Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Br J Dermatol ; 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38878280

ABSTRACT

BACKGROUND: The structured expression of several keratins in the skin is associated with differentiation status of the epidermal layers, whereas others are upregulated only during wound healing, in skin disorders and in cancers. One of these stress keratins, K17, is correlated with poor prognosis in various cancer types and its loss has been shown to decelerate tumour growth. K17 expression can also be detected in cutaneous squamous cell carcinomas (SCCs), where UV-irradiation and infection with cutaneous human papillomaviruses (HPVs) are important co-factors. It was previously reported that K17 is upregulated in papillomavirus (PV)-induced benign skin lesions in mice and induces an immunological status that is beneficial for tumour growth. OBJECTIVES: In order to investigate whether K17 upregulation is induced by PVs, we analysed K17 levels in skin tumour specimens of different animal models and humans. METHODS: Various immunofluorescence stainings were performed to identify K17 expression as well as levels of E-Cadherin, vimentin and CD271. Tissues were further analysed by PCRs, qPCRs and ELISA to control for PV activity. K17knockdown cells were generated and effects on viral life cycle were investigated by infection assays, qPCR and Western blotting. RESULTS: We could show that K17 is commonly expressed in skin tumours and that its presence is not directly linked to viral oncoprotein expression. Rather, K17 expression seems to be a marker of epithelial differentiation and its absence in tumour tissue is associated with an epithelial-to-mesenchymal transition. We further showed that the absence of K17 in skin tumours increases markers of cancer stem-like cells and negatively affects viral protein synthesis. CONCLUSIONS: Collectively, our data indicate that K17 expression is a common feature in skin tumourigenesis. While it is not primarily targeted by PV oncoproteins, our in vivo and in vitro data suggest that it is an important regulator of epithelial differentiation and thus may play a role in controlling viral protein synthesis.

2.
J Med Virol ; 95(6): e28850, 2023 06.
Article in English | MEDLINE | ID: mdl-37322807

ABSTRACT

Infection with certain cutaneous human papillomaviruses (HPV), in conjunction with chronic ultraviolet (UV) exposure, are the major cofactors of non-melanoma skin cancer (NMSC), the most frequent cancer type worldwide. Cutaneous squamous cell carcinomas (SCCs) as well as tumors in general represent three-dimensional entities determined by both temporal and spatial constraints. Whole tissue proteomics is a straightforward approach to understand tumorigenesis in better detail, but studies focusing on different progression states toward a dedifferentiated SCC phenotype on a spatial level are rare. Here, we applied an innovative proteomic workflow on formalin-fixed, paraffin-embedded (FFPE) epithelial tumors derived from the preclinical animal model Mastomys coucha. This rodent is naturally infected with its genuine cutaneous papillomavirus and closely mimics skin carcinogenesis in the context of cutaneous HPV infections in humans. We deciphered cellular networks by comparing diverse epithelial tissues with respect to their differentiation level and infection status. Our study reveals novel regulatory proteins and pathways associated with virus-induced tumor initiation and progression of SCCs. This approach provides the basis to better comprehend the multistep process of skin carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell , Papillomavirus Infections , Skin Neoplasms , Animals , Humans , Proteomics , Papillomaviridae/genetics , Murinae , Keratinocytes , Carcinogenesis
3.
J Chem Inf Model ; 63(12): 3878-3891, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37310029

ABSTRACT

Integrins are a family of α/ß heterodimeric cell surface adhesion receptors which are capable of transmitting signals bidirectionally across membranes. They are known for their therapeutic potential in a wide range of diseases. However, the development of integrin-targeting medications has been impacted by unexpected downstream effects including unwanted agonist-like effects. Allosteric modulation of integrins is a promising approach to potentially overcome these limitations. Applying mixed-solvent molecular dynamics (MD) simulations to integrins, the current study uncovers hitherto unknown allosteric sites within the integrin α I domains of LFA-1 (αLß2; CD11a/CD18), VLA-1 (α1ß1; CD49a/CD29), and Mac-1 (αMß2, CD11b/CD18). We show that these pockets are putatively accessible to small-molecule modulators. The findings reported here may provide opportunities for the design of novel allosteric integrin inhibitors lacking the unwanted agonism observed with earlier as well as current integrin-targeting drugs.


Subject(s)
CD18 Antigens , Molecular Dynamics Simulation , CD18 Antigens/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/metabolism , Receptors, Cell Surface
4.
Biochem Pharmacol ; 211: 115504, 2023 05.
Article in English | MEDLINE | ID: mdl-36921634

ABSTRACT

Integrins are a family of cell surface receptors well-recognized for their therapeutic potential in a wide range of diseases. However, the development of integrin targeting medications has been impacted by unexpected downstream effects, reflecting originally unforeseen interference with the bidirectional signalling and cross-communication of integrins. We here selected one of the most severely affected target integrins, the integrin lymphocyte function-associated antigen-1 (LFA-1, αLß2, CD11a/CD18), as a prototypic integrin to systematically assess and overcome these known shortcomings. We employed a two-tiered ligand-based virtual screening approach to identify a novel class of allosteric small molecule inhibitors targeting this integrin's αI domain. The newly discovered chemical scaffold was derivatized, yielding potent bis-and tris-aryl-bicyclic-succinimides which inhibit LFA-1 in vitro at low nanomolar concentrations. The characterisation of these compounds in comparison to earlier LFA-1 targeting modalities established that the allosteric LFA-1 inhibitors (i) are devoid of partial agonism, (ii) selectively bind LFA-1 versus other integrins, (iii) do not trigger internalization of LFA-1 itself or other integrins and (iv) display oral availability. This profile differentiates the new generation of allosteric LFA-1 inhibitors from previous ligand mimetic-based LFA-1 inhibitors and anti-LFA-1 antibodies, and is projected to support novel immune regulatory regimens selectively targeting the integrin LFA-1. The rigorous computational and experimental assessment schedule described here is designed to be adaptable to the preclinical discovery and development of novel allosterically acting compounds targeting integrins other than LFA-1, providing an exemplary approach for the early characterisation of next generation integrin inhibitors.


Subject(s)
Lymphocyte Function-Associated Antigen-1 , Signal Transduction , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Ligands , Intercellular Adhesion Molecule-1/metabolism
5.
Neuro Oncol ; 25(6): 1031-1043, 2023 06 02.
Article in English | MEDLINE | ID: mdl-36215168

ABSTRACT

BACKGROUND: IDH mutant gliomas are grouped into astrocytomas or oligodendrogliomas depending on the codeletion of chromosome arms 1p and 19q. Although the genomic alterations of IDH mutant gliomas have been well described, transcriptional changes unique to either tumor type have not been fully understood. Here, we identify Tripartite Motif Containing 67 (TRIM67), an E3 ubiquitin ligase with essential roles during neuronal development, as an oncogene distinctly upregulated in oligodendrogliomas. METHODS: We used several cell lines, including patient-derived oligodendroglioma tumorspheres, to knock down or overexpress TRIM67. We coupled high-throughput assays, including RNA sequencing, total lysate-mass spectrometry (MS), and coimmunoprecipitation (co-IP)-MS with functional assays including immunofluorescence (IF) staining, co-IP, and western blotting (WB) to assess the in vitro phenotype associated with TRIM67. Patient-derived oligodendroglioma tumorspheres were orthotopically implanted in mice to determine the effect of TRIM67 on tumor growth and survival. RESULTS: TRIM67 overexpression alters the abundance of cytoskeletal proteins and induces membrane bleb formation. TRIM67-associated blebbing was reverted with the nonmuscle class II myosin inhibitor blebbistatin and selective ROCK inhibitor fasudil. NOGO-A/Rho GTPase/ROCK2 signaling is altered upon TRIM67 ectopic expression, pointing to the underlying mechanism for TRIM67-induced blebbing. Phenotypically, TRIM67 expression resulted in higher cell motility and reduced cell adherence. In orthotopic implantation models of patient-derived oligodendrogliomas, TRIM67 accelerated tumor growth, reduced overall survival, and led to increased vimentin expression at the tumor margin. CONCLUSIONS: Taken together, our results demonstrate that upregulated TRIM67 induces blebbing-based rounded cell morphology through Rho GTPase/ROCK-mediated signaling thereby contributing to glioma pathogenesis.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Oligodendroglioma , Animals , Mice , Humans , Oligodendroglioma/genetics , Nogo Proteins/genetics , Glioma/pathology , Astrocytoma/genetics , Cell Transformation, Neoplastic , Carcinogenesis , Chromosomes, Human, Pair 1 , Brain Neoplasms/pathology , Chromosomes, Human, Pair 19 , Isocitrate Dehydrogenase/genetics , Mutation , Tripartite Motif Proteins/genetics , Cytoskeletal Proteins/genetics
6.
Front Immunol ; 13: 1010790, 2022.
Article in English | MEDLINE | ID: mdl-36263027

ABSTRACT

Licensed L1-VLP-based immunizations against high-risk mucosal human papillomavirus (HPV) types have been a great success in reducing anogenital cancers, although they are limited in their cross-protection against HPV types not covered by the vaccine. Further, their utility in protection against cutaneous HPV types, of which some contribute to non-melanoma skin cancer (NMSC) development, is rather low. Next generation vaccines achieve broadly cross-protective immunity against highly conserved sequences of L2. In this exploratory study, we tested two novel HPV vaccine candidates, HPV16 RG1-VLP and CUT-PANHPVAX, in the preclinical natural infection model Mastomys coucha. After immunization with either vaccines, a mock control or MnPV L1-VLPs, the animals were experimentally infected and monitored. Besides vaccine-specific seroconversion against HPV L2 peptides, the animals also developed cross-reactive antibodies against the cutaneous Mastomys natalensis papillomavirus (MnPV) L2, which were cross-neutralizing MnPV pseudovirions in vitro. Further, both L2-based vaccines also conferred in vivo protection as the viral loads in plucked hair after experimental infection were lower compared to mock-vaccinated control animals. Importantly, the formation of neutralizing antibodies, whether directed against L1-VLPs or L2, was able to prevent skin tumor formation and even microscopical signs of MnPV infection in the skin. For the first time, our study shows the proof-of-principle of next generation L2-based vaccines even across different PV genera in an infection animal model with its genuine PV. It provides fundamental insights into the humoral immunity elicited by L2-based vaccines against PV-induced skin tumors, with important implications to the design of next generation HPV vaccines.


Subject(s)
Neoplasms , Oncogene Proteins, Viral , Papillomavirus Infections , Papillomavirus Vaccines , Vaccines, Virus-Like Particle , Mice , Animals , Humans , Neutralization Tests , Capsid Proteins , Mice, Inbred BALB C , Papillomaviridae , Antibodies, Neutralizing , Peptides
7.
Front Immunol ; 13: 811094, 2022.
Article in English | MEDLINE | ID: mdl-35359995

ABSTRACT

Notably, the majority of papillomaviruses associated with a high cancer risk have the potential to translate different isoforms of the L1 major capsid protein. In an infection model, the cutaneous Mastomys natalensis papillomavirus (MnPV) circumvents the humoral immune response of its natural host by first expressing a 30 amino acid extended L1 isoform (L1LONG). Although inducing a robust seroconversion, the raised antibodies are not neutralizing in vitro. In contrast, neutralizing antibodies induced by the capsid-forming isoform (L1SHORT) appear delayed by several months. We now provide evidence that, although L1LONG vaccination showed a strong seroconversion, these antibodies were not protective. As a consequence, virus-free animals subsequently infected with MnPV still accumulated high numbers of transcriptionally active viral genomes, ultimately leading to skin tumor formation. In contrast, vaccination with L1SHORT was completely protective. This shows that papillomavirus L1LONG expression is a unique strategy to escape from antiviral immune surveillance.


Subject(s)
Papillomavirus Infections , Skin Neoplasms , Animals , Capsid Proteins , Papillomaviridae , Protein Isoforms
8.
Sci Adv ; 8(12): eabh4050, 2022 03 25.
Article in English | MEDLINE | ID: mdl-35319989

ABSTRACT

Radiotherapy is a mainstay cancer therapy whose antitumor effects partially depend on T cell responses. However, the role of Natural Killer (NK) cells in radiotherapy remains unclear. Here, using a reverse translational approach, we show a central role of NK cells in the radiation-induced immune response involving a CXCL8/IL-8-dependent mechanism. In a randomized controlled pancreatic cancer trial, CXCL8 increased under radiotherapy, and NK cell positively correlated with prolonged overall survival. Accordingly, NK cells preferentially infiltrated irradiated pancreatic tumors and exhibited CD56dim-like cytotoxic transcriptomic states. In experimental models, NF-κB and mTOR orchestrated radiation-induced CXCL8 secretion from tumor cells with senescence features causing directional migration of CD56dim NK cells, thus linking senescence-associated CXCL8 release to innate immune surveillance of human tumors. Moreover, combined high-dose radiotherapy and adoptive NK cell transfer improved tumor control over monotherapies in xenografted mice, suggesting NK cells combined with radiotherapy as a rational cancer treatment strategy.


Subject(s)
Interleukin-8 , Killer Cells, Natural , Neoplasms , Adoptive Transfer , Animals , Humans , Immunity , Interleukin-8/immunology , Interleukin-8/metabolism , Killer Cells, Natural/immunology , Mice , Neoplasms/immunology , Neoplasms/radiotherapy , Xenograft Model Antitumor Assays
10.
Br J Pharmacol ; 177(12): 2696-2711, 2020 06.
Article in English | MEDLINE | ID: mdl-31985813

ABSTRACT

BACKGROUND AND PURPOSE: Antibodies targeting cell surface receptors are considered to enable highly selective therapeutic interventions for immune disorders and cancer. Their biological profiles are found, generally, to represent the net effects of antibody-target interactions. The former therapeutic anti-integrin αLß2 antibody efalizumab seems to defeat this paradigm by eliciting, via mechanisms currently unknown, much broader effects than would be predicted based on its target specificity. EXPERIMENTAL APPROACH: To elucidate the mechanisms behind these broad effects, we investigated in primary human lymphocytes in vitro the effects of anti-αLß2 antibodies on the expression of αLß2 as well as unrelated α4 integrins, in comparison to Fab fragments and small-molecule inhibitors. KEY RESULTS: We demonstrate that anti-αLß2 mAbs directly induce the internalization of α4 integrins. The endocytotic phenomenon is a direct consequence of their antibody nature. It is inhibited when monovalent Fab fragments or small-molecule inhibitors are used. It is independent of crosslinking via anti-Fc mAbs and of αLß2 activation. The cross-modulatory effect is unidirectional and not observed in a similar fashion with the α4 integrin antibody natalizumab. CONCLUSION AND IMPLICATIONS: The present study identifies endocytotic cross-modulation as a hitherto unknown non-canonical functionality of anti-αLß2 antibodies. This cross-modulation has the potential to fundamentally alter an antibody's benefit risk profile, as evident with efalizumab. The newly described phenomenon may be of relevance to other therapeutic antibodies targeting cluster-forming receptors. Thus, pharmacologists should be cognizant of this action when investigating such antibodies.


Subject(s)
Antibodies, Monoclonal , Lymphocyte Function-Associated Antigen-1 , Antibodies, Monoclonal/pharmacology , Humans , Immunoglobulin Fab Fragments , Receptors, Cell Surface
12.
13.
Biochem Pharmacol ; 119: 42-55, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27613223

ABSTRACT

The integrin leucocyte function-associated antigen-1 (αLß2, LFA-1) plays crucial roles in T cell adhesion, migration and immunological synapse (IS) formation. Consequently, αLß2 is an important therapeutic target in autoimmunity. Three major classes of αLß2 inhibitors with distinct modes of action have been described to date: Monoclonal antibodies (mAbs), small molecule α/ß I allosteric and small molecule α I allosteric inhibitors. The objective of this study was to systematically compare these three modes of αLß2 inhibition for their αLß2 inhibitory as well as their potential agonist-like effects. All inhibitors assessed were found to potently block αLß2-mediated leucocyte adhesion. None of the inhibitors induced ZAP70 phosphorylation, indicating absence of agonistic outside-in signalling. Paradoxically, however, the α/ß I allosteric inhibitor XVA143 induced conformational changes within αLß2 characteristic for an intermediate affinity state. This effect was not observed with the α I allosteric inhibitor LFA878 or the anti-αLß2 mAb efalizumab. On the other hand, efalizumab triggered the unscheduled internalization of αLß2 in CD4+ and CD8+ T cells while LFA878 and XVA143 did not affect or only mildly reduced αLß2 surface expression, respectively. Moreover, efalizumab, in contrast to the small molecule inhibitors, disturbed the fine-tuned internalization/recycling of engaged TCR/CD3, concomitantly decreasing ZAP70 expression levels. In conclusion, different modes of αLß2 inhibition are associated with fundamentally different biologic effect profiles. The differential established here is expected to provide important translational guidance as novel αLß2 inhibitors will be advanced from bench to bedside.


Subject(s)
Antibodies, Monoclonal/pharmacology , Integrins/antagonists & inhibitors , Lymphocyte Function-Associated Antigen-1/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Humanized , Cell Adhesion , Cell Line , Gene Expression Regulation/drug effects , Humans , Integrins/agonists , Mice
14.
Chimia (Aarau) ; 70(12): 864-867, 2016 Dec 21.
Article in English | MEDLINE | ID: mdl-28661358

ABSTRACT

Experimental pharmacology is undergoing fundamental changes. This article describes the challenges and opportunities associated with these changes from the perspective of the Swiss Society of Pharmacology (SSEP), the society which aims to advance experimental pharmacology in Switzerland and abroad.


Subject(s)
Pharmacology , Societies, Scientific/organization & administration , Societies, Scientific/trends , History, 20th Century , Humans , Pharmacology/history , Pharmacology/trends , Societies, Scientific/history , Switzerland
15.
Ger Med Sci ; 13: Doc23, 2015.
Article in English | MEDLINE | ID: mdl-26693218

ABSTRACT

This article addresses the question of when a trial master file (TMF) can be considered sufficiently accurate and complete: What attributes does the TMF need to have so that a clinical trial can be adequately reconstructed from documented data and procedures? Clinical trial sponsors face significant challenges in assembling the TMF, especially when dealing with large, international, multicenter studies; despite all newly introduced archiving techniques it is becoming more and more difficult to ensure that the TMF is complete. This is directly reflected in the number of inspection findings reported and published by the EMA in 2014. Based on quality risk management principles in clinical trials the authors defined the quality expectations for the different document types in a TMF and furthermore defined tolerance limits for missing documents. This publication provides guidance on what type of documents and processes are most important, and in consequence, indicates on which documents and processes trial team staff should focus in order to achieve a high-quality TMF. The members of this working group belong to the CQAG Group (Clinical Quality Assurance Germany) and are QA (quality assurance) experts (auditors or compliance functions) with long-term experience in the practical handling of TMFs.


Subject(s)
Clinical Trials as Topic/methods , Data Curation , Filing , Records/standards , Risk Management/organization & administration , Data Accuracy , Filing/methods , Filing/standards , Guidelines as Topic , Humans , Quality Improvement/organization & administration
16.
Br J Pharmacol ; 172(20): 4875-87, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26224111

ABSTRACT

BACKGROUND AND PURPOSE: The integrin αLß2 plays central roles in leukocyte adhesion and T cell activation, rendering αLß2 an attractive therapeutic target. Compounds with different modes of αLß2 inhibition are in development, currently. Consequently, there is a foreseeable need for bedside assays, which allow assessment of the different effects of diverse types of αLß2 inhibitors in the peripheral blood of treated patients. EXPERIMENTAL APPROACH: Here, we describe a flow cytometry-based technology that simultaneously quantitates αLß2 conformational change upon inhibitor binding, αLß2 expression and T cell activation at the single-cell level in human blood. Two classes of allosteric low MW inhibitors, designated α I and α/ß I allosteric αLß2 inhibitors, were investigated. The first application revealed intriguing inhibitor class-specific profiles. KEY RESULTS: Half-maximal inhibition of T cell activation was associated with 80% epitope loss induced by α I allosteric inhibitors and with 40% epitope gain induced by α/ß I allosteric inhibitors. This differential establishes that inhibitor-induced αLß2 epitope changes do not directly predict the effect on T cell activation. Moreover, we show here for the first time that α/ß I allosteric inhibitors, in contrast to α I allosteric inhibitors, provoked partial downmodulation of αLß2, revealing a novel property of this inhibitor class. CONCLUSIONS AND IMPLICATIONS: The multi-parameter whole blood αLß2 assay described here may enable therapeutic monitoring of αLß2 inhibitors in patients' blood. The assay dissects differential effect profiles of different classes of αLß2 inhibitors.


Subject(s)
Lymphocyte Function-Associated Antigen-1/blood , Naphthalenes/pharmacology , Oxazines/pharmacology , Phthalic Acids/pharmacology , beta-Alanine/analogs & derivatives , Biological Assay , Epitopes/immunology , Flow Cytometry , Humans , Lymphocyte Activation , T-Lymphocytes/immunology , beta-Alanine/pharmacology
17.
Methods Mol Biol ; 757: 15-30, 2012.
Article in English | MEDLINE | ID: mdl-21909903

ABSTRACT

Standard adhesion assays measure cell binding either to immobilized ligands or to cell monolayers in flat-well microtiter plates under static conditions. Typically, these test systems require several washing steps to separate adherent from nonadherent cells. Here, we describe an adhesion assay which avoids these washing steps by employing V-bottom 96-well plates. In this assay, fluorescently labeled leukocytes are allowed to adhere to V-well plates coated with soluble ligand for a fixed time. Thereafter, centrifugal force is applied to separate adherent cells from nonadherent cells. Nonadherent cells accumulate in the nadir of the V-shaped wells and are quantified using a fluorometer with a narrow aperture. This simple and reproducible method has been validated with different classes of adhesion molecule families (selectins and integrins) and is adaptable to several other adhesive interactions. The assay format is suitable for screening applications and may also be used for diagnostic testing. The receptor/ligand interaction chosen as an example to describe the assay methodology is the interaction between the integrin lymphocyte function-associated molecule-1 (LFA-1, α(L)ß(2)) and intercellular adhesion molecule-1 (ICAM-1).


Subject(s)
Cell Adhesion/physiology , Fluorometry , Cell Adhesion/drug effects , Cells, Cultured , Dimethyl Sulfoxide/pharmacology , Humans , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , Leukocytes/metabolism , Leukocytes, Mononuclear/metabolism , Ligands , Lymphocyte Function-Associated Antigen-1/metabolism , Protein Binding , Staining and Labeling
18.
J Biol Chem ; 286(49): 42115-42122, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-21965670

ABSTRACT

The activation of α/ß heterodimeric integrins is the result of highly coordinated rearrangements within both subunits. The molecular interactions between the two subunits, however, remain to be characterized. In this study, we use the integrin α(L)ß(2) to investigate the functional role of the C-linker polypeptide that connects the C-terminal end of the inserted (I) domain with the ß-propeller domain on the α subunit and is located at the interface with the ßI domain of the ß chain. We demonstrate that shortening of the C-linker by eight or more amino acids results in constitutively active α(L)ß(2) in which the αI domain is no longer responsive to the regulation by the ßI domain. Despite this intersubunit uncoupling, both I domains remain individually sensitive to intrasubunit conformational changes induced by allosteric modulators. Interestingly, the length and not the sequence of the C-linker appears to be critical for its functionality in α/ß intersubunit communication. Using two monoclonal antibodies (R7.1 and CBR LFA-1/1) we further demonstrate that shortening of the C-linker results in the gradual loss of combinational epitopes that require both the αI and ß-propeller domains for full reactivity. Taken together, our findings highlight the role of the C-linker as a spring-like element that allows relaxation of the αI domain in the resting state and controlled tension of the αI domain during activation, exerted by the ß chain.


Subject(s)
Integrin alpha1/chemistry , Allosteric Site , Animals , Antibodies, Monoclonal/chemistry , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Dimerization , Humans , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Microscopy, Fluorescence/methods , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Transfection
19.
J Clin Endocrinol Metab ; 95(5): 2316-24, 2010 May.
Article in English | MEDLINE | ID: mdl-20357178

ABSTRACT

BACKGROUND: Mutations in the IGF-I receptor (IGF1R) gene can be responsible for intrauterine and postnatal growth disorders. OBJECTIVE: Here we report on a novel mutation in the IGF1R gene in a female patient. The aim of our study was to analyze the functional impact of this mutation. PATIENT: At birth, the girl's length was 47 cm [-1.82 sd score (SDS)], and her weight was 2250 g (-2.26 SDS). Clinical examination revealed microcephaly and retarded cognitive development. She showed no postnatal catch-up growth but had relatively high IGF-I levels (+1.83 to +2.17 SDS). RESULTS: Denaturing HPLC screening and direct DNA sequencing disclosed a heterozygous missense mutation resulting in an amino acid exchange from valine to glutamic acid at position 599 (V599E-IGF1R). Using various cell systems, we found that the V599E-IGF1R mutant was not tyrosine phosphorylated and had an impaired downstream signaling in the presence of IGF-I. Flow cytometry and live cell confocal laser scanning microscopy revealed a lack of cell surface expression due to an extensive retention of V599E-IGF1R proteins within the endoplasmic reticulum. CONCLUSION: The V599E-IGF1R mutation interferes with the receptor's trafficking path, thereby abrogating proreceptor processing and plasma membrane localization. Diminished cell surface receptor density solely expressed from the patient's wild-type allele is supposed to lead to insufficient IGF-I signaling. We hypothesize that this mechanism results in intrauterine and postnatal growth retardation of the affected patient. The reported retention of the nascent IGF1R in the endoplasmic reticulum presents a novel mechanism of IGF-I resistance.


Subject(s)
Endoplasmic Reticulum/metabolism , Fetal Growth Retardation/genetics , Growth Disorders/genetics , Heterozygote , Mutation, Missense , Receptor, IGF Type 1/genetics , Amino Acid Substitution , Birth Weight , Body Size , Cognition Disorders/genetics , Exons/genetics , Female , Glutamic Acid/genetics , Humans , Infant, Newborn , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Intellectual Disability/genetics , Microcephaly/genetics , Polymerase Chain Reaction , Receptor, IGF Type 1/metabolism , Valine/genetics
20.
J Pharmacol Exp Ther ; 330(3): 792-801, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19491325

ABSTRACT

There is a pressing need for immunosuppressants with an improved safety profile. The search for novel approaches to blocking T-cell activation led to the development of the selective protein kinase C (PKC) inhibitor AEB071 (sotrastaurin). In cell-free kinase assays AEB071 inhibited PKC, with K(i) values in the subnanomolar to low nanomolar range. Upon T-cell stimulation, AEB071 markedly inhibited in situ PKC catalytic activity and selectively affected both the canonical nuclear factor-kappaB and nuclear factor of activated T cells (but not activator protein-1) transactivation pathways. In primary human and mouse T cells, AEB071 treatment effectively abrogated at low nanomolar concentration markers of early T-cell activation, such as interleukin-2 secretion and CD25 expression. Accordingly, the CD3/CD28 antibody- and alloantigen-induced T-cell proliferation responses were potently inhibited by AEB071 in the absence of nonspecific antiproliferative effects. Unlike former PKC inhibitors, AEB071 did not enhance apoptosis of murine T-cell blasts in a model of activation-induced cell death. Furthermore, AEB071 markedly inhibited lymphocyte function-associated antigen-1-mediated T-cell adhesion at nanomolar concentrations. The mode of action of AEB071 is different from that of calcineurin inhibitors, and AEB071 and cyclosporine A seem to have complementary effects on T-cell signaling pathways.


Subject(s)
Immunosuppressive Agents/pharmacology , Macrophage Activation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrroles/antagonists & inhibitors , Quinazolines/antagonists & inhibitors , T-Lymphocytes/drug effects , Animals , CD28 Antigens/metabolism , Calcium/metabolism , Cell Adhesion/drug effects , Cytokines/biosynthesis , Electrophoretic Mobility Shift Assay , Flow Cytometry , Genes, Reporter , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Jurkat Cells , Mice , Mice, Knockout , NF-kappa B/drug effects , NFATC Transcription Factors/metabolism , Protein Kinase C/genetics , Receptors, Antigen, T-Cell/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...