1
|
Costa ADSD, Jeong H, Subbiah R, Park K, Choi IS, Shin JH. Intercellular junction-driven stromal cell stacking in a confined 3D microcavity. APL Bioeng 2024; 8:046109. [PMID: 39525363 PMCID: PMC11549968 DOI: 10.1063/5.0197187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Understanding the detailed mechanisms driving fibroblast migration within native tissue settings during pathophysiological events presents a critical research challenge. In this study, we elucidate how stromal cells migrate and contribute to the development of three-dimensional (3D) cellular aggregates within confined microcavities. Integrin α5β1 and β-catenin (β-cat) are central in guiding this collective migration and achieving optimal filling of the microcavity. When β-cat is suppressed, cells tend to migrate more sporadically, leading to less efficient cellular organization. Furthermore, we also detail the pivotal roles of Cx43 and N-cadherin (N-cad) in orchestrating collective migration and in shaping efficient cellular stacking. Suppressing gap junctions, especially Cx43, significantly impacts the extracellular matrix expression, integrin α5 and β1, and other elements in the 3D construct, emphasizing the importance of physicochemical cell-cell interactions. The distribution patterns of N-cad and focal adhesion kinase (FAK) further corroborate the essential roles in forming cell-cell junctions and FAK in establishing the foundational layer that underpins the cell stacking within the microcavity. Interestingly, neither Rho-associated protein kinase (ROCK) nor RhoA significantly alter the cell migration pattern toward microcavity. These findings provide fresh perspectives on fibroblast activities in 3D space, enriching our understanding and offering implications for advancements in wound healing and tissue engineering.
Collapse
Affiliation(s)
| | - Hyuntae Jeong
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Ramesh Subbiah
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University (OHSU), Portland, Oregon 97201, USA
| | | | - In-Suk Choi
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jennifer H. Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
2
|
Ghézali G, Ribot J, Curry N, Pillet LE, Boutet-Porretta F, Mozheiko D, Calvo CF, Ezan P, Perfettini I, Lecoin L, Janel S, Zapata J, Escartin C, Etienne-Manneville S, Kaminski CF, Rouach N. Connexin 30 locally controls actin cytoskeleton and mechanical remodeling in motile astrocytes. Glia 2024; 72:1915-1929. [PMID: 38982826 DOI: 10.1002/glia.24590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/04/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024]
Abstract
During brain maturation, astrocytes establish complex morphologies unveiling intense structural plasticity. Connexin 30 (Cx30), a gap-junction channel-forming protein expressed postnatally, dynamically regulates during development astrocyte morphological properties by controlling ramification and extension of fine processes. However, the underlying mechanisms remain unexplored. Here, we found in vitro that Cx30 interacts with the actin cytoskeleton in astrocytes and inhibits its structural reorganization and dynamics during cell migration. This translates into an alteration of local physical surface properties, as assessed by correlative imaging using stimulated emission depletion (STED) super resolution imaging and atomic force microscopy (AFM). Specifically, Cx30 impaired astrocyte cell surface topology and cortical stiffness in motile astrocytes. As Cx30 alters actin organization, dynamics, and membrane physical properties, we assessed whether it controls astrocyte migration. We found that Cx30 reduced persistence and directionality of migrating astrocytes. Altogether, these data reveal Cx30 as a brake for astrocyte structural and mechanical plasticity.
Collapse
Affiliation(s)
- Grégory Ghézali
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Doctoral School N° 158, Sorbonne Université, Paris, France
| | - Jérôme Ribot
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Nathan Curry
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Laure-Elise Pillet
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Doctoral School N°562, Université Paris Cité, Paris, France
| | - Flora Boutet-Porretta
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Doctoral School N° 158, Sorbonne Université, Paris, France
| | - Daria Mozheiko
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Doctoral School N° 158, Sorbonne Université, Paris, France
| | - Charles-Félix Calvo
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Pascal Ezan
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Isabelle Perfettini
- Institut Pasteur, Université de Paris, CNRS, Cell Polarity, Migration and Cancer Unit, Paris, France
| | - Laure Lecoin
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Sébastien Janel
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Jonathan Zapata
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | | | - Clemens F Kaminski
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Cárdenas-García SP, Ijaz S, Pereda AE. The components of an electrical synapse as revealed by expansion microscopy of a single synaptic contact. eLife 2024; 13:e91931. [PMID: 38994821 PMCID: PMC11333041 DOI: 10.7554/elife.91931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
Most nervous systems combine both transmitter-mediated and direct cell-cell communication, known as 'chemical' and 'electrical' synapses, respectively. Chemical synapses can be identified by their multiple structural components. Electrical synapses are, on the other hand, generally defined by the presence of a 'gap junction' (a cluster of intercellular channels) between two neuronal processes. However, while gap junctions provide the communicating mechanism, it is unknown whether electrical transmission requires the contribution of additional cellular structures. We investigated this question at identifiable single synaptic contacts on the zebrafish Mauthner cells, at which gap junctions coexist with specializations for neurotransmitter release and where the contact unequivocally defines the anatomical limits of a synapse. Expansion microscopy of these single contacts revealed a detailed map of the incidence and spatial distribution of proteins pertaining to various synaptic structures. Multiple gap junctions of variable size were identified by the presence of their molecular components. Remarkably, most of the synaptic contact's surface was occupied by interleaving gap junctions and components of adherens junctions, suggesting a close functional association between these two structures. In contrast, glutamate receptors were confined to small peripheral portions of the contact, indicating that most of the synaptic area functions as an electrical synapse. Thus, our results revealed the overarching organization of an electrical synapse that operates with not one, but multiple gap junctions, in close association with structural and signaling molecules known to be components of adherens junctions. The relationship between these intercellular structures will aid in establishing the boundaries of electrical synapses found throughout animal connectomes and provide insight into the structural organization and functional diversity of electrical synapses.
Collapse
Affiliation(s)
- Sandra P Cárdenas-García
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Sundas Ijaz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| |
Collapse
|
4
|
Ebrahim T, Ebrahim AS, Kandouz M. Diversity of Intercellular Communication Modes: A Cancer Biology Perspective. Cells 2024; 13:495. [PMID: 38534339 DOI: 10.3390/cells13060495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/27/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024] Open
Abstract
From the moment a cell is on the path to malignant transformation, its interaction with other cells from the microenvironment becomes altered. The flow of molecular information is at the heart of the cellular and systemic fate in tumors, and various processes participate in conveying key molecular information from or to certain cancer cells. For instance, the loss of tight junction molecules is part of the signal sent to cancer cells so that they are no longer bound to the primary tumors and are thus free to travel and metastasize. Upon the targeting of a single cell by a therapeutic drug, gap junctions are able to communicate death information to by-standing cells. The discovery of the importance of novel modes of cell-cell communication such as different types of extracellular vesicles or tunneling nanotubes is changing the way scientists look at these processes. However, are they all actively involved in different contexts at the same time or are they recruited to fulfill specific tasks? What does the multiplicity of modes mean for the overall progression of the disease? Here, we extend an open invitation to think about the overall significance of these questions, rather than engage in an elusive attempt at a systematic repertory of the mechanisms at play.
Collapse
Affiliation(s)
- Thanzeela Ebrahim
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Mustapha Kandouz
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48202, USA
| |
Collapse
|
5
|
Lin CY, Chang YM, Tseng HY, Shih YL, Yeh HH, Liao YR, Tang HH, Hsu CL, Chen CC, Yan YT, Kao CF. Epigenetic regulator RNF20 underlies temporal hierarchy of gene expression to regulate postnatal cardiomyocyte polarization. Cell Rep 2023; 42:113416. [PMID: 37967007 DOI: 10.1016/j.celrep.2023.113416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 09/19/2023] [Accepted: 10/25/2023] [Indexed: 11/17/2023] Open
Abstract
Differentiated cardiomyocytes (CMs) must undergo diverse morphological and functional changes during postnatal development. However, the mechanisms underlying initiation and coordination of these changes remain unclear. Here, we delineate an integrated, time-ordered transcriptional network that begins with expression of genes for cell-cell connections and leads to a sequence of structural, cell-cycle, functional, and metabolic transitions in mouse postnatal hearts. Depletion of histone H2B ubiquitin ligase RNF20 disrupts this gene network and impairs CM polarization. Subsequently, assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis confirmed that RNF20 contributes to chromatin accessibility in this context. As such, RNF20 is likely to facilitate binding of transcription factors at the promoters of genes involved in cell-cell connections and actin organization, which are crucial for CM polarization and functional integration. These results suggest that CM polarization is one of the earliest events during postnatal heart development and provide insights into how RNF20 regulates CM polarity and the postnatal gene program.
Collapse
Affiliation(s)
- Chia-Yeh Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsin-Yi Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yen-Ling Shih
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Hsiao-Hui Yeh
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - You-Rou Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Han-Hsuan Tang
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chia-Ling Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, 128, Academia Road, Section 2, Nankang, Taipei, Taiwan.
| |
Collapse
|
6
|
Cárdenas-García SP, Ijaz S, Pereda AE. The components of an electrical synapse as revealed by expansion microscopy of a single synaptic contact. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550347. [PMID: 37546897 PMCID: PMC10402082 DOI: 10.1101/2023.07.25.550347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Most nervous systems combine both transmitter-mediated and direct cell-cell communication, known as 'chemical' and 'electrical' synapses, respectively. Chemical synapses can be identified by their multiple structural components. Electrical synapses are, on the other hand, generally defined by the presence of a 'gap junction' (a cluster of intercellular channels) between two neuronal processes. However, while gap junctions provide the communicating mechanism, it is unknown whether electrical transmission requires the contribution of additional cellular structures. We investigated this question at identifiable single synaptic contacts on the zebrafish Mauthner cells, at which gap junctions coexist with specializations for neurotransmitter release and where the contact defines the anatomical limits of a synapse. Expansion microscopy of these contacts revealed a detailed map of the incidence and spatial distribution of proteins pertaining to various synaptic structures. Multiple gap junctions of variable size were identified by the presence of their molecular components. Remarkably, most of the synaptic contact's surface was occupied by interleaving gap junctions and components of adherens junctions, suggesting a close functional association between these two structures. In contrast, glutamate receptors were confined to small peripheral portions of the contact, indicating that most of the synaptic area works as an electrical synapse. Thus, our results revealed the overarching organization of an electrical synapse that operates with not one, but multiple gap junctions, in close association with structural and signaling molecules known to be components of AJs. The relationship between these intercellular structures will aid in establishing the boundaries of electrical synapses found throughout animal connectomes and provide insight into the structural organization and functional diversity of electrical synapses.
Collapse
Affiliation(s)
- Sandra P. Cárdenas-García
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sundas Ijaz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Alberto E. Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
7
|
Serra R, Simard JM. Adherens, tight, and gap junctions in ependymal cells: A systematic review of their contribution to CSF-brain barrier. Front Neurol 2023; 14:1092205. [PMID: 37034077 PMCID: PMC10079940 DOI: 10.3389/fneur.2023.1092205] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction The movement of fluids and solutes across the ependymal barrier, and their changes in physiologic and disease states are poorly understood. This gap in knowledge contributes strongly to treatment failures and complications in various neurological disorders. Methods We systematically searched and reviewed original research articles treating ependymal intercellular junctions on PubMed. Reviews, opinion papers, and abstracts were excluded. Research conducted on tissue samples, cell lines, CSF, and animal models was considered. Results A total of 45 novel articles treating tight, adherens and gap junctions of the ependyma were included in our review, spanning from 1960 to 2022. The findings of this review point toward a central and not yet fully characterized role of the ependymal lining ultrastructure in fluid flow interactions in the brain. In particular, tight junctions circumferentially line the apical equator of ependymal cells, changing between embryonal and adult life in several rodent models, shaping fluid and solute transit in this location. Further, adherens and gap junctions appear to have a pivotal role in several forms of congenital hydrocephalus. Conclusions These findings may provide an opportunity for medical management of CSF disorders, potentially allowing for tuning of CSF secretion and absorption. Beyond hydrocephalus, stroke, trauma, this information has relevance for metabolite clearance and drug delivery, with potential to affect many patients with a variety of neurological disorders. This critical look at intercellular junctions in ependyma and the surrounding interstitial spaces is meant to inspire future research on a central and rather unknown component of the CSF-brain interface.
Collapse
Affiliation(s)
- Riccardo Serra
- Department of Neurosurgery, University of Maryland, Baltimore, MD, United States
- *Correspondence: Riccardo Serra
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland, Baltimore, MD, United States
- Department of Pathology, University of Maryland, Baltimore, MD, United States
- Department of Physiology, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
8
|
Stem cell laden nano and micro collagen/PLGA bimodal fibrous patches for myocardial regeneration. Biomater Res 2022; 26:79. [PMID: 36514148 DOI: 10.1186/s40824-022-00319-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/13/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Although the use of cardiac patches is still controversial, cardiac patch has the significance in the field of the tissue engineered cardiac regeneration because it overcomes several shortcomings of intra-myocardial injection by providing a template for cells to form a cohesive sheet. So far, fibrous scaffolds fabricated using electrospinning technique have been increasingly explored for preparation of cardiac patches. One of the problems with the use of electrospinning is that nanofibrous structures hardly allow the infiltration of cells for development of 3D tissue construct. In this respect, we have prepared novel bi-modal electrospun scaffolds as a feasible strategy to address the challenges in cardiac tissue engineering . METHODS Nano/micro bimodal composite fibrous patch composed of collagen and poly (D, L-lactic-co-glycolic acid) (Col/PLGA) was fabricated using an independent nozzle control multi-electrospinning apparatus, and its feasibility as the stem cell laden cardiac patch was systemically investigated. RESULTS Nano/micro bimodal distributions of Col/PLGA patches without beaded fibers were obtained in the range of the 4-6% collagen concentration. The poor mechanical properties of collagen and the hydrophobic property of PLGA were improved by co-electrospinning. In vitro experiments using bone marrow-derived mesenchymal stem cells (BMSCs) revealed that Col/PLGA showed improved cyto-compatibility and proliferation capacity compared to PLGA, and their extent increased with increase in collagen content. The results of tracing nanoparticle-labeled as well as GFP transfected BMSCs strongly support that Col/PLGA possesses the long-term stem cells retention capability, thereby allowing stem cells to directly function as myocardial and vascular endothelial cells or to secrete the recovery factors, which in turn leads to improved heart function proved by histological and echocardiographic findings. CONCLUSION Col/PLGA bimodal cardiac patch could significantly attenuate cardiac remodeling and fully recover the cardiac function, as a consequence of their potent long term stem cell engraftment capability.
Collapse
|
9
|
Roads to Stat3 Paved with Cadherins. Cells 2022; 11:cells11162537. [PMID: 36010614 PMCID: PMC9406956 DOI: 10.3390/cells11162537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
The engagement of cadherins, cell-to-cell adhesion proteins, triggers a dramatic increase in the levels and activity of the Rac/Cdc42 GTPases, through the inhibition of proteasomal degradation. This leads to an increase in transcription and secretion of IL6 family cytokines, activation of their common receptor, gp130, in an autocrine manner and phosphorylation of the signal transducer and activator of transcription-3 (Stat3) on tyrosine-705 by the Jak kinases. Stat3 subsequently dimerizes, migrates to the nucleus and activates the transcription of genes involved in cell division and survival. The Src oncogene also increases Rac levels, leading to secretion of IL6 family cytokines and gp130 activation, which triggers a Stat3-ptyr705 increase. Interestingly, at the same time, Src downregulates cadherins in a quantitative manner, while cadherins are required to preserve gp130 levels for IL6 family signalling. Therefore, a fine balance between Src527F/Rac/IL6 and Src527F/cadherin/gp130 levels is in existence, which is required for Stat3 activation. This further demonstrates the important role of cadherins in the activation of Stat3, through preservation of gp130 function. Conversely, the absence of cadherin engagement correlates with low Stat3 activity: In sparsely growing cells, both gp130 and Stat3-ptyr705 levels are very low, despite the fact that cSrc is active in the FAK (focal adhesion kinase)/cSrc complex, which further indicates that the engagement of cadherins is important for Stat3 activation, not just their presence. Furthermore, the caveolin-1 protein downregulates Stat3 through binding and sequestration of cadherins to the scaffolding domain of caveolin-1. We hypothesize that the cadherins/Rac/gp130 axis may be a conserved pathway to Stat3 activation in a number of systems. This fact could have significant implications in Stat3 biology, as well as in drug testing and development.
Collapse
|
10
|
Interaction of Glia Cells with Glioblastoma and Melanoma Cells under the Influence of Phytocannabinoids. Cells 2022; 11:cells11010147. [PMID: 35011711 PMCID: PMC8750637 DOI: 10.3390/cells11010147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/10/2021] [Accepted: 12/31/2021] [Indexed: 01/27/2023] Open
Abstract
Brain tumor heterogeneity and progression are subject to complex interactions between tumor cells and their microenvironment. Glioblastoma and brain metastasis can contain 30–40% of tumor-associated macrophages, microglia, and astrocytes, affecting migration, proliferation, and apoptosis. Here, we analyzed interactions between glial cells and LN229 glioblastoma or A375 melanoma cells in the context of motility and cell–cell interactions in a 3D model. Furthermore, the effects of phytocannabinoids, cannabidiol (CBD), tetrahydrocannabidiol (THC), or their co-application were analyzed. Co-culture of tumor cells with glial cells had little effect on 3D spheroid formation, while treatment with cannabinoids led to significantly larger spheroids. The addition of astrocytes blocked cannabinoid-induced effects. None of the interventions affected cell death. Furthermore, glial cell-conditioned media led to a significant slowdown in collective, but not single-cell migration speed. Taken together, glial cells in glioblastoma and brain metastasis micromilieu impact the tumor spheroid formation, cell spreading, and motility. Since the size of spheroid remained unaffected in glial cell tumor co-cultures, phytocannabinoids increased the size of spheroids without any effects on migration. This aspect might be of relevance since phytocannabinoids are frequently used in tumor therapy for side effects.
Collapse
|
11
|
Unal YC, Yavuz B, Ozcivici E, Mese G. The role of connexins in breast cancer: from misregulated cell communication to aberrant intracellular signaling. Tissue Barriers 2022; 10:1962698. [PMID: 34355641 PMCID: PMC8794248 DOI: 10.1080/21688370.2021.1962698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
In spite of clinical advancements and improved diagnostic techniques, breast cancers are the leading cause of cancer-associated deaths in women worldwide. Although 70% of early breast cancers can be cured, there are no efficient therapies against metastatic breast cancers. Several factors including connexins and gap junctions play roles in breast tumorigenesis. Connexins are critical for cellular processes as a linkage between connexin mutations and hereditary disorders demonstrated their importance for tissue homeostasis. Further, alterations in their expression, localization and channel activities were observed in many cancers including breast cancer. Both channel-dependent and independent functions of connexins were reported in initiation and progression of cancers. Unlike initial reports suggesting tumor suppressor functions, connexins and gap junctions have stage, context and isoform dependent effects in breast cancers similar to other cancers. In this review, we tried to describe the current understanding of connexins in tumorigenesis specifically in breast cancers.
Collapse
Affiliation(s)
- Yagmur Ceren Unal
- Faculty of Science, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Busra Yavuz
- Faculty of Science, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Engin Ozcivici
- Department of Bioengineering, Faculty of Engineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Gulistan Mese
- Faculty of Science, Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, Turkey
| |
Collapse
|
12
|
Connexin 43 Gene Ablation Does Not Alter Human Pluripotent Stem Cell Germ Lineage Specification. Biomolecules 2021; 12:biom12010015. [PMID: 35053163 PMCID: PMC8773696 DOI: 10.3390/biom12010015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/23/2023] Open
Abstract
During embryonic germ layer development, cells communicate with each other and their environment to ensure proper lineage specification and tissue development. Connexin (Cx) proteins facilitate direct cell–cell communication through gap junction channels. While previous reports suggest that gap junctional intercellular communication may contribute to germ layer formation, there have been limited comprehensive expression analyses or genetic ablation studies on Cxs during human pluripotent stem cell (PSC) germ lineage specification. We screened the mRNA profile and protein expression patterns of select human Cx isoforms in undifferentiated human induced pluripotent stem cells (iPSCs), and after directed differentiation into the three embryonic germ lineages: ectoderm, definitive endoderm, and mesoderm. Transcript analyses by qPCR revealed upregulation of Cx45 and Cx62 in iPSC-derived ectoderm; Cx45 in mesoderm; and Cx30.3, Cx31, Cx32, Cx36, Cx37, and Cx40 in endoderm relative to control human iPSCs. Generated Cx43 (GJA1) CRISPR-Cas9 knockout iPSCs successfully differentiated into cells of all three germ layers, suggesting that Cx43 is dispensable during directed iPSC lineage specification. Furthermore, qPCR screening of select Cx transcripts in our GJA1-/- iPSCs showed no significant Cx upregulation in response to the loss of Cx43 protein. Future studies will reveal possible compensation by additional Cxs, suggesting targets for future CRISPR-Cas9 ablation studies in human iPSC lineage specification.
Collapse
|
13
|
Ferroptosis Meets Cell-Cell Contacts. Cells 2021; 10:cells10092462. [PMID: 34572111 PMCID: PMC8471828 DOI: 10.3390/cells10092462] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/15/2022] Open
Abstract
Ferroptosis is a regulated form of cell death characterized by iron dependency and increased lipid peroxidation. Initially assumed to be selectively induced in tumour cells, there is increasing evidence that ferroptosis plays an important role in pathophysiology and numerous cell types and tissues. Deregulated ferroptosis has been linked to human diseases, such as neurodegenerative diseases, cardiovascular disorders, and cancer. Along these lines, ferroptosis is a promising pathway to overcoming therapy resistance of cancer cells. It is therefore of utmost importance to understand the cellular signalling pathways and the molecular mechanisms underlying ferroptosis regulation, including context-specific effects mediated by the neighbouring cells through cell–cell contacts. Here, we give an overview on the molecular events and machinery linked to ferroptosis induction and commitment. We further summarize and discuss current knowledge about the role of cell–cell contacts, which differ in ferroptosis regulation between normal somatic cells and cancer cells. We present emerging concepts on the underlying mechanisms, address open questions, and discuss the possible impact of cell–cell contacts on exploiting ferroptosis in cancer therapy.
Collapse
|
14
|
Desmin Correlated with Cx43 May Facilitate Intercellular Electrical Coupling during Chronic Heart Failure. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6621132. [PMID: 34285704 PMCID: PMC8275391 DOI: 10.1155/2021/6621132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/16/2021] [Accepted: 05/18/2021] [Indexed: 01/17/2023]
Abstract
Desmin is one of five major intermediate filament proteins in cardiomyocytes. Desmin contributes to the maintenance of healthy muscle. The desmin content in cardiomyocytes directly affects the long-term prognosis of patients with heart failure, and lack of desmin leads to myocyte contractile dysfunction. However, the mechanism is elusive. In this study, we measured desmin expression using western blotting and qPCR in the failed hearts of human patients and rats. Our results showed that desmin content was reduced at the protein level in failed hearts and isolated cardiomyocytes. The association of desmin and the gap junction proteins connexin 43 (Cx43) and zonula occludens-1 (ZO-1) was also investigated. Immunoprecipitation assay showed that desmin was associated with Cx43 in cardiomyocytes. To compare the electrical integration of skeletal myoblasts in cocultures with cardiac myocytes, familial amyloid polyneuropathy (FAP) activation rate was found in 33% desmin overexpressing skeletal myoblasts. Desmin not only affected Cx43 and ZO-1 expression but also facilitated the complex of Cx43 and ZO-1 in skeletal myoblasts, which enhanced cell-to-cell electrical coupling of skeletal myoblasts with cardiac myocytes. Desmin has potential as a novel therapeutic target for heart failure. Preservation of desmin may attenuate heart failure.
Collapse
|
15
|
Sinha G, Ferrer AI, Ayer S, El-Far MH, Pamarthi SH, Naaldijk Y, Barak P, Sandiford OA, Bibber BM, Yehia G, Greco SJ, Jiang JG, Bryan M, Kumar R, Ponzio NM, Etchegaray JP, Rameshwar P. Specific N-cadherin-dependent pathways drive human breast cancer dormancy in bone marrow. Life Sci Alliance 2021; 4:4/7/e202000969. [PMID: 34078741 PMCID: PMC8200294 DOI: 10.26508/lsa.202000969] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/19/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
The challenge for treating breast cancer (BC) is partly due to long-term dormancy driven by cancer stem cells (CSCs) capable of evading immune response and resist chemotherapy. BC cells show preference for the BM, resulting in poor prognosis. CSCs use connexin 43 (Cx43) to form gap junctional intercellular communication with BM niche cells, fibroblasts, and mesenchymal stem cells (MSCs). However, Cx43 is an unlikely target to reverse BC dormancy because of its role as a hematopoietic regulator. We found N-cadherin (CDH2) and its associated pathways as potential drug targets. CDH2, highly expressed in CSCs, interacts intracellularly with Cx43, colocalizes with Cx43 in BC cells within BM biopsies of patients, and is required for Cx43-mediated gap junctional intercellular communication with BM niche cells. Notably, CDH2 and anti-apoptotic pathways maintained BC dormancy. We thereby propose these pathways as potential pharmacological targets to prevent dormancy and chemosensitize resistant CSCs.
Collapse
Affiliation(s)
- Garima Sinha
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA.,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Alejandra I Ferrer
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA.,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Seda Ayer
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Markos H El-Far
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA.,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Sri Harika Pamarthi
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Yahaira Naaldijk
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Pradeep Barak
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.,ONI, Linacre House, Oxford, UK
| | - Oleta A Sandiford
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Bernadette M Bibber
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA.,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Ghassan Yehia
- Genome Editing Shared Resource, Office of Research and Economic Development, Rutgers University, New Brunswick, NJ, USA
| | - Steven J Greco
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Jie-Gen Jiang
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.,ONI, Linacre House, Oxford, UK
| | - Margarette Bryan
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Rakesh Kumar
- Department of Biotechnology, Rajiv Gandhi Centre for Biotechnology, Kerala, India
| | - Nicholas M Ponzio
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.,ONI, Linacre House, Oxford, UK
| | | | - Pranela Rameshwar
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA .,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| |
Collapse
|
16
|
Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int J Mol Sci 2021; 22:ijms22094413. [PMID: 33922534 PMCID: PMC8122935 DOI: 10.3390/ijms22094413] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Collapse
|
17
|
Yang Y, Liu W, Wei J, Cui Y, Zhang D, Xie J. Transforming growth factor-β1-induced N-cadherin drives cell-cell communication through connexin43 in osteoblast lineage. Int J Oral Sci 2021; 13:15. [PMID: 33850101 PMCID: PMC8044142 DOI: 10.1038/s41368-021-00119-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/29/2020] [Accepted: 02/01/2021] [Indexed: 02/05/2023] Open
Abstract
Gap junction (GJ) has been indicated to have an intimate correlation with adhesion junction. However, the direct interaction between them partially remains elusive. In the current study, we aimed to elucidate the role of N-cadherin, one of the core components in adhesion junction, in mediating connexin 43, one of the functional constituents in gap junction, via transforming growth factor-β1(TGF-β1) induction in osteoblasts. We first elucidated the expressions of N-cadherin induced by TGF-β1 and also confirmed the upregulation of Cx43, and the enhancement of functional gap junctional intercellular communication (GJIC) triggered by TGF-β1 in both primary osteoblasts and MC3T3 cell line. Colocalization analysis and Co-IP experimentation showed that N-cadherin interacts with Cx43 at the site of cell-cell contact. Knockdown of N-cadherin by siRNA interference decreased the Cx43 expression and abolished the promoting effect of TGF-β1 on Cx43. Functional GJICs in living primary osteoblasts and MC3T3 cell line were also reduced. TGF-β1-induced increase in N-cadherin and Cx43 was via Smad3 activation, whereas knockdown of Smad3 signaling by using siRNA decreased the expressions of both N-cadherin and Cx43. Overall, these data indicate the direct interactions between N-cadherin and Cx43, and reveal the intervention of adhesion junction in functional gap junction in living osteoblasts.
Collapse
Affiliation(s)
- Yueyi Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenjing Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - JieYa Wei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Xu W, Dielubanza E, Maisel A, Leung K, Mustoe T, Hong S, Galiano R. Staphylococcus aureus impairs cutaneous wound healing by activating the expression of a gap junction protein, connexin-43 in keratinocytes. Cell Mol Life Sci 2021; 78:935-947. [PMID: 32409862 PMCID: PMC11072219 DOI: 10.1007/s00018-020-03545-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 10/24/2022]
Abstract
Chronic wounds have been considered as major medical problems that may result in expensive healthcare. One of the common causes of chronic wounds is bacterial contamination that leads to persistent inflammation and unbalanced host cell immune responses. Among the bacterial strains that have been identified from chronic wounds, Staphylococcus aureus is the most common strain. We previously observed that S. aureus impaired mouse cutaneous wound healing by delaying re-epithelialization. Here, we investigated the mechanism of delayed re-epithelialization caused by S. aureus infection. With the presence of S. aureus exudate, the migration of in vitro cultured human keratinocytes was significantly inhibited and connexin-43 (Cx43) was upregulated. Inhibition of keratinocyte migration by S. aureus exudate disappeared in keratinocytes where the expression of Cx43 knocked down. Protein kinase phosphorylation array showed that phosphorylation of Akt-S473 was upregulated by S. aureus exudate. In vivo study of Cx43 in S. aureus-infected murine splinted cutaneous wound model showed upregulation of Cx43 in the migrating epithelial edge by S. aureus infection. Treatment with a PI3K/Akt inhibitor reduced Cx43 expression and overcame the wound closure impairment by S. aureus infection in the mouse model. This may contribute to the development of treatment to bacterium-infected wounds.
Collapse
Affiliation(s)
- Wei Xu
- Department of Life Sciences, College of Science and Engineering, Texas A&M University-Corpus Christi, Corpus Christi, TX, 78412, USA.
| | - Elodi Dielubanza
- Laboratory for Wound Repair and Regenerative Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Amanda Maisel
- Laboratory for Wound Repair and Regenerative Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Kai Leung
- Division of Combat Wound Repair, US Army Institute of Surgical Research, JB Fort Sam Houston, San Antonio, TX, 78234, USA
| | - Thomas Mustoe
- Laboratory for Wound Repair and Regenerative Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Seok Hong
- Laboratory for Wound Repair and Regenerative Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Robert Galiano
- Laboratory for Wound Repair and Regenerative Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
19
|
Wan L, Jiang D, Correa-Gallegos D, Ramesh P, Zhao J, Ye H, Zhu S, Wannemacher J, Volz T, Rinkevich Y. Connexin43 gap junction drives fascia mobilization and repair of deep skin wounds. Matrix Biol 2021; 97:58-71. [PMID: 33508427 DOI: 10.1016/j.matbio.2021.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 12/23/2022]
Abstract
Deep and voluminous skin wounds are repaired with scars, by mobilization of fibroblasts and extracellular matrix from fascia, deep below the skin. The molecular trigger of this novel repair mechanism is incompletely understood. Here we reveal that the gap junction alpha-1 protein (Connexin43, Cx43) is the key to patch repair of deep wounds. By combining full-thickness wound models with fibroblast lineage specific transgenic lines, we show Cx43 expression is substantially upregulated in specialized fibroblasts of the fascia deep beneath the skin that are responsible for scar formation. Using live imaging of fascia fibroblasts and fate tracing of the fascia extracellular matrix we show that Cx43 inhibition disrupts calcium oscillations in cultured fibroblasts and that this inhibits collective migration of fascia EPFs necessary to mobilize fascia matrix into open wounds. Cell-cell communication through Cx43 thus mediates matrix movement and scar formation, and is necessary for patch repair of voluminous wounds. These mechanistic findings have broad clinical implications toward treating fibrosis, aggravated scarring and impaired wound healing.
Collapse
Affiliation(s)
- Li Wan
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany
| | - Dongsheng Jiang
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany
| | - Donovan Correa-Gallegos
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany
| | - Pushkar Ramesh
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany
| | - Jiakuan Zhao
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany
| | - Haifeng Ye
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany
| | - Shaohua Zhu
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany
| | - Juliane Wannemacher
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany
| | - Thomas Volz
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Dermatology and Allergology, Munich, Germany
| | - Yuval Rinkevich
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Munich, Germany; Helmholtz Zentrum München, Institute of Regenerative Biology and Medicine, Munich, Germany.
| |
Collapse
|
20
|
Ouabain Promotes Gap Junctional Intercellular Communication in Cancer Cells. Int J Mol Sci 2020; 22:ijms22010358. [PMID: 33396341 PMCID: PMC7801950 DOI: 10.3390/ijms22010358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/17/2022] Open
Abstract
Gap junctions are molecular structures that allow communication between neighboring cells. It has been shown that gap junctional intercellular communication (GJIC) is notoriously reduced in cancer cells compared to their normal counterparts. Ouabain, a plant derived substance, widely known for its therapeutic properties on the heart, has been shown to play a role in several types of cancer, although its mechanism of action is not yet fully understood. Since we have previously shown that ouabain enhances GJIC in epithelial cells (MDCK), here we probed whether ouabain affects GJIC in a variety of cancer cell lines, including cervico-uterine (CasKi, SiHa and Hela), breast (MDA-MB-321 and MCF7), lung (A549), colon (SW480) and pancreas (HPAF-II). For this purpose, we conducted dye transfer assays to measure and compare GJIC in monolayers of cells with and without treatment with ouabain (0.1, 1, 10, 50 and 500 nM). We found that ouabain induces a statistically significant enhancement of GJIC in all of these cancer cell lines, albeit with distinct sensitivity. Additionally, we show that synthesis of new nucleotides or protein subunits is not required, and that Csrc, ErK1/2 and ROCK-Rho mediate the signaling mechanisms. These results may contribute to explaining how ouabain influences cancer.
Collapse
|
21
|
Meng L, Yan D. NLR-1/CASPR Anchors F-Actin to Promote Gap Junction Formation. Dev Cell 2020; 55:574-587.e3. [PMID: 33238150 DOI: 10.1016/j.devcel.2020.10.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/05/2020] [Accepted: 10/28/2020] [Indexed: 12/31/2022]
Abstract
Gap junctions are present in most tissues and play essential roles in various biological processes. However, we know surprisingly little about the molecular mechanisms underlying gap junction formation. Here, we uncover the essential role of a conserved EGF- and laminin-G-domain-containing protein nlr-1/CASPR in the regulation of gap junction formation in multiple tissues across different developmental stages in C. elegans. NLR-1 is located in the gap junction perinexus, a region adjacent to but not overlapping with gap junctions, and forms puncta before the clusters of gap junction channels appear on the membrane. We show that NLR-1 can directly bind to actin to recruit F-actin networks at the gap junction formation plaque, and the formation of F-actin patches plays a critical role in the assembly of gap junction channels. Our findings demonstrate that nlr-1/CASPR acts as an early stage signal for gap junction formation through anchoring of F-actin networks.
Collapse
Affiliation(s)
- Lingfeng Meng
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Dong Yan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Regeneration Next, and Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
22
|
Muscarella AM, Dai W, Mitchell PG, Zhang W, Wang H, Jia L, Stossi F, Mancini MA, Chiu W, Zhang XHF. Unique cellular protrusions mediate breast cancer cell migration by tethering to osteogenic cells. NPJ Breast Cancer 2020; 6:42. [PMID: 32964116 PMCID: PMC7477119 DOI: 10.1038/s41523-020-00183-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Migration and invasion are key properties of metastatic cancer cells. These properties can be acquired through intrinsic reprogramming processes such as epithelial-mesenchymal transition. In this study, we discovered an alternative "migration-by-tethering" mechanism through which cancer cells gain the momentum to migrate by adhering to mesenchymal stem cells or osteoblasts. This tethering is mediated by both heterotypic adherens junctions and gap junctions, and leads to a unique cellular protrusion supported by cofilin-coated actin filaments. Inhibition of gap junctions or depletion of cofilin reduces migration-by-tethering. We observed evidence of these protrusions in bone segments harboring experimental and spontaneous bone metastasis in animal models. These data exemplify how cancer cells may acquire migratory ability without intrinsic reprogramming. Furthermore, given the important roles of osteogenic cells in early-stage bone colonization, our observations raise the possibility that migration-by-tethering may drive the relocation of disseminated tumor cells between different niches in the bone microenvironment.
Collapse
Affiliation(s)
- Aaron M. Muscarella
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Wei Dai
- Department of Cell Biology and Neuroscience, Institute for Quantitative Biomedicine, Rutgers University, 174 Frelinghuysen Road, Piscataway, NJ 08854 USA
| | - Patrick G. Mitchell
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- National Center for Macromolecular Imaging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305 USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Luyu Jia
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Fabio Stossi
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Michael A. Mancini
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Wah Chiu
- Department of Bioengineering, Stanford University, Stanford, CA 94305 USA
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| |
Collapse
|
23
|
Liu W, Cui Y, Wei J, Sun J, Zheng L, Xie J. Gap junction-mediated cell-to-cell communication in oral development and oral diseases: a concise review of research progress. Int J Oral Sci 2020; 12:17. [PMID: 32532966 PMCID: PMC7293327 DOI: 10.1038/s41368-020-0086-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/30/2020] [Accepted: 05/19/2020] [Indexed: 02/05/2023] Open
Abstract
Homoeostasis depends on the close connection and intimate molecular exchange between extracellular, intracellular and intercellular networks. Intercellular communication is largely mediated by gap junctions (GJs), a type of specialized membrane contact composed of variable number of channels that enable direct communication between cells by allowing small molecules to pass directly into the cytoplasm of neighbouring cells. Although considerable evidence indicates that gap junctions contribute to the functions of many organs, such as the bone, intestine, kidney, heart, brain and nerve, less is known about their role in oral development and disease. In this review, the current progress in understanding the background of connexins and the functions of gap junctions in oral development and diseases is discussed. The homoeostasis of tooth and periodontal tissues, normal tooth and maxillofacial development, saliva secretion and the integrity of the oral mucosa depend on the proper function of gap junctions. Knowledge of this pattern of cell-cell communication is required for a better understanding of oral diseases. With the ever-increasing understanding of connexins in oral diseases, therapeutic strategies could be developed to target these membrane channels in various oral diseases and maxillofacial dysplasia.
Collapse
Affiliation(s)
- Wenjing Liu
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jieya Wei
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianxun Sun
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
24
|
Calhoun PJ, Phan AV, Taylor JD, James CC, Padget RL, Zeitz MJ, Smyth JW. Adenovirus targets transcriptional and posttranslational mechanisms to limit gap junction function. FASEB J 2020; 34:9694-9712. [PMID: 32485054 DOI: 10.1096/fj.202000667r] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 01/19/2023]
Abstract
Adenoviruses are responsible for a spectrum of pathogenesis including viral myocarditis. The gap junction protein connexin43 (Cx43, gene name GJA1) facilitates rapid propagation of action potentials necessary for each heartbeat. Gap junctions also propagate innate and adaptive antiviral immune responses, but how viruses may target these structures is not understood. Given this immunological role of Cx43, we hypothesized that gap junctions would be targeted during adenovirus type 5 (Ad5) infection. We find reduced Cx43 protein levels due to decreased GJA1 mRNA transcripts dependent upon β-catenin transcriptional activity during Ad5 infection, with early viral protein E4orf1 sufficient to induce β-catenin phosphorylation. Loss of gap junction function occurs prior to reduced Cx43 protein levels with Ad5 infection rapidly inducing Cx43 phosphorylation events consistent with altered gap junction conductance. Direct Cx43 interaction with ZO-1 plays a critical role in gap junction regulation. We find loss of Cx43/ZO-1 complexing during Ad5 infection by co-immunoprecipitation and complementary studies in human induced pluripotent stem cell derived-cardiomyocytes reveal Cx43 gap junction remodeling by reduced ZO-1 complexing. These findings reveal specific targeting of gap junction function by Ad5 leading to loss of intercellular communication which would contribute to dangerous pathological states including arrhythmias in infected hearts.
Collapse
Affiliation(s)
- Patrick J Calhoun
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Allen V Phan
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | | | - Carissa C James
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Rachel L Padget
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Michael J Zeitz
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
| | - James W Smyth
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| |
Collapse
|
25
|
Jourdeuil K, Taneyhill LA. The gap junction protein connexin 43 controls multiple aspects of cranial neural crest cell development. J Cell Sci 2020; 133:jcs235440. [PMID: 31964703 PMCID: PMC7044449 DOI: 10.1242/jcs.235440] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022] Open
Abstract
Gap junctions are intercellular channels between cells that facilitate cell-cell communication. Connexin 43 (Cx43; also known as GJA1), the predominant gap junction protein in vertebrates, is expressed in premigratory cranial neural crest cells and is maintained throughout the neural crest cell epithelial-to-mesenchymal transition (EMT), but its function in these cells is unknown. To this end, we used a combination of in vivo and ex vivo experiments to assess gap junction formation, and Cx43 function, in chick cranial neural crest cells. Our results demonstrate that gap junctions exist between premigratory and migratory cranial neural crest cells and depend on Cx43 for their function. In the embryo, Cx43 knockdown just prior to EMT delays the emergence of Cx43-depleted neural crest cells from the neural tube, but these cells eventually successfully emigrate and join the migratory stream. This delay can be rescued by introduction of full-length Cx43 into Cx43-depleted cells. Furthermore, Cx43 depletion reduces the size of the premigratory neural crest cell domain through an early effect on neural crest cell specification. Collectively, these data identify new roles for Cx43 in chick cranial neural crest cell development.
Collapse
Affiliation(s)
- Karyn Jourdeuil
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
26
|
Esseltine JL, Brooks CR, Edwards NA, Subasri M, Sampson J, Séguin C, Betts DH, Laird DW. Dynamic regulation of connexins in stem cell pluripotency. Stem Cells 2019; 38:52-66. [DOI: 10.1002/stem.3092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 07/18/2019] [Accepted: 08/08/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Jessica L. Esseltine
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry; University of Western Ontario; London Ontario Canada
- Division of BioMedical Sciences, Faculty of Medicine; Memorial University of Newfoundland; St. John's Newfoundland and Labrador Canada
| | - Courtney R. Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry; University of Western Ontario; London Ontario Canada
| | - Nicole A. Edwards
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry; University of Western Ontario; London Ontario Canada
| | - Mathushan Subasri
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry; University of Western Ontario; London Ontario Canada
| | - Jacinda Sampson
- Department of Neurology; Stanford University Medical Center; Palo Alto California
| | - Cheryle Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry; University of Western Ontario; London Ontario Canada
| | - Dean H. Betts
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry; University of Western Ontario; London Ontario Canada
| | - Dale W. Laird
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry; University of Western Ontario; London Ontario Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry; University of Western Ontario; London Ontario Canada
| |
Collapse
|
27
|
Promotion of growth factor signaling as a critical function of β-catenin during HCC progression. Nat Commun 2019; 10:1909. [PMID: 31015417 PMCID: PMC6478918 DOI: 10.1038/s41467-019-09780-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 03/29/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. β-catenin is widely thought to be a major oncogene in HCC based on the frequency of mutations associated with aberrant Wnt signaling in HCC patients. Challenging this model, our data reveal that β-catenin nuclear accumulation is restricted to the late stage of the disease. Until then, β-catenin is primarily located at the plasma membrane in complex with multiple cadherin family members where it drives tumor cell survival by enhancing the signaling of growth factor receptors such as EGFR. Therefore, our study reveals the evolving nature of β-catenin in HCC to establish it as a compound tumor promoter during the progression of the disease. Aberrant Wnt/b-catenin signaling is thought to be a major driver of hepatocellular carcinoma. Here, the authors show that β-Catenin is predominantly integrated within the AJ complex during the early stages of this cancer and enhance EGFR signaling to promote tumour survival.
Collapse
|
28
|
McClure MJ, Ramey AN, Rashid M, Boyan BD, Schwartz Z. Integrin-α7 signaling regulates connexin 43, M-cadherin, and myoblast fusion. Am J Physiol Cell Physiol 2019; 316:C876-C887. [PMID: 30892939 DOI: 10.1152/ajpcell.00282.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regenerative medicine treatments for severe skeletal muscle injuries are limited, resulting in persistent functional deficits. Clinical options include neglecting the wound with the expectation that fibrosis will develop or using an autologous muscle graft with minimal functional improvement. A regenerative matrix can be used, but muscle fiber development on these matrices remains a challenge in vivo. Here, we explored the fundamental mechanisms that mediate cell-substrate signaling and its effect on cell-cell communication during myoblast fusion and tube formation to improve outcomes following implantation of matrices used to stimulate muscle regeneration. We previously reported that integrin-α7 was increased on anisotropic biomaterials, suggesting a role for α7β1 signaling in myoblast communication via connexin 43 and M-cadherin. Our results demonstrated that α7 silencing blocked expression of myogenic differentiation factor 1 (Myod), myogenin (Myog), myogenic factor 6 (Myf6), myosin heavy chain type 1 (Myh1), and transmembrane protein 8c (Tmem8c), indicating that myoblast fusion was inhibited. Expression of α5 and M-cadherin decreased but β1 and connexin 43 increased. We examined protein production and observed reduced extracellular-signal regulated kinase 1/2 (ERK) in α7-silenced cells that correlated with upregulation of connexin 43 and M-cadherin, suggesting a compensatory pathway. These results indicate that α7 signaling plays a critical role in ex vivo fusion and implicates a relationship with connexin 43 and M-cadherin.
Collapse
Affiliation(s)
- Michael J McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia
| | - Allison N Ramey
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia
| | - Mashaba Rashid
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia
| | - Barbara D Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia.,Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University , Richmond, Virginia.,Department of Periodontics, University of Texas Health Sciences Center at San Antonio , San Antonio, Texas
| |
Collapse
|
29
|
Aftab Q, Mesnil M, Ojefua E, Poole A, Noordenbos J, Strale PO, Sitko C, Le C, Stoynov N, Foster LJ, Sin WC, Naus CC, Chen VC. Cx43-Associated Secretome and Interactome Reveal Synergistic Mechanisms for Glioma Migration and MMP3 Activation. Front Neurosci 2019; 13:143. [PMID: 30941001 PMCID: PMC6433981 DOI: 10.3389/fnins.2019.00143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/07/2019] [Indexed: 12/23/2022] Open
Abstract
Extracellular matrix (ECM) remodeling, degradation and glioma cell motility are critical aspects of glioblastoma multiforme (GBM). Despite being a rich source of potential biomarkers and targets for therapeutic advance, the dynamic changes occurring within the extracellular environment that are specific to GBM motility have yet to be fully resolved. The gap junction protein connexin43 (Cx43) increases glioma migration and invasion in a variety of in vitro and in vivo models. In this study, the upregulation of Cx43 in C6 glioma cells induced morphological changes and the secretion of proteins associated with cell motility. Demonstrating the selective engagement of ECM remodeling networks, secretome analysis revealed the near-binary increase of osteopontin and matrix metalloproteinase-3 (MMP3), with gelatinase and NFF-3 assays confirming the proteolytic activities. Informatic analysis of interactome and secretome downstream of Cx43 identifies networks of glioma motility that appear to be synergistically engaged. The data presented here implicate ECM remodeling and matrikine signals downstream of Cx43/MMP3/osteopontin and ARK1B10 inhibition as possible avenues to inhibit GBM.
Collapse
Affiliation(s)
- Qurratulain Aftab
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Marc Mesnil
- Signalisation et Transports Ioniques Membranaires (STIM), CNRS ERL 7003, University of Poitiers, Poitiers, France
| | - Emmanuel Ojefua
- Department of Chemistry, Brandon University, Brandon, MB, Canada
| | - Alisha Poole
- Department of Chemistry, Brandon University, Brandon, MB, Canada
| | - Jenna Noordenbos
- Department of Chemistry, Brandon University, Brandon, MB, Canada
| | - Pierre-Olivier Strale
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Chris Sitko
- Department of Chemistry, Brandon University, Brandon, MB, Canada
| | - Caitlin Le
- Department of Chemistry, Brandon University, Brandon, MB, Canada
| | - Nikolay Stoynov
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada
| | - Wun-Chey Sin
- Signalisation et Transports Ioniques Membranaires (STIM), CNRS ERL 7003, University of Poitiers, Poitiers, France
| | - Christian C Naus
- Signalisation et Transports Ioniques Membranaires (STIM), CNRS ERL 7003, University of Poitiers, Poitiers, France
| | - Vincent C Chen
- Department of Chemistry, Brandon University, Brandon, MB, Canada
| |
Collapse
|
30
|
Cell-cell contacts protect against t-BuOOH-induced cellular damage and ferroptosis in vitro. Arch Toxicol 2019; 93:1265-1279. [PMID: 30798349 DOI: 10.1007/s00204-019-02413-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a recently discovered pathway of regulated necrosis dependent on iron and lipid peroxidation. It has gained broad attention since it is a promising approach to overcome resistance to apoptosis in cancer chemotherapy. We have recently identified tertiary-butyl hydroperoxide (t-BuOOH) as a novel inducer of ferroptosis. t-BuOOH is a widely used compound to induce oxidative stress in vitro. t-BuOOH induces lipid peroxidation and consequently ferroptosis in murine and human cell lines. t-BuOOH additionally results in a loss of mitochondrial membrane potential, formation of DNA double-strand breaks, and replication block. Here, we specifically address the question whether cell-cell contacts regulate t-BuOOH-induced ferroptosis and cellular damage. To this end, murine NIH3T3 or human HaCaT cells were seeded to confluence, but below their saturation density to allow the establishment of cell-cell contacts without inducing quiescence. Cells were then treated with t-BuOOH (50 or 200 µM, respectively). We revealed that cell-cell contacts reduce basal and t-BuOOH-triggered lipid peroxidation and consequently block ferroptosis. Similar results were obtained with the specific ferroptosis inducer erastin. Cell-cell contacts further protect against t-BuOOH-induced loss of mitochondrial membrane potential, and formation of DNA double-strand breaks. Interestingly, cell-cell contacts failed to prevent t-BuOOH-mediated replication block or formation of the oxidative base lesion 8-oxo-dG. Since evidence of protection against cell death was both (i) observed after treatment with hydrogen peroxide, methyl methanesulfonate or UV-C, and (ii) seen in several cell lines, we conclude that protection by cell-cell contacts is a widespread phenomenon. The impact of cell-cell contacts on toxicity might have important implications in cancer chemotherapy.
Collapse
|
31
|
The Functional Implications of Endothelial Gap Junctions and Cellular Mechanics in Vascular Angiogenesis. Cancers (Basel) 2019; 11:cancers11020237. [PMID: 30781714 PMCID: PMC6406946 DOI: 10.3390/cancers11020237] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis—the sprouting and growth of new blood vessels from the existing vasculature—is an important contributor to tumor development, since it facilitates the supply of oxygen and nutrients to cancer cells. Endothelial cells are critically affected during the angiogenic process as their proliferation, motility, and morphology are modulated by pro-angiogenic and environmental factors associated with tumor tissues and cancer cells. Recent in vivo and in vitro studies have revealed that the gap junctions of endothelial cells also participate in the promotion of angiogenesis. Pro-angiogenic factors modulate gap junction function and connexin expression in endothelial cells, whereas endothelial connexins are involved in angiogenic tube formation and in the cell migration of endothelial cells. Several mechanisms, including gap junction function-dependent or -independent pathways, have been proposed. In particular, connexins might have the potential to regulate cell mechanics such as cell morphology, cell migration, and cellular stiffness that are dynamically changed during the angiogenic processes. Here, we review the implication for endothelial gap junctions and cellular mechanics in vascular angiogenesis.
Collapse
|
32
|
Pogoda K, Kameritsch P, Mannell H, Pohl U. Connexins in the control of vasomotor function. Acta Physiol (Oxf) 2019; 225:e13108. [PMID: 29858558 DOI: 10.1111/apha.13108] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 12/13/2022]
Abstract
Vascular endothelial cells, as well as smooth muscle cells, show heterogeneity with regard to their receptor expression and reactivity. For the vascular wall to act as a functional unit, the various cells' responses require integration. Such an integration is not only required for a homogeneous response of the vascular wall, but also for the vasomotor behaviour of consecutive segments of the microvascular arteriolar tree. As flow resistances of individual sections are connected in series, sections require synchronization and coordination to allow effective changes of conductivity and blood flow. A prerequisite for the local coordination of individual vascular cells and different sections of an arteriolar tree is intercellular communication. Connexins are involved in a dual manner in this coordination. (i) By forming gap junctions between cells, they allow an intercellular exchange of signalling molecules and electrical currents. In particular, the spread of electrical currents allows for coordination of cell responses over longer distances. (ii) Connexins are able to interact with other proteins to form signalling complexes. In this way, they can modulate and integrate individual cells' responses also in a channel-independent manner. This review outlines mechanisms allowing the vascular connexins to exert their coordinating function and to regulate the vasomotor reactions of blood vessels both locally, and in vascular networks. Wherever possible, we focus on the vasomotor behaviour of small vessels and arterioles which are the main vessels determining vascular resistance, blood pressure and local blood flow.
Collapse
Affiliation(s)
- K. Pogoda
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
| | - P. Kameritsch
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
| | - H. Mannell
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
| | - U. Pohl
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
- Munich Cluster for Systems Neurology (SyNergy); Munich Germany
| |
Collapse
|
33
|
Rasouli SJ, El-Brolosy M, Tsedeke AT, Bensimon-Brito A, Ghanbari P, Maischein HM, Kuenne C, Stainier DY. The flow responsive transcription factor Klf2 is required for myocardial wall integrity by modulating Fgf signaling. eLife 2018; 7:e38889. [PMID: 30592462 PMCID: PMC6329608 DOI: 10.7554/elife.38889] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 12/24/2018] [Indexed: 12/17/2022] Open
Abstract
Complex interplay between cardiac tissues is crucial for their integrity. The flow responsive transcription factor KLF2, which is expressed in the endocardium, is vital for cardiovascular development but its exact role remains to be defined. To this end, we mutated both klf2 paralogues in zebrafish, and while single mutants exhibit no obvious phenotype, double mutants display a novel phenotype of cardiomyocyte extrusion towards the abluminal side. This extrusion requires cardiac contractility and correlates with the mislocalization of N-cadherin from the lateral to the apical side of cardiomyocytes. Transgenic rescue data show that klf2 expression in endothelium, but not myocardium, prevents this cardiomyocyte extrusion phenotype. Transcriptome analysis of klf2 mutant hearts reveals that Fgf signaling is affected, and accordingly, we find that inhibition of Fgf signaling in wild-type animals can lead to abluminal cardiomyocyte extrusion. These studies provide new insights into how Klf2 regulates cardiovascular development and specifically myocardial wall integrity.
Collapse
Affiliation(s)
- Seyed Javad Rasouli
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Mohamed El-Brolosy
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Ayele Taddese Tsedeke
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Anabela Bensimon-Brito
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Parisa Ghanbari
- Department of Cardiac Development and RemodelingMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Hans-Martin Maischein
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Carsten Kuenne
- Bioinformatics Core UnitMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Didier Y Stainier
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| |
Collapse
|
34
|
Gap junction protein Connexin-43 is a direct transcriptional regulator of N-cadherin in vivo. Nat Commun 2018; 9:3846. [PMID: 30242148 PMCID: PMC6155008 DOI: 10.1038/s41467-018-06368-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/29/2018] [Indexed: 11/16/2022] Open
Abstract
Connexins are the primary components of gap junctions, providing direct links between cells under many physiological processes. Here, we demonstrate that in addition to this canonical role, Connexins act as transcriptional regulators. We show that Connexin 43 (Cx43) controls neural crest cell migration in vivo by directly regulating N-cadherin transcription. This activity requires interaction between Cx43 carboxy tail and the basic transcription factor-3, which drives the translocation of Cx43 tail to the nucleus. Once in the nucleus they form a complex with PolII which directly binds to the N-cadherin promoter. We found that this mechanism is conserved between amphibian and mammalian cells. Given the strong evolutionary conservation of connexins across vertebrates, this may reflect a common mechanism of gene regulation by a protein whose function was previously ascribed only to gap junctional communication. Connexins are components of gap junctions that link cells and allow intercellular communication. Here, the authors show that the Connexin 43 carboxy tail interacts with basic transcription factor-3, leading to nuclear translocation and direct regulation of N-cadherin expression and neural crest migration.
Collapse
|
35
|
Expression and function of connexin 43 protein in mouse and human retinal pigment epithelial cells as hemichannels and gap junction proteins. Exp Eye Res 2018; 168:128-137. [DOI: 10.1016/j.exer.2018.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 01/08/2023]
|
36
|
Basheer WA, Xiao S, Epifantseva I, Fu Y, Kleber AG, Hong T, Shaw RM. GJA1-20k Arranges Actin to Guide Cx43 Delivery to Cardiac Intercalated Discs. Circ Res 2017; 121:1069-1080. [PMID: 28923791 DOI: 10.1161/circresaha.117.311955] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 01/21/2023]
Abstract
RATIONALE Delivery of Cx43 (connexin 43) to the intercalated disc is a continuous and rapid process critical for intercellular coupling. By a pathway of targeted delivery involving microtubule highways, vesicles of Cx43 hemichannels are efficiently trafficked to adherens junctions at intercalated discs. It has also been identified that actin provides rest stops for Cx43 forward trafficking and that Cx43 has a 20 kDa internally translated small C terminus isoform, GJA1-20k (Gap Junction Protein Alpha 1- 20 kDa), which is required for full-length Cx43 trafficking, but by an unknown mechanism. OBJECTIVE We explored the mechanism by which the GJA1-20k isoform is required for full-length Cx43 forward trafficking to intercalated discs. METHODS AND RESULTS Using an in vivo Adeno-associated virus serotype 9-mediated gene transfer system, we confirmed in whole animal that GJA1-20k markedly increases endogenous myocardial Cx43 gap junction plaque size at the intercalated discs. In micropatterned cell pairing systems, we found that exogenous GJA1-20k expression stabilizes filamentous actin without affecting actin protein expression and that GJA1-20k complexes with both actin and tubulin. We also found that filamentous actin regulates microtubule organization as inhibition of actin polymerization with a low dose of latrunculin A disrupts the targeting of microtubules to cell-cell junctions. GJA1-20k protects actin filament from latrunculin A disruption, preserving microtubule trajectory to the cell-cell border. For therapeutic implications, we found that prior in vivo Adeno-associated virus serotype 9-mediated gene delivery of GJA1-20k to the heart protects Cx43 localization to the intercalated discs against acute ischemic injury. CONCLUSIONS The internally translated GJA1-20k isoform stabilizes actin filaments, which guides growth trajectories of the Cx43 microtubule trafficking machinery, increasing delivery of Cx43 hemichannels to cardiac intercalated discs. Exogenous GJA1-20k helps to maintain cell-cell coupling in instances of anticipated myocardial ischemia.
Collapse
Affiliation(s)
- Wassim A Basheer
- From the Cedars-Sinai Heart Institute (W.A.B., S.X., I.E., Y.F., T.H., R.M.S.) and Department of Medicine (T.H., R.M.S.), Cedars-Sinai Medical Center and UCLA, Los Angeles, CA; and Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (A.G.K.)
| | - Shaohua Xiao
- From the Cedars-Sinai Heart Institute (W.A.B., S.X., I.E., Y.F., T.H., R.M.S.) and Department of Medicine (T.H., R.M.S.), Cedars-Sinai Medical Center and UCLA, Los Angeles, CA; and Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (A.G.K.)
| | - Irina Epifantseva
- From the Cedars-Sinai Heart Institute (W.A.B., S.X., I.E., Y.F., T.H., R.M.S.) and Department of Medicine (T.H., R.M.S.), Cedars-Sinai Medical Center and UCLA, Los Angeles, CA; and Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (A.G.K.)
| | - Ying Fu
- From the Cedars-Sinai Heart Institute (W.A.B., S.X., I.E., Y.F., T.H., R.M.S.) and Department of Medicine (T.H., R.M.S.), Cedars-Sinai Medical Center and UCLA, Los Angeles, CA; and Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (A.G.K.)
| | - Andre G Kleber
- From the Cedars-Sinai Heart Institute (W.A.B., S.X., I.E., Y.F., T.H., R.M.S.) and Department of Medicine (T.H., R.M.S.), Cedars-Sinai Medical Center and UCLA, Los Angeles, CA; and Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (A.G.K.)
| | - TingTing Hong
- From the Cedars-Sinai Heart Institute (W.A.B., S.X., I.E., Y.F., T.H., R.M.S.) and Department of Medicine (T.H., R.M.S.), Cedars-Sinai Medical Center and UCLA, Los Angeles, CA; and Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (A.G.K.)
| | - Robin M Shaw
- From the Cedars-Sinai Heart Institute (W.A.B., S.X., I.E., Y.F., T.H., R.M.S.) and Department of Medicine (T.H., R.M.S.), Cedars-Sinai Medical Center and UCLA, Los Angeles, CA; and Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (A.G.K.).
| |
Collapse
|
37
|
Montague K, Lowe AS, Uzquiano A, Knüfer A, Astick M, Price SR, Guthrie S. The assembly of developing motor neurons depends on an interplay between spontaneous activity, type II cadherins and gap junctions. Development 2017; 144:830-836. [PMID: 28246212 DOI: 10.1242/dev.144063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/10/2017] [Indexed: 01/12/2023]
Abstract
A core structural and functional motif of the vertebrate central nervous system is discrete clusters of neurons or 'nuclei'. Yet the developmental mechanisms underlying this fundamental mode of organisation are largely unknown. We have previously shown that the assembly of motor neurons into nuclei depends on cadherin-mediated adhesion. Here, we demonstrate that the emergence of mature topography among motor nuclei involves a novel interplay between spontaneous activity, cadherin expression and gap junction communication. We report that nuclei display spontaneous calcium transients, and that changes in the activity patterns coincide with the course of nucleogenesis. We also find that these activity patterns are disrupted by manipulating cadherin or gap junction expression. Furthermore, inhibition of activity disrupts nucleogenesis, suggesting that activity feeds back to maintain integrity among motor neurons within a nucleus. Our study suggests that a network of interactions between cadherins, gap junctions and spontaneous activity governs neuron assembly, presaging circuit formation.
Collapse
Affiliation(s)
- Karli Montague
- Wolfson Centre for Age-related Diseases, King's College London, Guy's Hospital Campus, London SE1 1UL, UK
| | - Andrew S Lowe
- Department of Developmental Neurobiology, King's College London, Guy's Hospital Campus, London SE1 1UL, UK
| | - Ana Uzquiano
- École de Neuroscience-Paris Île-de-France, ENP-DIM, 15 Rue de L'École de Médécine, Paris 75006, France
| | - Athene Knüfer
- Department of Developmental Neurobiology, King's College London, Guy's Hospital Campus, London SE1 1UL, UK
| | - Marc Astick
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université de Bruxelles, Route de Lennik 808, Bruxelles B1070, Belgium
| | - Stephen R Price
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Sarah Guthrie
- Department of Developmental Neurobiology, King's College London, Guy's Hospital Campus, London SE1 1UL, UK
| |
Collapse
|
38
|
Boucher J, Monvoisin A, Vix J, Mesnil M, Thuringer D, Debiais F, Cronier L. Connexins, important players in the dissemination of prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:202-215. [PMID: 28693897 DOI: 10.1016/j.bbamem.2017.06.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/22/2017] [Accepted: 06/29/2017] [Indexed: 12/25/2022]
Abstract
Over the past 50years, increasing experimental evidences have established that connexins (Cxs) and gap junctional intercellular communication (GJIC) ensure an important role in both the onset and development of cancerous processes. In the present review, we focus on the impact of Cxs and GJIC during the development of prostate cancer (PCa), from the primary growth mainly localized in acinar glands and ducts to the distant metastasis mainly concentrated in bone. As observed in several other types of solid tumours, Cxs and especially Cx43 exhibit an ambivalent role with a tumour suppressor effect in the early stages and, conversely, a rather pro-tumoural profile for most of invasion and dissemination steps to secondary sites. We report here the current knowledge on the function of Cxs during PCa cells migration, cytoskeletal dynamics, proteinases activities and the cross talk with the surrounding stromal cells in the microenvironment of the tumour and the bones. In addition, we discuss the role of Cxs in the bone tropism even if the prostate model is rarely used to study the complete sequence of cancer dissemination compared to breast cancer or melanoma. Even if not yet fully understood, these recent findings on Cxs provide new insights into their molecular mechanisms associated with progression and bone targeted behaviour of PCa. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Jonathan Boucher
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | - Arnaud Monvoisin
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | - Justine Vix
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France; Department of Rheumatology, C.H.U. la Milétrie, Poitiers, France
| | - Marc Mesnil
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | | | - Françoise Debiais
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France; Department of Rheumatology, C.H.U. la Milétrie, Poitiers, France
| | - Laurent Cronier
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France.
| |
Collapse
|
39
|
Lembong J, Sabass B, Stone HA. Calcium oscillations in wounded fibroblast monolayers are spatially regulated through substrate mechanics. Phys Biol 2017; 14:045006. [PMID: 28378710 DOI: 10.1088/1478-3975/aa6b67] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The maintenance of tissue integrity is essential for the life of multicellular organisms. Healing of a skin wound is a paradigm for how various cell types localize and repair tissue perturbations in an orchestrated fashion. To investigate biophysical mechanisms associated with wound localization, we focus on a model system consisting of a fibroblast monolayer on an elastic substrate. We find that the creation of an edge in the monolayer causes cytosolic calcium oscillations throughout the monolayer. The oscillation frequency increases with cell density, which shows that wound-induced calcium oscillations occur collectively. Inhibition of myosin II reduces the number of oscillating cells, demonstrating a coupling between actomyosin activity and calcium response. The spatial distribution of oscillating cells depends on the stiffness of the substrate. For soft substrates with a Young's modulus E ~ 360 Pa, oscillations occur on average within 0.2 mm distance from the wound edge. Increasing substrate stiffness leads to an average localization of oscillations away from the edge (up to ~0.6 mm). In addition, we use traction force microscopy to determine stresses between cells and substrate. We find that an increase of substrate rigidity leads to a higher traction magnitude. For E < ~2 kPa, the traction magnitude is strongly concentrated at the monolayer edge, while for E > ~8 kPa, traction magnitude is on average almost uniform beneath the monolayer. Thus, the spatial occurrence of calcium oscillations correlates with the cell-substrate traction. Overall, the experiments with fibroblasts demonstrate a collective, chemomechanical localization mechanism at the edge of a wound with a potential physiological role.
Collapse
Affiliation(s)
- Josephine Lembong
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, United States of America. Current address: Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America. These authors contributed equally to this work
| | | | | |
Collapse
|
40
|
Epifantseva I, Shaw RM. Intracellular trafficking pathways of Cx43 gap junction channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:40-47. [PMID: 28576298 DOI: 10.1016/j.bbamem.2017.05.018] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/19/2017] [Accepted: 05/25/2017] [Indexed: 12/11/2022]
Abstract
Gap Junction (GJ) channels, including the most common Connexin 43 (Cx43), have fundamental roles in excitable tissues by facilitating rapid transmission of action potentials between adjacent cells. For instance, synchronization during each heartbeat is regulated by these ion channels at the cardiomyocyte cell-cell border. Cx43 protein has a short half-life, and rapid synthesis and timely delivery of those proteins to particular subdomains are crucial for the cellular organization of gap junctions and maintenance of intracellular coupling. Impairment in gap junction trafficking contributes to dangerous complications in diseased hearts such as the arrhythmias of sudden cardiac death. Of recent interest are the protein-protein interactions with the Cx43 carboxy-terminus. These interactions have significant impact on the full length Cx43 lifecycle and also contribute to trafficking of Cx43 as well as possibly other functions. We are learning that many of the known non-canonical roles of Cx43 can be attributed to the recently identified six endogenous Cx43 truncated isoforms which are produced by internal translation. In general, alternative translation is a new leading edge for proteome expansion and therapeutic drug development. This review highlights recent mechanisms identified in the trafficking of gap junction channels, involvement of other proteins contributing to the delivery of channels to the cell-cell border, and understanding of possible roles of the newly discovered alternatively translated isoforms in Cx43 biology. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Irina Epifantseva
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Robin M Shaw
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.; Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA..
| |
Collapse
|
41
|
Zheng B, Wang J, Tang L, Tan C, Zhao Z, Xiao Y, Ge R, Zhu D. Involvement of Rictor/mTORC2 in cardiomyocyte differentiation of mouse embryonic stem cells in vitro. Int J Biol Sci 2017; 13:110-121. [PMID: 28123351 PMCID: PMC5264266 DOI: 10.7150/ijbs.16312] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 10/21/2016] [Indexed: 11/05/2022] Open
Abstract
Rictor is a key regulatory/structural subunit of the mammalian target of rapamycin complex 2 (mTORC2) and is required for phosphorylation of Akt at serine 473. It plays an important role in cell survival, actin cytoskeleton organization and other processes in embryogenesis. However, the role of Rictor/mTORC2 in the embryonic cardiac differentiation has been uncovered. In the present study, we examined a possible link between Rictor expression and cardiomyocyte differentiation of the mouse embryonic stem (mES) cells. Knockdown of Rictor by shRNA significantly reduced the phosphorylation of Akt at serine 473 followed by a decrease in cardiomyocyte differentiation detected by beating embryoid bodies. The protein levels of brachyury (mesoderm protein), Nkx2.5 (cardiac progenitor cell protein) and α-Actinin (cardiomyocyte biomarker) decreased in Rictor knockdown group during cardiogenesis. Furthermore, knockdown of Rictor specifically inhibited the ventricular-like cells differentiation of mES cells with reduced level of ventricular-specific protein, MLC-2v. Meanwhile, patch-clamp analysis revealed that shRNA-Rictor significantly increased the number of cardiomyocytes with abnormal electrophysiology. In addition, the expressions and distribution patterns of cell-cell junction proteins (Cx43/Desmoplakin/N-cadherin) were also affected in shRNA-Rictor cardiomyocytes. Taken together, the results demonstrated that Rictor/mTORC2 might play an important role in the cardiomyocyte differentiation of mES cells. Knockdown of Rictor resulted in inhibiting ventricular-like myocytes differentiation and induced arrhythmias symptom, which was accompanied by interfering the expression and distribution patterns of cell-cell junction proteins. Rictor/mTORC2 might become a new target for regulating cardiomyocyte differentiation and a useful reference for application of the induced pluripotent stem cells.
Collapse
Affiliation(s)
- Bei Zheng
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| | - Jiadan Wang
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| | - Leilei Tang
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| | - Chao Tan
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| | - Zhe Zhao
- Undergraduate students in Research Training Project at Zhejiang University
| | - Yi Xiao
- Undergraduate students in Research Training Project at Zhejiang University
| | - Renshan Ge
- The Population Council at the Rockefeller University, New York, NY 10021, USA.; Institute of Reproductive Biomedicine, the 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, CHINA
| | - Danyan Zhu
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou 310058, CHINA
| |
Collapse
|
42
|
Santolim LV, Amaral MECD, Fachi JL, Mendes MF, Oliveira CAD. Vitamin E and caloric restriction promote hepatic homeostasis through expression of connexin 26, N-cad, E-cad and cholesterol metabolism genes. J Nutr Biochem 2017; 39:86-92. [PMID: 27816814 DOI: 10.1016/j.jnutbio.2016.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 09/09/2016] [Accepted: 09/13/2016] [Indexed: 12/30/2022]
Abstract
Connexins (Cx) and cadherins are responsible for cell homeostasis. The Cx activity is directly related to cholesterol. The present work investigates whether vitamin E, with or without caloric restriction (CR), alters the mRNA expression of Cx26, Cx32, Cx43, N-cadherins (N-cads), E-cadherins (E-cads) and alpha-smooth muscle actin (α-SMA), and evaluates their relation to cholesterol metabolism in rat liver. Animals were divided into different groups: control with ad libitum diet (C), control+vitamin E (CV), aloric restriction with intake to 60% of group C (CR), and the intake of group CR+vitamin E (RV). There were increases of manganese superoxide dismutase (Mn-SOD) and glutathione S-transferase mu 1, indicating antioxidant effects of CR and vitamin E. An increase of nitric oxide in the CR group was in agreement with the Mn-SOD data. Supplementation with vitamin E, with or without CR, upregulated the expression of Cx26 mRNA and increased low-density lipoprotein cholesterol (LDL-c) in the CV group. Reductions of Cx32 and Cx43 were associated with lower LDL-c. Increases in Hmgcr and low-density lipoprotein receptor (LDLr) in the CV and RV groups could be explained by the effect of vitamin E. A reduction of LDLr in the CR group was due to the reduced dietary intake. Increases in cadherins in the CV, CR and RV groups were indicative of tissue maintenance, which was also supported by increases of α-SMA in groups CV and RV. Finally, vitamin E, with or without CR, increased Cx26, probably modulated by expression of the Hmgcr and LDLr genes. This suggests important relationship of Cxs and cholesterol metabolism genes.
Collapse
Affiliation(s)
- Leonardo Vinícius Santolim
- Graduate Program in Biomedical Sciences, Centro Universitário Hermínio Ometto, UNIARARAS, Araras, SP 13607339, Brazil
| | | | - José Luís Fachi
- School of Biomedicine, Centro Universitário Hermínio Ometto, UNIARARAS, Araras, SP 13607339, Brazil
| | - Maíra Felonato Mendes
- Graduate Program in Biomedical Sciences, Centro Universitário Hermínio Ometto, UNIARARAS, Araras, SP 13607339, Brazil
| | - Camila Andréa de Oliveira
- Graduate Program in Biomedical Sciences, Centro Universitário Hermínio Ometto, UNIARARAS, Araras, SP 13607339, Brazil.
| |
Collapse
|
43
|
Soon ASC, Chua JW, Becker DL. Connexins in endothelial barrier function - novel therapeutic targets countering vascular hyperpermeability. Thromb Haemost 2016; 116:852-867. [PMID: 27488046 DOI: 10.1160/th16-03-0210] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 07/15/2016] [Indexed: 12/14/2022]
Abstract
Prolonged vascular hyperpermeability is a common feature of many diseases. Vascular hyperpermeability is typically associated with changes in the expression patterns of adherens and tight junction proteins. Here, we focus on the less-appreciated contribution of gap junction proteins (connexins) to basal vascular permeability and endothelial dysfunction. First, we assess the association of connexins with endothelial barrier integrity by introducing tools used in connexin biology and relating the findings to customary readouts in vascular biology. Second, we explore potential mechanistic ties between connexins and junction regulation. Third, we review the role of connexins in microvascular organisation and development, focusing on interactions of the endothelium with mural cells and tissue-specific perivascular cells. Last, we see how connexins contribute to the interactions between the endothelium and components of the immune system, by using neutrophils as an example. Mounting evidence of crosstalk between connexins and other junction proteins suggests that we rethink the way in which different junction components contribute to endothelial barrier function. Given the multiple points of connexin-mediated communication arising from the endothelium, there is great potential for synergism between connexin-targeted inhibitors and existing immune-targeted therapeutics. As more drugs targeting connexins progress through clinical trials, it is hoped that some might prove effective at countering vascular hyperpermeability.
Collapse
Affiliation(s)
| | | | - David Laurence Becker
- David L. Becker, PhD, Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, 308232 Singapore, Tel: +65 6592 3961, Fax: +65 6515 0417, E-mail:
| |
Collapse
|
44
|
Connexins, E-cadherin, Claudin-7 and β-catenin transiently form junctional nexuses during the post-natal mammary gland development. Dev Biol 2016; 416:52-68. [PMID: 27291930 DOI: 10.1016/j.ydbio.2016.06.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 05/15/2016] [Accepted: 06/03/2016] [Indexed: 12/21/2022]
Abstract
Gap junctions are intercellular channels made of connexins (Cxs) that allow direct communication between adjacent cells. Modulation of Cxs has been associated with abnormal development and function of the mammary gland and breast cancer. However, the mechanisms underlying their expression during normal mammary gland are not yet known. Cxs interact with components of tight and adherens junctions. Thus, we hypothesized that the expression levels of Cxs vary during mammary gland development and are regulated through stage-dependent interactions with members of the tight and adherens junctions. Our specific objectives were to: 1) determine the expression of Cxs and tight and adherens junction proteins throughout development and 2) characterize Cxs interactions with components of tight and adherens junctions. Murine mammary glands were sampled at various developmental stages (pre-pubescent to post-weaning). RT-qPCR and western-blot analyses demonstrated differential expression patterns for all gap (Cx43, Cx32, Cx26, Cx30), tight (Claudin-1, -3, -4, -7) and adherens (β-catenin, E- and P-cadherins) junctions throughout development. Interestingly, co-immunoprecipitation demonstrated interactions between these different types of junctions. Cx30 interacted with Cx26 just at the late pregnancy stage. While Cx43 showed a persistent interaction with β-catenin from virginity to post-weaning, its interactions with E-cadherin and Claudin-7 were transient. Cx32 interacted with Cx26, E-cadherin and β-catenin during lactation. Immunofluorescence results confirmed the existence of a junctional nexus that remodeled during mammary gland development. Together, our results confirm that the expression levels of Cxs vary concomitantly and that Cxs form junctional nexuses with tight and adherens junctions, suggesting the existence of common regulatory pathways.
Collapse
|
45
|
Altomare L, Cochis A, Carletta A, Rimondini L, Farè S. Thermo-responsive methylcellulose hydrogels as temporary substrate for cell sheet biofabrication. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2016; 27:95. [PMID: 26984360 DOI: 10.1007/s10856-016-5703-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 03/05/2016] [Indexed: 06/05/2023]
Abstract
Methylcellulose (MC), a water-soluble polymer derived from cellulose, was investigated as a possible temporary substrate having thermo-responsive properties favorable for cell culturing. MC-based hydrogels were prepared by a dispersion technique, mixing MC powder (2, 4, 6, 8, 10, 12 % w/v) with selected salts (sodium sulphate, Na2SO4), sodium phosphate, calcium chloride, or phosphate buffered saline, to evaluate the influence of different compositions on the thermo-responsive behavior. The inversion test was used to determine the gelation temperatures of the different hydrogel compositions; thermo-mechanical properties and thermo-reversibility of the MC hydrogels were investigated by rheological analysis. Gelation temperatures and rheological behavior depended on the MC concentration and type and concentration of salt used in hydrogel preparation. In vitro cytotoxicity tests, performed using L929 mouse fibroblasts, showed no toxic release from all the tested hydrogels. Among the investigated compositions, the hydrogel composed of 8 % w/v MC with 0.05 M Na2SO4 had a thermo-reversibility temperature at 37 °C. For that reason, this formulation was thus considered to verify the possibility of inducing in vitro spontaneous detachment of cells previously seeded on the hydrogel surface. A continuous cell layer (cell sheet) was allowed to grow and then detached from the hydrogel surface without the use of enzymes, thanks to the thermo-responsive behavior of the MC hydrogel. Immunofluorescence observation confirmed that the detached cell sheet was composed of closely interacting cells.
Collapse
Affiliation(s)
- Lina Altomare
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "G. Natta", Politecnico di Milano, Piazza L. Da Vinci 32, Milan, MI, Italy
- INSTM, Consorzio Nazionale di Scienza e Tecnologia dei Materiali, Local Unit Politecnico di Milano, Milan, Italy
| | - Andrea Cochis
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100, Novara, NO, Italy
- INSTM, Consorzio Nazionale di Scienza e Tecnologia dei Materiali, Local Unit Università del Piemonte Orientale, Novara, Italy
| | - Andrea Carletta
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "G. Natta", Politecnico di Milano, Piazza L. Da Vinci 32, Milan, MI, Italy
| | - Lia Rimondini
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100, Novara, NO, Italy.
- INSTM, Consorzio Nazionale di Scienza e Tecnologia dei Materiali, Local Unit Università del Piemonte Orientale, Novara, Italy.
| | - Silvia Farè
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "G. Natta", Politecnico di Milano, Piazza L. Da Vinci 32, Milan, MI, Italy
- INSTM, Consorzio Nazionale di Scienza e Tecnologia dei Materiali, Local Unit Politecnico di Milano, Milan, Italy
| |
Collapse
|
46
|
Connexin 43 expression is associated with increased malignancy in prostate cancer cell lines and functions to promote migration. Oncotarget 2016; 6:11640-51. [PMID: 25960544 PMCID: PMC4484482 DOI: 10.18632/oncotarget.3449] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/24/2015] [Indexed: 12/13/2022] Open
Abstract
Impaired expression of connexins, the gap junction subunits that facilitate direct cell-cell communication, have been implicated in prostate cancer growth. To elucidate the crucial role of connexins in prostate cancer progression, we performed a systematic quantitative RT-PCR screening of connexin expression in four representative prostate cancer cell lines across the spectrum of malignancy. Transcripts of several connexin subunits were detected in all four cell lines, and connexin 43 (Cx43) showed marked elevation at both RNA and protein levels in cells with increased metastatic potential. Analysis of gap-junction-mediated intercellular communication revealed homocellular coupling in PC-3 cells, which had the highest Cx43 expression, with minimal coupling in LNCaP cells where Cx43 expression was very low. Treatment with the gap junction inhibitor carbenoxolone or connexin mimetic peptide ACT-1 did not impair cell growth, suggesting that growth is independent of functional gap junctions. PC-3 cells with Cx43 expression reduced by shRNA showed decreased migration in monolayer wound healing assay, as well as decreased transwell invasion capacities when compared to control cells expressing non-targeting shRNA. These results, together with the correlation between Cx43 expression levels and the metastatic capacity of the cell lines, suggest a role of Cx43 in prostate cancer invasion and metastasis.
Collapse
|
47
|
Good ME, Begandt D, DeLalio LJ, Johnstone SR, Isakson BE. Small Interfering RNA-Mediated Connexin Gene Knockdown in Vascular Endothelial and Smooth Muscle Cells. Methods Mol Biol 2016; 1437:71-82. [PMID: 27207287 DOI: 10.1007/978-1-4939-3664-9_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Global knockout of vascular connexins can result in premature/neonatal death, severe developmental complications, or compensatory up-regulation of different connexin isoforms. Thus, specific connexin gene knockdown using RNAi-mediated technologies is a technique that allows investigators to efficiently monitor silencing effects of single or multiple connexin gene products. The present chapter describes the transient knockdown of connexins in vitro and ex vivo for cells of the blood vessel wall. In detail, different transfection methods for primary endothelial cells and ex vivo thoracodorsal arteries are described. Essential controls for validating transfection efficiency as well as targeted gene knockdown are explained. These protocols provide researchers with the ability to modify connexin gene expression levels in a multitude of experimental setups.
Collapse
Affiliation(s)
- Miranda E Good
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Daniela Begandt
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Leon J DeLalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.,Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Scott R Johnstone
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA. .,Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| |
Collapse
|
48
|
Nahta R, Al-Mulla F, Al-Temaimi R, Amedei A, Andrade-Vieira R, Bay SN, Brown DG, Calaf GM, Castellino RC, Cohen-Solal KA, Colacci A, Cruickshanks N, Dent P, Di Fiore R, Forte S, Goldberg GS, Hamid RA, Krishnan H, Laird DW, Lasfar A, Marignani PA, Memeo L, Mondello C, Naus CC, Ponce-Cusi R, Raju J, Roy D, Roy R, Ryan EP, Salem HK, Scovassi AI, Singh N, Vaccari M, Vento R, Vondráček J, Wade M, Woodrick J, Bisson WH. Mechanisms of environmental chemicals that enable the cancer hallmark of evasion of growth suppression. Carcinogenesis 2015; 36 Suppl 1:S2-18. [PMID: 26106139 DOI: 10.1093/carcin/bgv028] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
As part of the Halifax Project, this review brings attention to the potential effects of environmental chemicals on important molecular and cellular regulators of the cancer hallmark of evading growth suppression. Specifically, we review the mechanisms by which cancer cells escape the growth-inhibitory signals of p53, retinoblastoma protein, transforming growth factor-beta, gap junctions and contact inhibition. We discuss the effects of selected environmental chemicals on these mechanisms of growth inhibition and cross-reference the effects of these chemicals in other classical cancer hallmarks.
Collapse
Affiliation(s)
- Rita Nahta
- Departments of Pharmacology and Hematology & Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA 30322, USA, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy, Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada, Program in Genetics and Molecular Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA, Department of Environmental and Radiological Health Sciences/Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA, Instituto de Alta Investigacion, Universidad de Tarapaca, Arica 8097877, Chile, Division of Hematology and Oncology, Department of Pediatrics, Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA, Department of Medicine/Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901-1914, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 980033, USA, Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, 90127 Palermo, Italy, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Graduate School of Biomedical Sciences and Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084-1501, USA, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontari
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | - Rafaela Andrade-Vieira
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Sarah N Bay
- Program in Genetics and Molecular Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences/Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Gloria M Calaf
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA, Instituto de Alta Investigacion, Universidad de Tarapaca, Arica 8097877, Chile
| | - Robert C Castellino
- Division of Hematology and Oncology, Department of Pediatrics, Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Karine A Cohen-Solal
- Department of Medicine/Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901-1914, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Nichola Cruickshanks
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 980033, USA
| | - Paul Dent
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 980033, USA
| | - Riccardo Di Fiore
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, 90127 Palermo, Italy
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Gary S Goldberg
- Graduate School of Biomedical Sciences and Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084-1501, USA
| | - Roslida A Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
| | - Harini Krishnan
- Graduate School of Biomedical Sciences and Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084-1501, USA
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Ahmed Lasfar
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 60503, USA
| | - Paola A Marignani
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Richard Ponce-Cusi
- Instituto de Alta Investigacion, Universidad de Tarapaca, Arica 8097877, Chile
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Debasish Roy
- Department of Natural Science, The City University of New York at Hostos Campus, Bronx, NY 10451, USA
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences/Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Hosni K Salem
- Urology Dept., kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George's Medical University, Lucknow, UP 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Renza Vento
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, 90127 Palermo, Italy, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics AS CR, Brno 612 65, Czech Republic
| | - Mark Wade
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan 16163, Italy and
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
49
|
Intracellular Cleavage of the Cx43 C-Terminal Domain by Matrix-Metalloproteases: A Novel Contributor to Inflammation? Mediators Inflamm 2015; 2015:257471. [PMID: 26424967 PMCID: PMC4573893 DOI: 10.1155/2015/257471] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/13/2015] [Indexed: 01/11/2023] 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 connexin (Cx) proteins of which Cx43 is most widespread in the human body. Beyond its role in direct intercellular communication, Cx43 also forms nonjunctional hemichannels (HCs) in the plasma membrane that mediate the release of paracrine signaling molecules in the extracellular environment. Both HC and GJ channel function are regulated by protein-protein interactions and posttranslational modifications that predominantly take place in the C-terminal domain of Cx43. Matrix metalloproteases (MMPs) are a major group of zinc-dependent proteases, known to regulate not only extracellular matrix remodeling, but also processing of intracellular proteins. Together with Cx43 channels, both GJs and HCs, MMPs contribute to acute inflammation and a small number of studies reports on an MMP-Cx43 link. Here, we build further on these reports and present a novel hypothesis that describes proteolytic cleavage of the Cx43 C-terminal domain by MMPs and explores possibilities of how such cleavage events may affect Cx43 channel function. Finally, we set out how aberrant channel function resulting from cleavage can contribute to the acute inflammatory response during tissue injury.
Collapse
|
50
|
Mrozik KM, Cheong CM, Hewett D, Chow AWS, Blaschuk OW, Zannettino ACW, Vandyke K. Therapeutic targeting of N-cadherin is an effective treatment for multiple myeloma. Br J Haematol 2015; 171:387-99. [PMID: 26194766 DOI: 10.1111/bjh.13596] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/14/2015] [Indexed: 12/13/2022]
Abstract
Elevated expression of the cell adhesion molecule N-cadherin (cadherin 2, type 1, N-cadherin (neuronal); CDH2) is associated with poor prognosis in newly-diagnosed multiple myeloma (MM) patients. In this study, we investigated whether targeting of N-cadherin represents a potential treatment for the ~50% of MM patients with elevated N-cadherin. Initially, we stably knocked-down N-cadherin in the mouse MM plasma cell (PC) line 5TGM1 to assess the functional role of N-cadherin in MM pathogenesis. When compared with 5TGM1-scramble-shRNA cells, 5TGM1-Cdh2-shRNA cells had significantly reduced adhesion to bone marrow endothelial cells. However, N-cadherin knock-down did not affect 5TGM1 cell proliferation or adhesion to bone marrow stromal cells. In the C57BL/KaLwRij murine MM model, mice intravenously inoculated with 5TGM1-Cdh2-shRNA cells showed significantly decreased tumour burden after 4 weeks, compared with animals bearing 5TGM1-scramble-shRNA cells. Finally, the N-cadherin antagonist ADH-1 had no effect on tumour burden in the established disease setting, whereas up-front ADH-1 treatment resulted in significantly reduced tumour burden after 4 weeks. Our findings demonstrate that N-cadherin may play a key role in the extravasation of circulating MM PCs promoting bone marrow homing. Moreover, these studies suggest that N-cadherin may represent a viable therapeutic target to prevent the dissemination of MM PCs and delay MM disease progression.
Collapse
Affiliation(s)
- Krzysztof M Mrozik
- Myeloma Research Laboratory, School of Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Chee Man Cheong
- Myeloma Research Laboratory, School of Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Duncan Hewett
- Myeloma Research Laboratory, School of Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Annie W S Chow
- Myeloma Research Laboratory, School of Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Orest W Blaschuk
- Division of Urology, Department of Surgery, McGill University, Montreal, Canada
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, School of Medical Sciences, University of Adelaide, Adelaide, Australia.,Centre for Cancer Biology and Hanson Institute, SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia.,Centre for Stem Cell Research, Robinson Institute, University of Adelaide, Adelaide, Australia.,Centre for Personalised Cancer Medicine, University of Adelaide, Adelaide, Australia
| | - Kate Vandyke
- Myeloma Research Laboratory, School of Medical Sciences, University of Adelaide, Adelaide, Australia.,Centre for Cancer Biology and Hanson Institute, SA Pathology, Adelaide, Australia.,School of Medicine, University of Adelaide, Adelaide, Australia
| |
Collapse
|