1
|
Myo Min KK, Ffrench CB, McClure BJ, Ortiz M, Dorward EL, Samuel MS, Ebert LM, Mahoney MG, Bonder CS. Desmoglein-2 as a cancer modulator: friend or foe? Front Oncol 2023; 13:1327478. [PMID: 38188287 PMCID: PMC10766750 DOI: 10.3389/fonc.2023.1327478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Desmoglein-2 (DSG2) is a calcium-binding single pass transmembrane glycoprotein and a member of the large cadherin family. Until recently, DSG2 was thought to only function as a cell adhesion protein embedded within desmosome junctions designed to enable cells to better tolerate mechanical stress. However, additional roles for DSG2 outside of desmosomes are continuing to emerge, particularly in cancer. Herein, we review the current literature on DSG2 in cancer and detail its impact on biological functions such as cell adhesion, proliferation, migration, invasion, intracellular signaling, extracellular vesicle release and vasculogenic mimicry. An increased understanding of the diverse repertoire of the biological functions of DSG2 holds promise to exploit this cell surface protein as a potential prognostic biomarker and/or target for better patient outcomes. This review explores the canonical and non-canonical functions of DSG2, as well as the context-dependent impacts of DSG2 in the realm of cancer.
Collapse
Affiliation(s)
- Kay K. Myo Min
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Charlie B. Ffrench
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Barbara J. McClure
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Michael Ortiz
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Emma L. Dorward
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Michael S. Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Basil Hetzel Institute, Queen Elizabeth Hospital, SA, Adelaide, Australia
| | - Lisa M. Ebert
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Mỹ G. Mahoney
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Claudine S. Bonder
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
2
|
Paris O, Mennechet FJD, Kremer EJ. Human innate lymphoid cell activation by adenoviruses is modified by host defense proteins and neutralizing antibodies. Front Immunol 2022; 13:975910. [PMID: 36275713 PMCID: PMC9579290 DOI: 10.3389/fimmu.2022.975910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Innate lymphoid cells (ILCs), the complements of diverse CD4 T helper cells, help maintain tissue homeostasis by providing a link between innate and adaptive immune responses. While pioneering studies over the last decade have advanced our understanding how ILCs influence adaptive immune responses to pathogens, far less is known about whether the adaptive immune response feeds back into an ILC response. In this study, we isolated ILCs from blood of healthy donors, fine-tuned culture conditions, and then directly challenged them with human adenoviruses (HAdVs), with HAdVs and host defense proteins (HDPs) or neutralizing antibodies (NAbs), to mimic interactions in a host with pre-existing immunity. Additionally, we developed an ex vivo approach to identify how bystander ILCs respond to the uptake of HAdVs ± neutralizing antibodies by monocyte-derived dendritic cells. We show that ILCs take up HAdVs, which induces phenotypic maturation and cytokine secretion. Moreover, NAbs and HDPs complexes modified the cytokine profile generated by ILCs, consistent with a feedback loop for host antiviral responses and potential to impact adenovirus-based vaccine efficacy.
Collapse
|
3
|
Bernegger S, Jarzab M, Wessler S, Posselt G. Proteolytic Landscapes in Gastric Pathology and Cancerogenesis. Int J Mol Sci 2022; 23:2419. [PMID: 35269560 PMCID: PMC8910283 DOI: 10.3390/ijms23052419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Gastric cancer is a leading cause of cancer-related death, and a large proportion of cases are inseparably linked to infections with the bacterial pathogen and type I carcinogen Helicobacter pylori. The development of gastric cancer follows a cascade of transformative tissue events in an inflammatory environment. Proteases of host origin as well as H. pylori-derived proteases contribute to disease progression at every stage, from chronic gastritis to gastric cancer. In the present article, we discuss the importance of (metallo-)proteases in colonization, epithelial inflammation, and barrier disruption in tissue transformation, deregulation of cell proliferation and cell death, as well as tumor metastasis and neoangiogenesis. Proteases of the matrix metalloproteinase (MMP) and a disintegrin and metalloproteinase domain-containing protein (ADAM) families, caspases, calpain, and the H. pylori proteases HtrA, Hp1012, and Hp0169 cleave substrates including extracellular matrix molecules, chemokines, and cytokines, as well as their cognate receptors, and thus shape the pathogenic microenvironment. This review aims to summarize the current understanding of how proteases contribute to disease progression in the gastric compartment.
Collapse
Affiliation(s)
- Sabine Bernegger
- Division of Microbiology, Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, 5020 Salzburg, Austria; (S.B.); (M.J.); (S.W.)
| | - Miroslaw Jarzab
- Division of Microbiology, Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, 5020 Salzburg, Austria; (S.B.); (M.J.); (S.W.)
| | - Silja Wessler
- Division of Microbiology, Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, 5020 Salzburg, Austria; (S.B.); (M.J.); (S.W.)
- Cancer Cluster Salzburg and Allergy Cancer BioNano Research Centre, University of Salzburg, Hellbrunner Strasse 34, 5020 Salzburg, Austria
| | - Gernot Posselt
- Division of Microbiology, Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, 5020 Salzburg, Austria; (S.B.); (M.J.); (S.W.)
| |
Collapse
|
4
|
Flemming JP, Hill BL, Anderson-Pullinger L, Harshyne LA, Mahoney MG. Cytokine Profiling in Low- and High-Density Small Extracellular Vesicles from Epidermoid Carcinoma Cells. JID INNOVATIONS 2021; 1:100053. [PMID: 34909749 PMCID: PMC8659799 DOI: 10.1016/j.xjidi.2021.100053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 01/08/2023] Open
Abstract
Exosomes or small extracellular vesicles (sEVs) are membrane-bound nanoparticles that carry various macromolecules and act as autocrine and paracrine signaling messengers. In this study, sEVs from epidermoid carcinoma cells influenced by membrane presentation of the glycoprotein desmoglein 2 and its palmitoylation state were investigated. In this study, sEVs were isolated by sequential ultracentrifugation followed by iodixanol density gradient separation. They were then subjected to multiplex profiling of cytokines associated with the surface of intact sEVs. The results revealed a previously undescribed active sorting of cytokines onto the surface of low-density and high-density sEV subpopulations. Specifically, an altered surface presentation of desmoglein 2 decreased FGF-2 and VEGF in low-density sEVs. In addition, in response to desmoglein 2, IL-8 and RANTES were increased in low-density sEVs but only slightly decreased in high-density sEVs. Finally, IL-6 and G-CSF were increased dramatically in high-density sEVs. This comprehensive analysis of the cytokine production profile by squamous cell carcinoma‒derived sEVs highlights their contribution to immune evasion, pro-oncogenic and proangiogenic activity, and the potential to identify diagnostic disease biomarkers.
Collapse
Affiliation(s)
- Joseph P. Flemming
- Department of Dermatology & Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Brianna L. Hill
- Department of Dermatology & Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Larry A. Harshyne
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mỹ G. Mahoney
- Department of Dermatology & Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Otolaryngology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Raya-Sandino A, Luissint AC, Kusters DHM, Narayanan V, Flemming S, Garcia-Hernandez V, Godsel LM, Green KJ, Hagen SJ, Conway DE, Parkos CA, Nusrat A. Regulation of intestinal epithelial intercellular adhesion and barrier function by desmosomal cadherin desmocollin-2. Mol Biol Cell 2021; 32:753-768. [PMID: 33596089 PMCID: PMC8108520 DOI: 10.1091/mbc.e20-12-0775] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/26/2022] Open
Abstract
The role of desmosomal cadherin desmocollin-2 (Dsc2) in regulating barrier function in intestinal epithelial cells (IECs) is not well understood. Here, we report the consequences of silencing Dsc2 on IEC barrier function in vivo using mice with inducible intestinal-epithelial-specific Dsc2 knockdown (KD) (Dsc2ERΔIEC). While the small intestinal gross architecture was maintained, loss of epithelial Dsc2 influenced desmosomal plaque structure, which was smaller in size and had increased intermembrane space between adjacent epithelial cells. Functional analysis revealed that loss of Dsc2 increased intestinal permeability in vivo, supporting a role for Dsc2 in the regulation of intestinal epithelial barrier function. These results were corroborated in model human IECs in which Dsc2 KD resulted in decreased cell-cell adhesion and impaired barrier function. It is noteworthy that Dsc2 KD cells exhibited delayed recruitment of desmoglein-2 (Dsg2) to the plasma membrane after calcium switch-induced intercellular junction reassembly, while E-cadherin accumulation was unaffected. Mechanistically, loss of Dsc2 increased desmoplakin (DP I/II) protein expression and promoted intermediate filament interaction with DP I/II and was associated with enhanced tension on desmosomes as measured by a Dsg2-tension sensor. In conclusion, we provide new insights on Dsc2 regulation of mechanical tension, adhesion, and barrier function in IECs.
Collapse
Affiliation(s)
- Arturo Raya-Sandino
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Dennis H. M. Kusters
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Vani Narayanan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Sven Flemming
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | | | - Lisa M. Godsel
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Kathleen J. Green
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Susan J. Hagen
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Daniel E. Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Charles A. Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
6
|
Hemi- and Homozygous Loss-of-Function Mutations in DSG2 (Desmoglein-2) Cause Recessive Arrhythmogenic Cardiomyopathy with an Early Onset. Int J Mol Sci 2021; 22:ijms22073786. [PMID: 33917638 PMCID: PMC8038858 DOI: 10.3390/ijms22073786] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
About 50% of patients with arrhythmogenic cardiomyopathy (ACM) carry a pathogenic or likely pathogenic mutation in the desmosomal genes. However, there is a significant number of patients without positive familial anamnesis. Therefore, the molecular reasons for ACM in these patients are frequently unknown and a genetic contribution might be underestimated. Here, we used a next-generation sequencing (NGS) approach and in addition single nucleotide polymor-phism (SNP) arrays for the genetic analysis of two independent index patients without familial medical history. Of note, this genetic strategy revealed a homozygous splice site mutation (DSG2–c.378+1G>T) in the first patient and a nonsense mutation (DSG2–p.L772X) in combination with a large deletion in DSG2 in the second one. In conclusion, a recessive inheritance pattern is likely for both cases, which might contribute to the hidden medical history in both families. This is the first report about these novel loss-of-function mutations in DSG2 that have not been previously identi-fied. Therefore, we suggest performing deep genetic analyses using NGS in combination with SNP arrays also for ACM index patients without obvious familial medical history. In the future, this finding might has relevance for the genetic counseling of similar cases.
Collapse
|
7
|
Dudley JS, Murphy CR, Thompson MB, Lindsay LA, McAllan BM. Sex steroids influence the plasma membrane transformation in the uterus of the fat-tailed dunnart (Sminthopsis crassicaudata, Marsupialia). Reprod Fertil Dev 2019; 31:633-644. [PMID: 30449299 DOI: 10.1071/rd18202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022] Open
Abstract
The uterine epithelium undergoes remodelling to become receptive to blastocyst implantation during pregnancy in a process known as the plasma membrane transformation. There are commonalities in ultrastructural changes to the epithelium, which, in eutherian, pregnancies are controlled by maternal hormones, progesterone and oestrogens. The aim of this study was to determine the effects that sex steroids have on the uterine epithelium in the fat-tailed dunnart Sminthopsis crassicaudata, the first such study in a marsupial. Females were exposed to exogenous hormones while they were reproductively quiescent, thus not producing physiological concentrations of ovarian hormones. We found that changes to the protein E-cadherin, which forms part of the adherens junction, are controlled by progesterone and that changes to the desmoglein-2 protein, which forms part of desmosomes, are controlled by 17β-oestradiol. Exposure to a combination of progesterone and 17β-oestradiol causes changes to the microvilli on the apical surface and to the ultrastructure of the uterine epithelium. There is a decrease in lateral adhesion when the uterus is exposed to progesterone and 17β-oestradiol that mimics the hormone environment of uterine receptivity. We conclude that uterine receptivity and the plasma membrane transformation in marsupial and eutherian pregnancies are under the same endocrine control and may be an ancestral feature of therian mammals.
Collapse
Affiliation(s)
- Jessica S Dudley
- School of Medical Sciences and Bosch Institute, Anderson Stuart Building (F13), University of Sydney, Sydney, NSW 2006, Australia
| | - Christopher R Murphy
- School of Medical Sciences and Bosch Institute, Anderson Stuart Building (F13), University of Sydney, Sydney, NSW 2006, Australia
| | - Michael B Thompson
- School of Life and Environmental Science, Heydon-Laurence Building (A08), University of Sydney, Sydney, NSW 2006, Australia
| | - Laura A Lindsay
- School of Medical Sciences and Bosch Institute, Anderson Stuart Building (F13), University of Sydney, Sydney, NSW 2006, Australia
| | - Bronwyn M McAllan
- School of Medical Sciences and Bosch Institute, Anderson Stuart Building (F13), University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
8
|
Headrick AT, Rosenfeld JA, Yang Y, Tunuguntla H, Allen HD, Penny DJ, Kim JJ, Landstrom AP. Incidentally identified genetic variants in arrhythmogenic right ventricular cardiomyopathy-associated genes among children undergoing exome sequencing reflect healthy population variation. Mol Genet Genomic Med 2019; 7:e593. [PMID: 30985088 PMCID: PMC6565596 DOI: 10.1002/mgg3.593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/03/2019] [Accepted: 01/06/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND With expanding use of clinical whole exome sequencing (WES), genetic variants of uncertain significance are increasingly identified. As pathologic mutations in genes associated with arrhythmogenic right ventricular cardiomyopathy (ARVC) carry a risk of sudden death, determining the diagnostic relevance of incidentally identified variants associated with these genes is critical. METHODS WES variants from a large, predominantly pediatric cohort (N = 7,066 probands) were obtained for nine ARVC-associated genes (Baylor Miraca). For comparison, a control cohort was derived from the gnomAD database and an ARVC case cohort (N = 1,379 probands) was established from ARVC cases in the literature. Topologic mapping was performed and signal-to-noise analysis was conducted normalizing WES, or case variants, against control variant frequencies. Retrospective chart review was performed of WES cases evaluated clinically (Texas Children's Hospital). RESULTS Incidentally identified variants occurred in 14% of WES referrals and localized to genes which were rare among ARVC cases yet similar to controls. Amino acid-level signal-to-noise analysis of cases demonstrated "pathologic hotspots" localizing to critical domains of PKP2 and DSG2 while WES variants did not. PKP2 ARM7 and ARM8 domains and DSG2 N-terminal cadherin-repeat domains demonstrated high pathogenicity while normalized WES variant frequency was low. Review of clinical data available on WES referrals demonstrated none with evidence of ARVC among variant-positive individuals. CONCLUSIONS Incidentally identified variants are common among pediatric WES testing with gene frequencies similar to "background" variants. Incidentally identified variants are unlikely to be pathologic.
Collapse
Affiliation(s)
- Andrew T. Headrick
- Department of Pediatrics, Section of Pediatric CardiologyBaylor College of MedicineHoustonTexas
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics and Baylor Genetics LaboratoriesBaylor College of MedicineHoustonTexas
| | - Yaping Yang
- Department of Molecular and Human Genetics and Baylor Genetics LaboratoriesBaylor College of MedicineHoustonTexas
| | - Hari Tunuguntla
- Department of Pediatrics, Section of Pediatric CardiologyBaylor College of MedicineHoustonTexas
| | - Hugh D. Allen
- Department of Pediatrics, Section of Pediatric CardiologyBaylor College of MedicineHoustonTexas
| | - Daniel J. Penny
- Department of Pediatrics, Section of Pediatric CardiologyBaylor College of MedicineHoustonTexas
| | - Jeffrey J. Kim
- Department of Pediatrics, Section of Pediatric CardiologyBaylor College of MedicineHoustonTexas
| | - Andrew P. Landstrom
- Department of Pediatrics, Section of Pediatric CardiologyBaylor College of MedicineHoustonTexas
- Department of Pediatrics, Division of Pediatric CardiologyDuke University School of MedicineDurhamNorth Carolina
- Present address:
Department of Pediatrics, Division of Pediatric CardiologyDuke University School of MedicineDuke University Medical CenterDurhamNorth Carolina
| |
Collapse
|
9
|
Lu CS, Lee YN, Wang SW, Wu YJ, Su CH, Hsieh CL, Tien TY, Wang BJ, Chen MC, Chen CW, Yeh HI. KC21 Peptide Inhibits Angiogenesis and Attenuates Hypoxia-Induced Retinopathy. J Cardiovasc Transl Res 2019; 12:366-377. [PMID: 30790141 PMCID: PMC6707963 DOI: 10.1007/s12265-019-09865-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/20/2019] [Indexed: 12/31/2022]
Abstract
Desmogleins (Dsg2) are the major components of desmosomes. Dsg2 has five extracellular tandem cadherin domains (EC1-EC5) for cell-cell interaction. We had previously confirmed the Dsg2 antibody and its epitope (named KC21) derived from EC2 domain suppressing epithelial-mesenchymal transition and invasion in human cancer cell lines. Here, we screened six peptide fragments derived from EC2 domain and found that KR20, the parental peptide of KC21, was the most potent one on suppressing endothelial colony-forming cell (ECFC) tube-like structure formation. KC21 peptide also attenuated migration but did not disrupt viability and proliferation of ECFCs, consistent with the function to inhibit VEGF-mediated activation of p38 MAPK but not AKT and ERK. Animal studies showed that KC21 peptides suppressed capillary growth in Matrigel implant assay and inhibited oxygen-induced retinal neovascularization. The effects were comparable to bevacizumab (Bev). In conclusion, KC21 peptide is an angiogenic inhibitor potentially useful for treating angiogenesis-related diseases.
Collapse
Affiliation(s)
- Chi-Sheng Lu
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan.,Virginia Contract Research Organization Co., Ltd, Taipei, 11491, Taiwan
| | - Yi-Nan Lee
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan
| | - Shin-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan
| | - Yih-Jer Wu
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan.,Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan
| | - Cheng-Huang Su
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan
| | - Chin-Ling Hsieh
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan
| | - Ting Yi Tien
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan
| | - Bo-Jeng Wang
- Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan
| | - Min-Che Chen
- Asclepiumm Taiwan Co., Ltd, New Taipei City, 25160, Taiwan
| | - Chun-Wei Chen
- Asclepiumm Taiwan Co., Ltd, New Taipei City, 25160, Taiwan
| | - Hung-I Yeh
- Departments of Medical Research, Mackay Memorial Hospital, Taipei, 10449, Taiwan. .,Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan. .,Departments of Medical Research and Internal Medicine, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd, Taipei City, 10449, Taiwan.
| |
Collapse
|
10
|
Yulis M, Quiros M, Hilgarth R, Parkos CA, Nusrat A. Intracellular Desmoglein-2 cleavage sensitizes epithelial cells to apoptosis in response to pro-inflammatory cytokines. Cell Death Dis 2018; 9:389. [PMID: 29523777 PMCID: PMC5844960 DOI: 10.1038/s41419-018-0380-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 12/29/2022]
Abstract
Desmosomal cadherins mediate intercellular adhesion and have also been shown to regulate homeostatic signaling in epithelial cells. We have previously reported that select pro-inflammatory cytokines induce Dsg2 ectodomain cleavage and shedding from intestinal epithelial cells (IECs). Dsg2 extracellular cleaved fragments (Dsg2 ECF) function to induce paracrine pro-proliferative signaling in epithelial cells. In this study, we show that exposure of IECs to pro-inflammatory cytokines interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) resulted in Dsg2 intracellular cleavage and generation of a ~55 kDa fragment (Dsg2 ICF). Dsg2 intracellular cleavage is mediated by caspase-8 and occurs prior to Dsg2 extracellular cleavage and the execution of apoptosis. Expression of exogenous Dsg2 ICF in model IECs resulted in increased sensitivity to apoptotic stimuli and apoptosis execution. Additionally, expression of the Dsg2 ICF repressed the anti-apoptotic Bcl-2 family member proteins Bcl-XL and Mcl1. Taken together, our findings identify a novel mechanism by which pro-inflammatory mediators induce modification of Dsg2 to activate apoptosis and eliminate damaged cells, while also promoting release of Dsg2 ECF that promotes proliferation of neighboring cells and epithelial barrier recovery.
Collapse
Affiliation(s)
- Mark Yulis
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Miguel Quiros
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Roland Hilgarth
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Charles A Parkos
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Asma Nusrat
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
11
|
Overmiller AM, Pierluissi JA, Wermuth PJ, Sauma S, Martinez-Outschoorn U, Tuluc M, Luginbuhl A, Curry J, Harshyne LA, Wahl JK, South AP, Mahoney MG. Desmoglein 2 modulates extracellular vesicle release from squamous cell carcinoma keratinocytes. FASEB J 2017; 31:3412-3424. [PMID: 28438789 DOI: 10.1096/fj.201601138rr] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 04/11/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane-derived vesicles that serve as intercellular messengers carrying lipids, proteins, and genetic material. Substantial evidence has shown that cancer-derived EVs, secreted by tumor cells into the blood and other bodily fluids, play a critical role in modulating the tumor microenvironment and affecting the pathogenesis of cancer. Here we demonstrate for the first time that squamous cell carcinoma (SCC) EVs were enriched with the C-terminal fragment of desmoglein 2 (Dsg2), a desmosomal cadherin often overexpressed in malignancies. Overexpression of Dsg2 increased EV release and mitogenic content including epidermal growth factor receptor and c-Src. Inhibiting ectodomain shedding of Dsg2 with the matrix metalloproteinase inhibitor GM6001 resulted in accumulation of full-length Dsg2 in EVs and reduced EV release. When cocultured with Dsg2/green fluorescence protein-expressing SCC cells, green fluorescence protein signal was detected by fluorescence-activated cell sorting analysis in the CD90+ fibroblasts. Furthermore, SCC EVs activated Erk1/2 and Akt signaling and enhanced fibroblast cell proliferation. In vivo, Dsg2 was highly up-regulated in the head and neck SCCs, and EVs isolated from sera of patients with SCC were enriched in Dsg2 C-terminal fragment and epidermal growth factor receptor. This study defines a mechanism by which Dsg2 expression in cancer cells can modulate the tumor microenvironment, a step critical for tumor progression.-Overmiller, A. M., Pierluissi, J. A., Wermuth, P. J., Sauma, S., Martinez-Outschoorn, U., Tuluc, M., Luginbuhl, A., Curry, J., Harshyne, L. A., Wahl, J. K. III, South, A. P., Mahoney, M. G. Desmoglein 2 modulates extracellular vesicle release from squamous cell carcinoma keratinocytes.
Collapse
Affiliation(s)
- Andrew M Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jennifer A Pierluissi
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Peter J Wermuth
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sami Sauma
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Madalina Tuluc
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam Luginbuhl
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Joseph Curry
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Larry A Harshyne
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - James K Wahl
- Department of Oral Biology, University of Nebraska, Lincoln, Nebraska, USA
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mỹ G Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
12
|
Desmoglein-2 is overexpressed in non-small cell lung cancer tissues and its knockdown suppresses NSCLC growth by regulation of p27 and CDK2. J Cancer Res Clin Oncol 2016; 143:59-69. [PMID: 27629878 DOI: 10.1007/s00432-016-2250-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 08/31/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Desmoglein-2 (Dsg2) is a cell adhesion protein of the cadherin superfamily. Altered Dsg2 expression is associated with tumorigenesis. This study determined Dsg2 expression in non-small cell lung cancer (NSCLC) tissue specimens for association with clinicopathological and survival data and then assessed the effect of Dsg2 knockdown on regulation of NSCLC cell malignant behaviors in vitro and in nude mouse xenografts. METHODS qRT-PCR and Western blot were used to detect Dsg2 expression in 28 paired NSCLC and normal tissue samples. Immunohistochemistry was used to detect Dsg2 expression in 70 cases of paraffin-embedded NSCLC tissues. NSCLC A549, H1703, and H1299 cells were cultured with Dsg2 knockdown performed using Dsg2 siRNA. Cell viability, cell cycle, apoptosis, and colony formation were assessed. siRNA-transfected A549 cells were also used to generate tumor xenografts in nude mice. RESULTS Both Dsg2 mRNA and protein were highly expressed in NSCLC tissues and associated with NSCLC size, but not with overall survival of patients. Moreover, knockdown of Dsg2 expression reduced NSCLC cell proliferation and arrested them at the G1 phase of the cell cycle, but did not significantly affect NSCLC cell apoptosis. Dsg2 knockdown downregulated cyclin-dependent kinase 2 expression and upregulated p27 expression. Nude mouse xenograft assays showed that Dsg2 knockdown inhibited NSCLC xenograft growth in vivo. CONCLUSION This study revealed the importance of Dsg2 in suppression of NSCLC development and progression. Further studies will explore whether restoration of Dsg2 expression is a novel strategy in control of NSCLC.
Collapse
|
13
|
Abstract
BACKGROUND The intestinal epithelium of patients with Crohn's disease (CD) is characterized by defects in permeability and alterations in tight junction morphology sealing the paracellular cleft. Desmosomes are primarily considered to mediate strong intercellular cohesion. Because barrier properties of epithelial cells were shown to depend on the function of the desmosomal adhesion molecule desmoglein 2 (Dsg2), we here investigated the relevance of Dsg2 for CD. METHODS Biopsies from the terminal ileum of 14 patients with CD and 12 healthy controls were investigated for changes in cell adhesion molecules. Two intestinal epithelial cell lines were used for functional studies. A tandem peptide modulating Dsg binding was applied to strengthen Dsg2 interaction. RESULTS Dsg2 but not the adherens junction molecule E-cadherin was strongly reduced in the mucosa of patients with CD. TNF-α, a central cytokine in CD pathogenesis, led to loss of cell cohesion and increased permeability in cultured epithelial cells, which was paralleled by loss of Dsg2 at cell borders, reduction of the tight junction component claudin-1, and upregulation of claudin-2. These effects were mediated at least in part by increased activity of p38MAPK because inhibition of this kinase restored intercellular adhesion and blunted the permeability increase induced by TNF-α. Importantly, stabilizing desmosomal adhesion through tandem peptide ameliorated loss of barrier functions and prevented claudin-2 increase. CONCLUSIONS We show an important role of p38MAPK-mediated regulation of desmosomal adhesion resulting in upregulation of claudin-2 in CD. Our data suggest peptide-mediated strengthening of impaired Dsg2 adhesion as a novel therapeutic approach in CD.
Collapse
|
14
|
Kamekura R, Nava P, Feng M, Quiros M, Nishio H, Weber DA, Parkos CA, Nusrat A. Inflammation-induced desmoglein-2 ectodomain shedding compromises the mucosal barrier. Mol Biol Cell 2015. [PMID: 26224314 PMCID: PMC4569309 DOI: 10.1091/mbc.e15-03-0147] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Proinflammatory cytokines promote desmoglein-2 (Dsg2) ectodomain shedding in intestinal epithelial cells. Epithelial exposure to Dsg2 ectodomains compromises intercellular adhesion while also promoting proliferation. These findings identify mechanisms by which mucosal inflammation–induced cleavage of Dsg2 influences intestinal epithelial homeostasis. Desmosomal cadherins mediate intercellular adhesion and control epithelial homeostasis. Recent studies show that proteinases play an important role in the pathobiology of cancer by targeting epithelial intercellular junction proteins such as cadherins. Here we describe the proinflammatory cytokine-induced activation of matrix metalloproteinase 9 and a disintegrin and metalloproteinase domain–containing protein 10, which promote the shedding of desmosomal cadherin desmoglein-2 (Dsg2) ectodomains in intestinal epithelial cells. Epithelial exposure to Dsg2 ectodomains compromises intercellular adhesion by promoting the relocalization of endogenous Dsg2 and E-cadherin from the plasma membrane while also promoting proliferation by activation of human epidermal growth factor receptor 2/3 signaling. Cadherin ectodomains were detected in the inflamed intestinal mucosa of mice with colitis and patients with ulcerative colitis. Taken together, our findings reveal a novel response pathway in which inflammation-induced modification of columnar epithelial cell cadherins decreases intercellular adhesion while enhancing cellular proliferation, which may serve as a compensatory mechanism to promote repair.
Collapse
Affiliation(s)
- Ryuta Kamekura
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322 Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 0608556, Japan
| | - Porfirio Nava
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322 Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies, Mexico DF 07360, Mexico
| | - Mingli Feng
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322 Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Miguel Quiros
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322 Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Hikaru Nishio
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322
| | - Dominique A Weber
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322
| | - Charles A Parkos
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322 Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Asma Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322 Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
15
|
Sumagin R, Parkos CA. Epithelial adhesion molecules and the regulation of intestinal homeostasis during neutrophil transepithelial migration. Tissue Barriers 2015; 3:e969100. [PMID: 25838976 DOI: 10.4161/21688362.2014.969100] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 08/14/2014] [Indexed: 12/19/2022] Open
Abstract
Epithelial adhesion molecules play essential roles in regulating cellular function and maintaining mucosal tissue homeostasis. Some form epithelial junctional complexes to provide structural support for epithelial monolayers and act as a selectively permeable barrier separating luminal contents from the surrounding tissue. Others serve as docking structures for invading viruses and bacteria, while also regulating the immune response. They can either obstruct or serve as footholds for the immune cells recruited to mucosal surfaces. Currently, it is well appreciated that adhesion molecules collectively serve as environmental cue sensors and trigger signaling events to regulate epithelial function through their association with the cell cytoskeleton and various intracellular adapter proteins. Immune cells, particularly neutrophils (PMN) during transepithelial migration (TEM), can modulate adhesion molecule expression, conformation, and distribution, significantly impacting epithelial function and tissue homeostasis. This review discusses the roles of key intestinal epithelial adhesion molecules in regulating PMN trafficking and outlines the potential consequences on epithelial function.
Collapse
Key Words
- AJs, adherens junctions
- CAR, coxsackie and adenovirus receptor
- CLMP, CAR-like protein
- CTLs, cytotoxic T lymphocytes
- CTX, thymocyte Xenopus
- DMs, Desmosomes
- Dsc-2, desmocollin-2
- Dsg-2, desmoglein-2
- E-cadherin, epithelial cadherin
- EGFR, Epithelial growth factor receptor
- EMT, epithelial-mesenchymal transition
- EpCAM, epithelial cell adhesion molecule
- IBD, inflammatory bowel diseases
- ICAM-1, intercellular adhesion molecule-1
- IECs, intestinal epithelial cells
- JAM, junctional adhesion molecules
- LAD, leukocyte adhesion deficiency
- LTB-4, lipid leukotriene B4
- MIP1 α, macrophage inflammatory protein 1 alpha
- MLCK, myosin light chain kinase
- MMPs, matrix metalloproteases
- NF-κB, nuclear factor kappa B
- NO, nitric oxide
- PARS, protease-activated receptors
- PI3K, phosphatidylinositol 3-kinase
- PMN, polymorphonuclear cells
- SGD, specific granule deficiency
- SIRPa, signal regulatory protein alpha
- TEM, transepithelial migration
- TGF-β, transforming growth factor beta
- TIAM1, metastasis-inducing protein 1
- TJs, tight junctions
- TSP-1, thrombospondin-1
- adhesion molecules
- barrier
- cell migration
- epithelial cells
- neutrophils
- sLea, sialyl Lewis A
Collapse
Affiliation(s)
- Ronen Sumagin
- Department of Pathology and Laboratory Medicine; Epithelial Pathobiology and Mucosal Inflammation Unit; Emory University ; Atlanta, GA USA
| | - Charles A Parkos
- Department of Pathology and Laboratory Medicine; Epithelial Pathobiology and Mucosal Inflammation Unit; Emory University ; Atlanta, GA USA
| |
Collapse
|
16
|
Nava P, Kamekura R, Nusrat A. Cleavage of transmembrane junction proteins and their role in regulating epithelial homeostasis. Tissue Barriers 2014; 1:e24783. [PMID: 24665393 PMCID: PMC3879235 DOI: 10.4161/tisb.24783] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 04/19/2013] [Accepted: 04/23/2013] [Indexed: 02/07/2023] Open
Abstract
Epithelial tissues form a selective barrier that separates the external environment from the internal tissue milieu. Single epithelial cells are densely packed and associate via distinct intercellular junctions. Intercellular junction proteins not only control barrier properties of the epithelium but also play an important role in regulating epithelial homeostasis that encompasses cell proliferation, migration, differentiation and regulated shedding. Recent studies have revealed that several proteases target epithelial junction proteins during physiological maturation as well as in pathologic states such as inflammation and cancer. This review discusses mechanisms and biological consequences of transmembrane junction protein cleavage. The influence of junction protein cleavage products on pathogenesis of inflammation and cancer is discussed.
Collapse
Affiliation(s)
- Porfirio Nava
- Epithelial Pathobiology and Mucosal Inflammation Research Unit; Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA USA ; Department of Physiology; Biophysics and Neurosciences; Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV); México DF, Mexico
| | - Ryuta Kamekura
- Epithelial Pathobiology and Mucosal Inflammation Research Unit; Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA USA
| | - Asma Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Research Unit; Department of Pathology and Laboratory Medicine; Emory University School of Medicine; Atlanta, GA USA
| |
Collapse
|
17
|
Jiang K, Rankin CR, Nava P, Sumagin R, Kamekura R, Stowell SR, Feng M, Parkos CA, Nusrat A. Galectin-3 regulates desmoglein-2 and intestinal epithelial intercellular adhesion. J Biol Chem 2014; 289:10510-10517. [PMID: 24567334 DOI: 10.1074/jbc.m113.538538] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The desmosomal cadherins, desmogleins, and desmocollins mediate strong intercellular adhesion. Human intestinal epithelial cells express the desmoglein-2 isoform. A proteomic screen for Dsg2-associated proteins in intestinal epithelial cells identified a lectin referred to as galectin-3 (Gal3). Gal3 bound to N-linked β-galactosides in Dsg2 extracellular domain and co-sedimented with caveolin-1 in lipid rafts. Down-regulation of Gal3 protein or incubation with lactose, a galactose-containing disaccharide that competitively inhibits galectin binding to Dsg2, decreased intercellular adhesion in intestinal epithelial cells. In the absence of functional Gal3, Dsg2 protein was internalized from the plasma membrane and degraded in the proteasome. These results report a novel role of Gal3 in stabilizing a desmosomal cadherin and intercellular adhesion in intestinal epithelial cells.
Collapse
Affiliation(s)
- Kun Jiang
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Carl R Rankin
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Porfirio Nava
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322; Department of Physiology, Biophysics and Neuroscience, CINVESTAV IPN., Av. IPN 2508, Col. San Pedro Zacatenco, 07360 Ciudad de México, Distrito Federal, México
| | - Ronen Sumagin
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Ryuta Kamekura
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Sean R Stowell
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Mingli Feng
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Charles A Parkos
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Asma Nusrat
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322.
| |
Collapse
|
18
|
Kamekura R, Kolegraff KN, Nava P, Hilgarth RS, Feng M, Parkos CA, Nusrat A. Loss of the desmosomal cadherin desmoglein-2 suppresses colon cancer cell proliferation through EGFR signaling. Oncogene 2013; 33:4531-6. [PMID: 24166502 DOI: 10.1038/onc.2013.442] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/06/2013] [Accepted: 09/09/2013] [Indexed: 12/13/2022]
Abstract
Desmosomal cadherins mediate cell-cell adhesion in epithelial tissues and have been known to be altered in cancer. We have previously shown that one of the two intestinal epithelial desmosomal cadherins, desmocollin-2 (Dsc2) loss promotes colonic epithelial carcinoma cell proliferation and tumor formation. In this study we show that loss of the other intestinal desmosomal cadherin, desmoglein-2 (Dsg2) that pairs with Dsc2, results in decreased epithelial cell proliferation and suppressed xenograft tumor growth in mice. Dsg2-deficient cells demonstrated a compensatory increase in Dsc2 expression, and small interfering RNA-mediated loss of Dsc2 restored proliferation in Dsg2-deficient cells. Dsg2 downregulation inhibited epidermal growth factor receptor (EGFR) signaling and cell proliferation through altered phosphorylation of EGFR and downstream extracellular signal-regulated kinase activation in parallel with inhibited EGFR receptor internalization. Additionally, we demonstrated a central role of Dsc2 in controlling EGFR signaling and cell proliferation in intestinal epithelial cells. Consistent with these findings, analyses of human colon cancers demonstrated increased Dsg2 protein expression. Taken together, these data demonstrate that partner desmosomal cadherins Dsg2 and Dsc2 play opposing roles in controlling colonic carcinoma cell proliferation through differential effects on EGFR signaling.
Collapse
Affiliation(s)
- R Kamekura
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - K N Kolegraff
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - P Nava
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies (CINVESTAV), Mexico DF, Mexico
| | - R S Hilgarth
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - M Feng
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - C A Parkos
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - A Nusrat
- Epithelial Pathobiology and Mucosal Inflammation Research Unit, Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
19
|
Chen J, Nekrasova OE, Patel DM, Klessner JL, Godsel LM, Koetsier JL, Amargo EV, Desai BV, Green KJ. The C-terminal unique region of desmoglein 2 inhibits its internalization via tail-tail interactions. ACTA ACUST UNITED AC 2012; 199:699-711. [PMID: 23128240 PMCID: PMC3494854 DOI: 10.1083/jcb.201202105] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tail–tail interactions of desmoglein 2, promoted by its C-terminal unique region, inhibit its internalization, stabilizing it at the cell surface and promoting intercellular adhesion. Desmosomal cadherins, desmogleins (Dsgs) and desmocollins, make up the adhesive core of intercellular junctions called desmosomes. A critical determinant of epithelial adhesive strength is the level and organization of desmosomal cadherins on the cell surface. The Dsg subclass of desmosomal cadherins contains a C-terminal unique region (Dsg unique region [DUR]) with unknown function. In this paper, we show that the DUR of Dsg2 stabilized Dsg2 at the cell surface by inhibiting its internalization and promoted strong intercellular adhesion. DUR also facilitated Dsg tail–tail interactions. Forced dimerization of a Dsg2 tail lacking the DUR led to decreased internalization, supporting the conclusion that these two functions of the DUR are mechanistically linked. We also show that a Dsg2 mutant, V977fsX1006, identified in arrhythmogenic right ventricular cardiomyopathy patients, led to a loss of Dsg2 tail self-association and underwent rapid endocytosis in cardiac muscle cells. Our observations illustrate a new mechanism desmosomal cadherins use to control their surface levels, a key factor in determining their adhesion and signaling roles.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wang H, Beyer I, Persson J, Song H, Li Z, Richter M, Cao H, van Rensburg R, Yao X, Hudkins K, Yumul R, Zhang XB, Yu M, Fender P, Hemminki A, Lieber A. A new human DSG2-transgenic mouse model for studying the tropism and pathology of human adenoviruses. J Virol 2012; 86:6286-302. [PMID: 22457526 PMCID: PMC3372198 DOI: 10.1128/jvi.00205-12] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 03/16/2012] [Indexed: 12/12/2022] Open
Abstract
We have recently reported that a group of human adenoviruses (HAdVs) uses desmoglein 2 (DSG2) as a receptor for infection. Among these are the widely distributed serotypes HAdV-B3 and HAdV-B7, as well as a newly emerged strain derived from HAdV-B14. These serotypes do not infect rodent cells and could not up until now be studied in small-animal models. We therefore generated transgenic mice containing the human DSG2 locus. These mice expressed human DSG2 (hDSG2) at a level and in a pattern similar to those found for humans and nonhuman primates. As an initial application of hDSG2-transgenic mice, we used a green fluorescent protein (GFP)-expressing HAdV-B3 vector (Ad3-GFP) and studied GFP transgene expression by quantitative reverse transcription-PCR (qRT-PCR) and immunohistochemistry subsequent to intranasal and intravenous virus application. After intranasal application, we found efficient transduction of bronchial and alveolar epithelial cells in hDSG2-transgenic mice. Intravenous Ad3-GFP injection into hDSG2-transgenic mice resulted in hDSG2-dependent transduction of epithelial cells in the intestinal and colon mucosa. Our findings give an explanation for clinical symptoms associated with infection by DSG2-interacting HAdVs and provide a rationale for using Ad3-derived vectors in gene therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hua Cao
- Division of Medical Genetics
| | | | | | - Kelly Hudkins
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | | | - Xiao-Bing Zhang
- Loma Linda University, Department of Medicine, Division of Regenerative Medicine, Loma Linda, California, USA
| | - Mujun Yu
- Medical Laboratory Associates, Seattle, Washington, USA
| | - Pascal Fender
- Unit of Virus Host Cell Interactions, UMI3265, CNRS/EMBL/UJF, Grenoble, France
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Molecular Cancer Biology Research Program, Transplantation Laboratory & Haartman Institute, University of Helsinki, Helsinki, Finland
| | - André Lieber
- Division of Medical Genetics
- Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
21
|
Beyer I, van Rensburg R, Strauss R, Li Z, Wang H, Persson J, Yumul R, Feng Q, Song H, Bartek J, Fender P, Lieber A. Epithelial junction opener JO-1 improves monoclonal antibody therapy of cancer. Cancer Res 2011; 71:7080-90. [PMID: 21990319 DOI: 10.1158/0008-5472.can-11-2009] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The efficacy of monoclonal antibodies (mAb) used to treat solid tumors is limited by intercellular junctions which tightly link epithelial tumor cells to each another. In this study, we define a small, recombinant adenovirus serotype 3-derived protein, termed junction opener 1 (JO-1), which binds to the epithelial junction protein desmoglein 2 (DSG2). In mouse xenograft models employing Her2/neu- and EGFR-positive human cancer cell lines, JO-1 mediated cleavage of DSG2 dimers and activated intracellular signaling pathways which reduced E-cadherin expression in tight junctions. Notably, JO-1-triggered changes allowed for increased intratumoral penetration of the anti-Her2/neu mAb trastuzumab (Herceptin) and improved access to its target receptor, Her2/neu, which is partly trapped in tight junctions. This effect translated directly into increased therapeutic efficacy of trastuzumab in mouse xenograft models using breast, gastric, and ovarian cancer cells that were Her2/neu-positive. Furthermore, combining JO-1 with the EGFR-targeting mAb cetuximab (Erbitux) greatly improved therapeutic outcomes in a metastatic model of EGFR-positive lung cancer. A combination of JO-1 with an approach that triggered transient degradation of tumor stroma proteins elicited eradication of tumors. Taken together, our findings offer preclinical proof of concept to employ JO-1 in combination with mAb therapy.
Collapse
Affiliation(s)
- Ines Beyer
- Division of Medical Genetics, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|