge the view that secondary lymphedema is solely due to mechanical trauma and support the hypothesis that genetic susceptibility is an important risk factor for secondary lymphedema. A priori recognition of genetic risk (i) raises the potential for early detection and intervention for a high-risk group and (ii) allows the possibility of altering surgical approach and/or chemo- and radiation therapy, or direct medical treatment of secondary lymphedema with novel connexin-modifying drugs.
Collapse
Affiliation(s)
- David N Finegold
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Furuta N, Ikeda M, Hirayanagi K, Fujita Y, Amanuma M, Okamoto K. A novel GJA1 mutation in oculodentodigital dysplasia with progressive spastic paraplegia and sensory deficits. Intern Med 2012; 51:93-8. [PMID: 22214631 DOI: 10.2169/internalmedicine.51.5770] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oculodentodigital dysplasia (ODDD) is a rare autosomal dominant inherited disorder mainly affecting the development of the face, eyes, dentition, limbs, hair and heart. GJA1 (the gap junction protein α-1) has been determined to be a causative gene of ODDD, mapped to chromosome 6q22-24 identified as the connexin 43 gene (Cx43). We found a novel GJA1 mutation (W25C) as the possible causative gene in this sporadic ODDD patient with neurological features of motor deficits by pyramidal tract signs, and sensory deficits due to peripheral nerve disturbance. It is also notable that the MRI of this patient demonstrated widespread aberrant signal lesions in the brain and brainstem.
Collapse
Affiliation(s)
- Natsumi Furuta
- Department of Neurology, Gunma University Graduate School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Gregory M, Kahiri CN, Barr KJ, Smith CE, Hermo L, Cyr DG, Kidder GM. Male reproductive system defects and subfertility in a mutant mouse model of oculodentodigital dysplasia1. ACTA ACUST UNITED AC 2011; 34:e630-41. [DOI: 10.1111/j.1365-2605.2011.01224.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
38
|
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
|
39
|
Miron RJ, Hedbom E, Ruggiero S, Bosshardt DD, Zhang Y, Mauth C, Gemperli AC, Iizuka T, Buser D, Sculean A. Premature osteoblast clustering by enamel matrix proteins induces osteoblast differentiation through up-regulation of connexin 43 and N-cadherin. PLoS One 2011; 6:e23375. [PMID: 21858092 PMCID: PMC3156132 DOI: 10.1371/journal.pone.0023375] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 07/14/2011] [Indexed: 01/17/2023] Open
Abstract
In recent years, enamel matrix derivative (EMD) has garnered much interest in the dental field for its apparent bioactivity that stimulates regeneration of periodontal tissues including periodontal ligament, cementum and alveolar bone. Despite its widespread use, the underlying cellular mechanisms remain unclear and an understanding of its biological interactions could identify new strategies for tissue engineering. Previous in vitro research has demonstrated that EMD promotes premature osteoblast clustering at early time points. The aim of the present study was to evaluate the influence of cell clustering on vital osteoblast cell-cell communication and adhesion molecules, connexin 43 (cx43) and N-cadherin (N-cad) as assessed by immunofluorescence imaging, real-time PCR and Western blot analysis. In addition, differentiation markers of osteoblasts were quantified using alkaline phosphatase, osteocalcin and von Kossa staining. EMD significantly increased the expression of connexin 43 and N-cadherin at early time points ranging from 2 to 5 days. Protein expression was localized to cell membranes when compared to control groups. Alkaline phosphatase activity was also significantly increased on EMD-coated samples at 3, 5 and 7 days post seeding. Interestingly, higher activity was localized to cell cluster regions. There was a 3 fold increase in osteocalcin and bone sialoprotein mRNA levels for osteoblasts cultured on EMD-coated culture dishes. Moreover, EMD significantly increased extracellular mineral deposition in cell clusters as assessed through von Kossa staining at 5, 7, 10 and 14 days post seeding. We conclude that EMD up-regulates the expression of vital osteoblast cell-cell communication and adhesion molecules, which enhances the differentiation and mineralization activity of osteoblasts. These findings provide further support for the clinical evidence that EMD increases the speed and quality of new bone formation in vivo.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Dere E, Zlomuzica A. The role of gap junctions in the brain in health and disease. Neurosci Biobehav Rev 2011; 36:206-17. [PMID: 21664373 DOI: 10.1016/j.neubiorev.2011.05.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 05/25/2011] [Accepted: 05/27/2011] [Indexed: 11/19/2022]
Abstract
Gap junctions connect the cytosolic compartments of adjacent cells for direct electrotonic and metabolic cell-to-cell communication. Gap junctions between glial cells or neurons are ubiquitously expressed in the brain and play a role in brain development including cell differentiation, cell migration and survival, tissue homeostasis, as well as in human diseases including hearing loss, skin disease, neuropathies, epilepsy, brain trauma, and cardiovascular disease. Furthermore, gap junctions are involved in the synchronization and rhythmic oscillation of hippocampal and neocotical neuronal ensembles which might be important for memory formation and consolidation. In this review the accumulated evidence from mouse mutant and pharmacological studies using gap junction blockers is summarized and the progress made in dissecting the physiological, pathophysiological and behavioral roles of gap junction mediated intercellular communication in the brain is discussed.
Collapse
Affiliation(s)
- Ekrem Dere
- Université Pierre et Marie Curie, Paris 6, UFR des Sciences de la Vie, UMR 7102, Neurobiologie des Processus Adaptatifs, 9 quai St Bernard, 75005 Paris, France.
| | | |
Collapse
|
41
|
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: 206] [Impact Index Per Article: 13.7] [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
|
42
|
Su V, Nakagawa R, Koval M, Lau AF. Ubiquitin-independent proteasomal degradation of endoplasmic reticulum-localized connexin43 mediated by CIP75. J Biol Chem 2010; 285:40979-90. [PMID: 20940304 DOI: 10.1074/jbc.m110.170753] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Connexin43 (Cx43) is a transmembrane protein that forms gap junction channels. Regulation of Cx43 turnover is one mechanism to control the level of intercellular communication that occurs through gap junction channels. Proteasomal degradation of Cx43 is regulated in part through CIP75, a ubiquitin-like and ubiquitin-associated domain containing protein. CIP75 interacts with endoplasmic reticulum-localized Cx43, as demonstrated through co-immunoprecipitation and immunofluorescence microscopy experiments. CIP75 also binds to free monoubiquitin and lysine 48-linked tetraubiquitin chains in vitro and binds to ubiquitinated proteins in cellular lysates. However, analysis of Cx43 that immunoprecipitated with CIP75 demonstrated that the Cx43 associated with CIP75 was not ubiquitinated, and a mutant form of Cx43 that lacked lysines capable of ubiquitination retained the capacity to interact with CIP75. These results suggest that although CIP75 can interact with ubiquitinated cellular proteins, its interaction with Cx43 and stimulation of Cx43 proteasomal degradation does not require the ubiquitination of Cx43.
Collapse
Affiliation(s)
- Vivian Su
- Natural Products and Cancer Biology Program, Cancer Research Center of Hawaii, Honolulu, Hawaii 96813, USA
| | | | | | | |
Collapse
|
43
|
Tsutsumi M, Kitahata H, Nakata S, Sanno Y, Nagayama M, Denda M. Mathematical analysis of intercellular calcium propagation induced by adenosine triphosphate. Skin Res Technol 2010; 16:146-50. [PMID: 20456094 DOI: 10.1111/j.1600-0846.2009.00420.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We previously demonstrated that intracellular calcium propagation was induced by stimulation of epidermal keratinocytes in skin slices or in culture with adenosine triphosphate (ATP). The feature of the calcium wave propagation appeared to be different between differentiated cells and proliferating cells, and so the mechanisms involved might be different. PURPOSE Establish a new methodology to abstract cellular information from aggregative dynamics. METHODS We present a mathematical analysis of the calcium wave to evaluate the mechanism of calcium ion propagation. RESULTS A well-defined calcium wave was observed in differentiated cells in comparison with undifferentiated cells. Application of either 2APB [an inositol 1,4,5-trisphosphate (IP3) receptor blocker] or U73122 (an IP3 synthesis blocker) reduced the amplitude of the wave in differentiated cells. Mathematical analysis indicated that U73122 decreased the velocity of the wave, while 2APB altered the wave form. Thus, IP3 synthesis might be important for signal transmission and IP3 movement might be important for pattern formation. CONCLUSION The method we present here should be useful to analyze the effects of various reagents in in vitro studies.
Collapse
|
44
|
Geneau G, Lamiche C, Niger C, Strale PO, Clarhaut J, Defamie N, Debiais F, Mesnil M, Cronier L. Effect of endothelin-1 on osteoblastic differentiation is modified by the level of connexin43: comparative study on calvarial osteoblastic cells isolated from Cx43+/- and Cx43+/+ mice. Cell Tissue Res 2010; 340:103-15. [PMID: 20195637 DOI: 10.1007/s00441-009-0924-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 12/18/2009] [Indexed: 01/04/2023]
Abstract
Bone is a dynamic tissue that undergoes a precise remodeling process involving resorptive osteoclastic cells and bone-forming osteoblastic (OB) cells. The functional imbalance of either of these cell types can lead to severe skeletal diseases. The proliferation and differentiation of OB cells play a major role in bone development and turnover. These cellular processes are coordinated by connexin43 (Cx43)-based gap-junctional intercellular communication (GJIC) and by soluble factors such as endothelin-1 (ET-1). We have used the Cx43 heterozygous (Cx43(+/-)) murine model to study the possible cross-talk between Cx43 and ET-1 in cultured calvarial OB cells. On microcomputed tomographic analysis of 3-day-old pups, Cx43(+/-) mice showed hypomineralized calvaria in comparison with their Cx43(+/+) littermates. Characterization of cultured OB cells clearly demonstrated the effect of the partial deletion of the Cx43 gene on its expression, on GJIC, and subsequently on OB differentiation. In this model, ET-1 (10(-8) M) lost its mitogenic action in Cx43(+/-) OB cells compared with Cx43(+/+) cells. Moreover, a correlation between the inhibition of cell differentiation by ET-1 and the decreased amount and function of Cx43 was found in Cx43(+/+) OB cells but not in their Cx43(+/-) counterparts. Thus, as Cx43 is linked to OB differentiation, our data indicate that this mitogenic ET-1 peptide has pronounced effects on fully differentiated OB cells. With respect to roles in mechanotransduction and OB differentiation, Cx43 might modulate osteoblastic sensitivity to soluble factors.
Collapse
Affiliation(s)
- Graziello Geneau
- Institut de Physiologie et Biologie Cellulaires, University of Poitiers, CNRS UMR 6187, 86022, Poitiers Cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Laird DW. The gap junction proteome and its relationship to disease. Trends Cell Biol 2010; 20:92-101. [DOI: 10.1016/j.tcb.2009.11.001] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 10/30/2009] [Accepted: 11/02/2009] [Indexed: 02/07/2023]
|
46
|
Chtchetinin J, Gifford WD, Li S, Paznekas WA, Jabs EW, Lai A. Tyrosine-dependent basolateral targeting of human connexin43-eYFP in Madin-Darby canine kidney cells can be disrupted by the oculodentodigital dysplasia mutation L90V. FEBS J 2009; 276:6992-7005. [PMID: 19860828 PMCID: PMC2805759 DOI: 10.1111/j.1742-4658.2009.07407.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Polarized membrane sorting of connexin 43 (Cx43) has not been well-characterized. Based on the presence of a putative sorting signal, YKLV(286-289), within its C-terminal cytoplasmic domain, we hypothesized that Cx43 is selectively expressed on the basolateral surface of Madin-Darby canine kidney (MDCK) cells in a tyrosine-dependent manner. We generated stable MDCK cell lines expressing human wild-type and mutant Cx43-eYFP, and analyzed the membrane localization of Cx43-eYFP within polarized monolayers using confocal microscopy and selective surface biotinylation. We found that wild-type Cx43-eYFP was selectively targeted to the basolateral membrane domain of MDCK cells. Substitution of alanine for Y286 disrupted basolateral targeting of Cx43-eYFP. Additionally, substitution of a sequence containing the transferrin receptor internalization signal, LSYTRF, for PGYKLV(284-289) also disrupted basolateral targeting. Taken together, these results indicate that Y286 in its native amino acid sequence is necessary for targeting Cx43-eYFP to the basolateral membrane domain of MDCK cells. To determine whether the F52dup or L90V oculodentodigital dysplasia-associated mutations could affect polarized sorting of Cx43-eYFP, we analyzed the expression of these Cx43-eYFP mutant constructs and found that the L90V mutation disrupted basolateral expression. These findings raise the possibility that some oculodentodigitial dysplasia-associated mutations contribute to disease by altering polarized targeting of Cx43.
Collapse
Affiliation(s)
- Jana Chtchetinin
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Henry E Singleton Brain Cancer Research Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Wes D. Gifford
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Henry E Singleton Brain Cancer Research Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Sichen Li
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Henry E Singleton Brain Cancer Research Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - William A. Paznekas
- Institute of Genetic Medicine, Johns Hopkins University, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Ethylin Wang Jabs
- Institute of Genetic Medicine, Johns Hopkins University, 733 N. Broadway, Baltimore, MD 21205, USA
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, 1428 Madison Avenue, New York, NY 10029-6574, USA
| | - Albert Lai
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Henry E Singleton Brain Cancer Research Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
47
|
Gerhart SV, Jefferis R, Iovine MK. Cx40.8, a Cx43-like protein, forms gap junction channels inefficiently and may require Cx43 for its association at the plasma membrane. FEBS Lett 2009; 583:3419-24. [PMID: 19808035 DOI: 10.1016/j.febslet.2009.09.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 09/22/2009] [Accepted: 09/26/2009] [Indexed: 11/26/2022]
Abstract
In addition to having a Cx43 ortholog, the zebrafish genome also contains a Cx43-like gene, Cx40.8. Here, we investigate the expression of cx40.8 in zebrafish fins and the function of Cx40.8 in HeLa cells. We find that cx40.8 is present in the same population of dividing cells as cx43. Unlike Cx43, dye coupling assays suggest that Cx40.8 only inefficiently forms functional gap junction channels. However, co-transfection reveals that Cx40.8 can co-localize with Cx43 in gap junction plaques, and that the resulting plaques contain functional gap junction channels. Together, these data suggest the possibility that Cx40.8 may functionally interact with Cx43 to regulate cell proliferation in vivo.
Collapse
Affiliation(s)
- Sarah V Gerhart
- Lehigh University, Department of Biological Sciences, 111 Research Drive, Iacocca B-217, Bethlehem, PA 18015, United States
| | | | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Bone cells such as osteoclasts, osteoblasts, and osteocytes 'talk' to each other throughout adult life to maintain bone integrity. This review highlights frontier areas of research on intercellular communication among bone cells. RECENT FINDINGS Bone cells communicate to regulate the balance between bone resorption and bone formation. Recent findings have suggested that bone remodeling compartment is critical for osteoclast-osteoblast communication during bone remodeling. New molecules and mechanisms for bone cell communication, including 'coupling' of bone formation to resorption, have been revealed. Osteoclastic regulation of pH within the bone remodeling compartment is a mechanism that has been posited to rapidly activate osteoblastic bone formation, whereas osteocytes in the bone matrix regulate osteoclasts and osteoblasts on the bone surface through the lacuno-canaliculi network. SUMMARY Differentiation, activation, and apoptosis of bone cells are often dependent on the status of other types of bone cells. Bone cells in different lineages achieve intercellular communication not only by ligand-receptor interactions but also by molecules and ions traveling in the extracellular space or across gap junctions, processes that depend profoundly on the four-dimensional (space and time) architecture of bone tissue.
Collapse
|
49
|
Myllykoski M, Kuczera K, Kursula P. Complex formation between calmodulin and a peptide from the intracellular loop of the gap junction protein connexin43: Molecular conformation and energetics of binding. Biophys Chem 2009; 144:130-5. [PMID: 19716642 DOI: 10.1016/j.bpc.2009.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 07/31/2009] [Accepted: 08/01/2009] [Indexed: 11/18/2022]
Abstract
Gap junctions are formed by a family of transmembrane proteins, connexins. Connexin43 is a widely studied member of the family, being ubiquitously expressed in a variety of tissues and a target of a large number of disease mutations. The intracellular loop of connexin43 has been shown to include a calmodulin binding domain, but detailed 3-dimensional data on the structure of the complex are not available. In this study, we used a synthetic peptide from this domain to reveal the conformation of the calmodulin-peptide complex by small angle X-ray scattering. Upon peptide binding, calmodulin lost its dumbbell shape, adopting a more globular conformation. We also studied the energetics of the interaction using calorimetry and computational methods. All our data indicate that calmodulin binds to the peptide from cx43 in the classical 'collapsed' conformation.
Collapse
Affiliation(s)
- Matti Myllykoski
- Department of Biochemistry, FIN-90014, University of Oulu, Oulu, Finland
| | | | | |
Collapse
|
50
|
Das S, Smith TD, Sarma JD, Ritzenthaler JD, Maza J, Kaplan BE, Cunningham LA, Suaud L, Hubbard MJ, Rubenstein RC, Koval M. ERp29 restricts Connexin43 oligomerization in the endoplasmic reticulum. Mol Biol Cell 2009; 20:2593-604. [PMID: 19321666 DOI: 10.1091/mbc.e08-07-0790] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Connexin43 (Cx43) is a gap junction protein that forms multimeric channels that enable intercellular communication through the direct transfer of signals and metabolites. Although most multimeric protein complexes form in the endoplasmic reticulum (ER), Cx43 seems to exit from the ER as monomers and subsequently oligomerizes in the Golgi complex. This suggests that one or more protein chaperones inhibit premature Cx43 oligomerization in the ER. Here, we provide evidence that an ER-localized, 29-kDa thioredoxin-family protein (ERp29) regulates Cx43 trafficking and function. Interfering with ERp29 function destabilized monomeric Cx43 oligomerization in the ER, caused increased Cx43 accumulation in the Golgi apparatus, reduced transport of Cx43 to the plasma membrane, and inhibited gap junctional communication. ERp29 also formed a specific complex with monomeric Cx43. Together, this supports a new role for ERp29 as a chaperone that helps stabilize monomeric Cx43 to enable oligomerization to occur in the Golgi apparatus.
Collapse
Affiliation(s)
- Shamie Das
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|