Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 13: 1001122, 2022.
Article in English | MEDLINE | ID: mdl-36249782

ABSTRACT

Following injury the kidney undergoes a repair process, which results in replacement of the injured tissue with little evidence of damage. However, repetitive injuries or inability of the kidney to stop the repair process result in abnormal deposition of extracellular matrix (ECM) components leading to fibrosis and organ dysfunction. The synthesis/degradation of ECM components is finely regulated by several factors, including discoidin domain receptors (DDRs). These are receptor tyrosine kinases that are activated by collagens. Upon activation, DDRs control several cell functions that, when exacerbated, contribute to kidney injury and fibrosis. DDRs are undetectable in healthy kidney, but become rapidly upregulated in several kidney fibrotic conditions, thus making them attractive anti-fibrotic targets. DDRs contribute to kidney injury and fibrosis by promoting apoptosis of injured kidney cells, stimulating the production of pro-inflammatory cytokines, and regulating the production of ECM components. They achieve these effects by activating canonical intracellular molecules or by directly interacting with nuclear chromatin and promoting the transcription of pro-fibrotic genes. The goal of this review is to highlight canonical and non-canonical mechanisms whereby DDRs contribute to kidney injury/fibrosis. This review will summarize key findings obtained using cells and mice lacking DDRs and it will discuss the discovery and development of targeted DDR small molecule- and antisense-based inhibitors. Understanding the molecular mechanisms whereby DDRs control kidney injury and fibrosis might enable us to not only develop more selective and potent inhibitors, but to also determine when DDR inhibition needs to be achieved to prevent and/or halt the development of kidney fibrosis.

2.
Front Cell Dev Biol ; 10: 836797, 2022.
Article in English | MEDLINE | ID: mdl-35309920

ABSTRACT

Integrins and discoidin domain receptors (DDRs) 1 and 2 promote cell adhesion and migration on both fibrillar and non fibrillar collagens. Collagen I contains DDR and integrin selective binding motifs; however, the relative contribution of these two receptors in regulating cell migration is unclear. DDR1 has five isoforms (DDR1a-e), with most cells expressing the DDR1a and DDR1b isoforms. We show that human embryonic kidney 293 cells expressing DDR1b migrate more than DDR1a expressing cells on DDR selective substrata as well as on collagen I in vitro. In addition, DDR1b expressing cells show increased lung colonization after tail vein injection in nude mice. DDR1a and DDR1b differ from each other by an extra 37 amino acids in the DDR1b cytoplasmic domain. Interestingly, these 37 amino acids contain an NPxY motif which is a central control module within the cytoplasmic domain of ß integrins and acts by binding scaffold proteins, including talin. Using purified recombinant DDR1 cytoplasmic tail proteins, we show that DDR1b directly binds talin with higher affinity than DDR1a. In cells, DDR1b, but not DDR1a, colocalizes with talin and integrin ß1 to focal adhesions and enhances integrin ß1-mediated cell migration. Moreover, we show that DDR1b promotes cell migration by enhancing Rac1 activation. Mechanistically DDR1b interacts with the GTPase-activating protein (GAP) Breakpoint cluster region protein (BCR) thus reducing its GAP activity and enhancing Rac activation. Our study identifies DDR1b as a major driver of cell migration and talin and BCR as key players in the interplay between integrins and DDR1b in regulating cell migration.

3.
JCI Insight ; 7(3)2022 02 08.
Article in English | MEDLINE | ID: mdl-34941574

ABSTRACT

Discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase activated by collagen, contributes to chronic kidney disease. However, its role in acute kidney injury and subsequent development of kidney fibrosis is not clear. Thus, we performed a model of severe ischemia/reperfusion-induced acute kidney injury that progressed to kidney fibrosis in WT and Ddr1-null mice. We showed that Ddr1-null mice had reduced acute tubular injury, inflammation, and tubulointerstitial fibrosis with overall decreased renal monocyte chemoattractant protein (MCP-1) levels and STAT3 activation. We identified breakpoint cluster region (BCR) protein as a phosphorylated target of DDR1 that controls MCP-1 production in renal proximal tubule epithelial cells. DDR1-induced BCR phosphorylation or BCR downregulation increased MCP-1 secretion, suggesting that BCR negatively regulates the levels of MCP-1. Mechanistically, phosphorylation or downregulation of BCR increased ß-catenin activity and in turn MCP-1 production. Finally, we showed that DDR1-mediated STAT3 activation was required to stimulate the secretion of TGF-ß. Thus, DDR1 contributes to acute and chronic kidney injury by regulating BCR and STAT3 phosphorylation and in turn the production of MCP-1 and TGF-ß. These findings identify DDR1 an attractive therapeutic target for ameliorating both proinflammatory and profibrotic signaling in kidney disease.


Subject(s)
Discoidin Domain Receptor 1/genetics , Gene Expression Regulation , Inflammation/complications , Kidney Tubules, Proximal/metabolism , Proto-Oncogene Proteins c-bcr/genetics , RNA/genetics , STAT3 Transcription Factor/genetics , Acute Kidney Injury , Animals , Cell Line , Cells, Cultured , Discoidin Domain Receptor 1/biosynthesis , Female , Fibrosis/complications , Fibrosis/genetics , Fibrosis/pathology , Inflammation/genetics , Inflammation/pathology , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-bcr/biosynthesis , STAT3 Transcription Factor/biosynthesis , Signal Transduction
4.
ACS Med Chem Lett ; 11(1): 29-33, 2020 Jan 09.
Article in English | MEDLINE | ID: mdl-31938459

ABSTRACT

Herein, we report the discovery of a potent and selective dual DDR1/2 inhibitor, 7e (VU6015929), displaying low cytotoxicity, good kinome selectivity, and possessing an acceptable in vitro DMPK profile with good rodent in vivo pharmacokinetics. VU6015929 potently blocks collagen-induced DDR1 activation and collagen-IV production, suggesting DDR1 inhibition as an exciting target for antifibrotic therapy.

5.
J Am Soc Nephrol ; 30(9): 1605-1624, 2019 09.
Article in English | MEDLINE | ID: mdl-31383731

ABSTRACT

BACKGROUND: The discoidin domain receptor 1 (DDR1) is activated by collagens, upregulated in injured and fibrotic kidneys, and contributes to fibrosis by regulating extracellular matrix production, but how DDR1 controls fibrosis is poorly understood. DDR1 is a receptor tyrosine kinase (RTK). RTKs can translocate to the nucleus via a nuclear localization sequence (NLS) present on the receptor itself or a ligand it is bound to. In the nucleus, RTKs regulate gene expression by binding chromatin directly or by interacting with transcription factors. METHODS: To determine whether DDR1 translocates to the nucleus and whether this event is mediated by collagen-induced DDR1 activation, we generated renal cells expressing wild-type or mutant forms of DDR1 no longer able to bind collagen. Then, we determined the location of the DDR1 upon collagen stimulation. Using both biochemical assays and immunofluorescence, we analyzed the steps involved in DDR1 nuclear translocation. RESULTS: We show that although DDR1 and its natural ligand, collagen, lack an NLS, DDR1 is present in the nucleus of injured human and mouse kidney proximal tubules. We show that DDR1 nuclear translocation requires collagen-mediated receptor activation and interaction of DDR1 with SEC61B, a component of the Sec61 translocon, and nonmuscle myosin IIA and ß-actin. Once in the nucleus, DDR1 binds to chromatin to increase the transcription of collagen IV, a major collagen upregulated in fibrosis. CONCLUSIONS: These findings reveal a novel mechanism whereby activated DDR1 translates to the nucleus to regulate synthesis of profibrotic molecules.


Subject(s)
Collagen Type IV/genetics , Collagen Type I/metabolism , Discoidin Domain Receptor 1/metabolism , Kidney Tubules, Proximal/metabolism , Actins/metabolism , Acute Kidney Injury/metabolism , Animals , Biological Transport , Cell Line , Cell Nucleus , Chromatin/metabolism , Collagen Type I/pharmacology , Collagen Type IV/metabolism , Discoidin Domain Receptor 1/genetics , Humans , Kidney Tubules, Proximal/pathology , Male , Mice , Myosin Heavy Chains/metabolism , Nuclear Localization Signals , Retinoblastoma-Binding Protein 4/metabolism , SEC Translocation Channels/metabolism , Transcription, Genetic
7.
Matrix Biol ; 57-58: 258-271, 2017 01.
Article in English | MEDLINE | ID: mdl-27915093

ABSTRACT

Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase that binds to and is activated by collagens. DDR1 expression increases following kidney injury and accumulating evidence suggests that it contributes to the progression of injury. To this end, deletion of DDR1 is beneficial in ameliorating kidney injury induced by angiotensin infusion, unilateral ureteral obstruction, or nephrotoxic nephritis. Most of the beneficial effects observed in the DDR1-null mice are attributed to reduced inflammatory cell infiltration to the site of injury, suggesting that DDR1 plays a pro-inflammatory effect. The goal of this study was to determine whether, in addition to its pro-inflammatory effect, DDR1 plays a deleterious effect in kidney injury by directly regulating extracellular matrix production. We show that DDR1-null mice have reduced deposition of glomerular collagens I and IV as well as decreased proteinuria following the partial renal ablation model of kidney injury. Using mesangial cells isolated from DDR1-null mice, we show that these cells produce significantly less collagen compared to DDR1-null cells reconstituted with wild type DDR1. Moreover, mutagenesis analysis revealed that mutations in the collagen binding site or in the kinase domain significantly reduce DDR1-mediated collagen production. Finally, we provide evidence that blocking DDR1 kinase activity with an ATP-competitive small molecule inhibitor reduces collagen production. In conclusion, our studies indicate that the kinase activity of DDR1 plays a key role in DDR1-induced collagen synthesis and suggest that blocking collagen-mediated DDR1 activation may be beneficial in fibrotic diseases.


Subject(s)
Acute Kidney Injury/genetics , Collagen Type IV/genetics , Discoidin Domain Receptor 1/genetics , Kidney Glomerulus/metabolism , Nephritis/genetics , Ureteral Obstruction/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/surgery , Angiotensins , Animals , Binding Sites , Collagen Type IV/metabolism , Discoidin Domain Receptor 1/deficiency , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Kidney Glomerulus/pathology , Male , Mice , Mice, Knockout , Nephrectomy , Nephritis/chemically induced , Nephritis/metabolism , Nephritis/pathology , Protein Binding , Signal Transduction , Ureter/surgery , Ureteral Obstruction/pathology , Ureteral Obstruction/surgery
8.
Curr Top Membr ; 76: 231-53, 2015.
Article in English | MEDLINE | ID: mdl-26610916

ABSTRACT

Cell-extracellular matrix (ECM) interactions are essential for tissue development, homeostasis, and response to injury. Basement membranes (BMs) are specialized ECMs that separate epithelial or endothelial cells from stromal components and interact with cells via cellular receptors, including integrins and discoidin domain receptors. Disruption of cell-BM interactions due to either injury or genetic defects in either the ECM components or cellular receptors often lead to irreversible tissue injury and loss of organ function. Animal models that lack specific BM components or receptors either globally or in selective tissues have been used to help with our understanding of the molecular mechanisms whereby cell-BM interactions regulate organ function in physiological and pathological conditions. We review recently published works on animal models that explore how cell-BM interactions regulate kidney homeostasis in both health and disease.


Subject(s)
Basement Membrane/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney/cytology , Kidney/pathology , Receptors, Cell Surface/metabolism , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Kidney/metabolism , Protein Binding
9.
Drug Discov Today ; 20(2): 255-61, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25284748

ABSTRACT

Discoidin domain receptor (DDR) 1 and 2 are transmembrane receptors that belong to the family of receptor tyrosine kinases (RTK). Upon collagen binding, DDRs transduce cellular signaling involved in various cell functions, including cell adhesion, proliferation, differentiation, migration, and matrix homeostasis. Altered DDR function resulting from either mutations or overexpression has been implicated in several types of disease, including atherosclerosis, inflammation, cancer, and tissue fibrosis. Several established inhibitors, such as imatinib, dasatinib, and nilotinib, originally developed as Abelson murine leukemia (Abl) kinase inhibitors, have been found to inhibit DDR kinase activity. As we review here, recent discoveries of novel inhibitors and their co-crystal structure with the DDR1 kinase domain have made structure-based drug discovery for DDR1 amenable.


Subject(s)
Drug Discovery , Receptor Protein-Tyrosine Kinases/chemistry , Receptors, Mitogen/chemistry , Animals , Discoidin Domain Receptors , Humans , Molecular Structure , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Mitogen/antagonists & inhibitors
10.
Matrix Biol ; 34: 185-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24361528

ABSTRACT

Discoidin domain receptors, DDR1 and DDR2, lie at the intersection of two large receptor families, namely the extracellular matrix and tyrosine kinase receptors. As such, DDRs are uniquely positioned to function as sensors for extracellular matrix and to regulate a wide range of cell functions from migration and proliferation to cytokine secretion and extracellular matrix homeostasis/remodeling. While activation of DDRs by extracellular matrix collagens is required for normal development and tissue homeostasis, aberrant activation of these receptors following injury or in disease is detrimental. The availability of mice lacking DDRs has enabled us to identify key roles played by these receptors in disease initiation and progression. DDR1 promotes inflammation in atherosclerosis, lung fibrosis and kidney injury, while DDR2 contributes to osteoarthritis. Furthermore, both DDRs have been implicated in cancer progression. Yet the mechanisms whereby DDRs contribute to disease progression are poorly understood. In this review we highlight the mechanisms whereby DDRs regulate two important processes, namely inflammation and tissue fibrosis. In addition, we discuss the challenges of targeting DDRs in disease. Selective targeting of these receptors requires understanding of how they interact with and are activated by extracellular matrix, and whether their cellular function is dependent on or independent of receptor kinase activity.


Subject(s)
Fibrosis/genetics , Inflammation/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Animals , Cell Movement/genetics , Cell Proliferation , Discoidin Domain Receptor 1 , Discoidin Domain Receptors , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Fibrosis/pathology , Humans , Inflammation/pathology , Mice , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Signal Transduction/genetics
11.
J Am Soc Nephrol ; 23(6): 1027-38, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22440900

ABSTRACT

Mesangial cells and podocytes express integrins α1ß1 and α2ß1, which are the two major collagen receptors that regulate multiple cellular functions, including extracellular matrix homeostasis. Integrin α1ß1 protects from glomerular injury by negatively regulating collagen production, but the role of integrin α2ß1 in renal injury is unclear. Here, we subjected wild-type and integrin α2-null mice to injury with adriamycin or partial renal ablation. In both of these models, integrin α2-null mice developed significantly less proteinuria and glomerulosclerosis. In addition, selective pharmacological inhibition of integrin α2ß1 significantly reduced adriamycin-induced proteinuria, glomerular injury, and collagen deposition in wild-type mice. This inhibitor significantly reduced collagen synthesis in wild-type, but not integrin α2-null, mesangial cells in vitro, demonstrating that its effects are integrin α2ß1-dependent. Taken together, these results indicate that integrin α2ß1 contributes to glomerular injury by positively regulating collagen synthesis and suggest that its inhibition may be a promising strategy to reduce glomerular injury and proteinuria.


Subject(s)
Acute Kidney Injury/pathology , Doxorubicin/pharmacology , Integrin alpha2beta1/metabolism , Kidney Glomerulus/injuries , Acute Kidney Injury/metabolism , Albuminuria/physiopathology , Analysis of Variance , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Integrin alpha2beta1/drug effects , Kidney Function Tests , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Random Allocation , Receptors, Collagen/metabolism
12.
Exp Cell Res ; 318(9): 1001-10, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22417893

ABSTRACT

Glomerulosclerosis is characterized by excessive deposition of extracellular matrix within the glomeruli of the kidney, glomerular cell death, and subsequent loss of functional glomeruli. While in physiological situations the levels of extracellular matrix components are kept constant by a tight balance between formation and degradation, in the case of injury that results in fibrosis there is increased matrix deposition relative to its breakdown. Multiple factors control matrix synthesis and degradation, thus contributing to the development of glomerulosclerosis. This review focuses primarily on the role of cell-matrix interactions, which play a critical role in governing glomerular cell cues in both healthy and diseased kidneys. Cell-extracellular matrix interactions are made possible by various cellular receptors including integrins, discoidin domain receptors, and dystroglycan. Upon binding to a selective extracellular matrix protein, these receptors activate intracellular signaling pathways that can either downregulate or upregulate matrix synthesis and deposition. This, together with the observation that changes in the expression levels of matrix receptors have been documented in glomerular disease, clearly emphasizes the contribution of cell-matrix interactions in glomerular injury. Understanding the molecular mechanisms whereby extracellular matrix receptors regulate matrix homeostasis in the course of glomerular injury is therefore critical for devising more effective therapies to treat and ideally prevent glomerulosclerosis.


Subject(s)
Extracellular Matrix/metabolism , Glomerulonephritis/metabolism , Animals , Glomerulonephritis/pathology , Humans , Integrins/metabolism , Kidney/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology
13.
J Biol Chem ; 285(51): 40114-24, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20940300

ABSTRACT

Integrin α1ß1 is a collagen receptor that down-regulates collagen and reactive oxygen species (ROS) production, and mice lacking this receptor show increased ROS levels and exacerbated glomerular sclerosis following injury. Caveolin-1 (Cav-1) is a multifunctional protein that is tyrosine-phosphorylated in response to injury and has been implicated in ROS-mediated injury. Cav-1 interacts with integrins, and integrin α1ß1 binds/activates T cell protein-tyrosine phosphatase (TCPTP), which is homologous to the tyrosine phosphatase PTP1B known to dephosphorylate Cav-1. In this study, we analyzed whether phosphorylated Cav-1 (pCav-1) is a substrate of TCPTP and if integrin α1ß1 is essential for promoting TCPTP-mediated Cav-1 dephosphorylation. We found that Cav-1 phosphorylation is significantly higher in cells lacking integrin α1ß1 at base line and following oxidative stress. Overexpression of TCPTP leads to reduced pCav-1 levels only in cells expressing integrin α1ß1. Using solid phase binding assays, we demonstrated that 1) purified Cav-1 directly interacts with TCPTP and the integrin α1 subunit, 2) pCav-1 is a substrate of TCPTP, and 3) TCPTP-mediated Cav-1 dephosphorylation is highly increased by the addition of purified integrin α1ß1 or an integrin α1 cytoplasmic peptide to which TCPTP has been shown to bind. Thus, our results demonstrate that pCav-1 is a new substrate of TCPTP and that integrin α1ß1 acts as a negative regulator of Cav-1 phosphorylation by activating TCPTP. This could explain the protective function of integrin α1ß1 in oxidative stress-mediated damage and why integrin α1-null mice are more susceptible to fibrosis following injury.


Subject(s)
Caveolin 1/metabolism , Integrin alpha1beta1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Animals , CHO Cells , Caveolin 1/genetics , Collagen/genetics , Collagen/metabolism , Cricetinae , Cricetulus , Enzyme Activation/genetics , Fibrosis/genetics , HEK293 Cells , Humans , Integrin alpha1beta1/genetics , Mice , Mice, Mutant Strains , Oxidative Stress/genetics , Phosphorylation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Reactive Oxygen Species/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
J Am Soc Nephrol ; 19(4): 677-84, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18235087

ABSTRACT

Podocyte adhesion to the glomerular basement membrane is required for proper function of the glomerular filtration barrier. However, the mechanism whereby podocytes adhere to collagen IV networks, a major component of the glomerular basement membrane, is poorly understood. The predominant collagen IV network is composed of triple helical protomers containing the alpha3alpha4alpha5 chains. The protomers connect via the trimeric noncollagenous (NC1) domains to form hexamers at the interface. Because the NC1 domains of this network can potentially support integrin-dependent cell adhesion, it was determined whether individual NC1 monomers or alpha3alpha4alpha5 hexamers support podocyte adhesion. It was found that, although human podocytes did not adhere to NC1 domains proper, they did adhere via integrin alphavbeta3 to a KRGDS motif located adjacent to alpha3NC1 domains. Because the KRGDS motif is a site of phosphorylation, its interactions with integrin alphavbeta3 may play a critical role in cell signaling in physiologic and pathologic states.


Subject(s)
Collagen Type IV , Podocytes/physiology , Amino Acid Motifs , Animals , Cell Adhesion , Humans
15.
J Biol Chem ; 281(30): 20932-20939, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16731529

ABSTRACT

Exogenous soluble human alpha3 noncollagenous (NC1) domain of collagen IV inhibits angiogenesis and tumor growth. These biological functions are attributed to the binding of alpha3NC1 to integrin alphavbeta3. However, in some tumor cells that express integrin alphavbeta3, the alpha3NC1 domain does not inhibit proliferation, suggesting that integrin alphavbeta3 expression is not sufficient to mediate the anti-tumorigenic activity of this domain. Therefore, in the present study, we searched for novel binding receptors for the soluble alpha3NC1 domain in cells lacking alphavbeta3 integrin. In these cells, soluble alpha3NC1 bound integrin alpha3beta1; however, unlike alphavbeta3, alpha3beta1 integrin did not mediate cell adhesion to immobilized alpha3NC1 domain. Interestingly, in cells lacking integrin alpha3beta1, adhesion to the alpha3NC1 domain was enhanced due to activation of integrin alphavbeta3. These findings indicate that integrin alpha3beta1 is a receptor for the alpha3NC1 domain and transdominantly inhibits integrin alphavbeta3 activation. Thus integrin alpha3beta1, in conjunction with integrin alphavbeta3, modulates cellular responses to the alpha3NC1 domain, which may be pivotal in the mechanism underpinning its anti-angiogenic and anti-tumorigenic activities.


Subject(s)
Integrin alpha3beta1/physiology , Integrin alphaVbeta3/antagonists & inhibitors , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/metabolism , Gene Expression Regulation, Neoplastic , Humans , Integrin alphaVbeta3/chemistry , Integrins/metabolism , Neovascularization, Pathologic , Protein Structure, Tertiary , Recombinant Proteins/chemistry
16.
J Virol ; 79(17): 10923-30, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16103144

ABSTRACT

The core fusion machinery of all herpesviruses consists of three conserved glycoproteins, gB and gHgL, suggesting a common mechanism for virus cell fusion, but fusion of Epstein-Barr virus (EBV) with B cells and epithelial cells is initiated differently. Fusion with B cells requires a fourth protein, gp42, which complexes with gHgL and interacts with HLA class II, the B-cell coreceptor. Fusion with an epithelial cell does not require gp42 but requires interaction of gHgL with a novel epithelial cell coreceptor. Epithelial cell fusion can be inhibited by gp42 binding to gHgL and by antibodies to gH that fail to block B-cell fusion. This suggests that regions of gHgL initiating fusion with each cell are separable from each other and from regions involved in fusion itself. To address this possibility we mapped the region of gH recognized by a monoclonal antibody to gH that blocks EBV fusion with epithelial cells but not B cells by making a series of chimeras with the gH homolog of rhesus lymphocryptovirus. Proteins with mutations engineered within this region included those that preferentially mediate fusion with B cells, those that preferentially mediate fusion with epithelial cells, and those that mediate fusion with neither cell type. These results support the hypothesis that the core fusion function of gH is the same for B cells and epithelial cells and that it differs only in the way in which it is triggered into a functionally active state.


Subject(s)
B-Lymphocytes/virology , Epithelial Cells/virology , Herpesvirus 4, Human/physiology , Viral Fusion Proteins/physiology , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Glycoproteins/physiology , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Molecular Chaperones/genetics , Molecular Chaperones/physiology , Molecular Sequence Data , Point Mutation , Sequence Alignment , Viral Fusion Proteins/genetics , Viral Proteins/genetics , Viral Proteins/physiology , Virus Replication
17.
J Virol ; 78(10): 5007-14, 2004 May.
Article in English | MEDLINE | ID: mdl-15113881

ABSTRACT

Epstein-Barr virus (EBV) is a lymphotropic herpesvirus. However, access to B lymphocytes during primary infection may be facilitated by replication in mucosal epithelial cells. Attachment and penetration of EBV into these two cell types are fundamentally different. Both the distribution of receptors and the cellular origin of the virus impact the efficiency of infection. Epithelial cells potentially offer a wide range of receptors with which virus can interact. We report here on analyses of epithelial cells expressing different combinations of receptors. We find that the stoichiometry of the virus glycoprotein complex that includes gHgL and gp42 affects the use of gHgL not just for entry into epithelial cells but also for attachment. Penetration can be mediated efficiently with either a coreceptor for gp42 or gHgL, but the use of gHgL for attachment as well as penetration greatly compromises its ability to mediate entry.


Subject(s)
Herpesvirus 4, Human/physiology , Membrane Glycoproteins/physiology , Molecular Chaperones/physiology , Viral Envelope Proteins/physiology , Viral Proteins/physiology , Cell Line, Tumor , Epithelial Cells/virology , Glycoproteins/analysis , Glycoproteins/physiology , Histocompatibility Antigens Class II/physiology , Humans , Polyethylene Glycols/pharmacology , Receptors, Complement 3d/physiology , Receptors, Virus/physiology
18.
Nat Med ; 8(6): 594-9, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12042810

ABSTRACT

Epstein-Barr virus is ubiquitous and is causally implicated in lymphoid and epithelial malignancies. Virus invades oropharyngeal mucosa and establishes latency in B lymphocytes. Reactivating lymphocytes shed virus into saliva for spread to new hosts. A complex of three virus glycoproteins, gH, gL and gp42, is essential for entry. B-cell entry requires binding of gp42 to human leukocyte antigen (HLA) class II whereas entry into epithelial cells lacking HLA class II requires complexes without gp42. To accommodate infection of each, the virus carries both three-part and two-part complexes. We show here that HLA class II in the virus-producing cell alters the ratio of three-part to two-part complexes. As a consequence, virus originating in epithelial cells efficiently infects B cells whereas B-cell derived virus better infects epithelial cells. This molecular switch is a novel strategy that could alter tropism of virus from epithelium to B cells and then back to epithelium in a new host.


Subject(s)
B-Lymphocytes/virology , Epithelial Cells/virology , Herpesvirus 4, Human/physiology , Virus Replication/physiology , Animals , B-Lymphocytes/immunology , Cell Line , DNA, Viral/analysis , Epithelial Cells/immunology , Gene Deletion , Herpesvirus 4, Human/genetics , Histocompatibility Antigens Class II/analysis , Lymphocyte Activation , Thymidine Kinase/deficiency , Thymidine Kinase/genetics , Virus Shedding
SELECTION OF CITATIONS
SEARCH DETAIL
...