Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Signal ; 72: 109638, 2020 08.
Article in English | MEDLINE | ID: mdl-32320860

ABSTRACT

Glioblastoma (GBM) is an aggressive and devastating primary brain cancer which responds very poorly to treatment. The average survival time of patients is only 14-15 months from diagnosis so there is a clear and unmet need for the development of novel targeted therapies to improve patient outcomes. The multifunctional cytokine TGFß plays fundamental roles in development, adult tissue homeostasis, tissue wound repair and immune responses. Dysfunction of TGFß signalling has been implicated in both the development and progression of many tumour types including GBM, thereby potentially providing an actionable target for its treatment. This review will examine TGFß signalling mechanisms and their role in the development and progression of GBM. The targeting of TGFß signalling using a variety of approaches including the TGFß binding protein Decorin will be highlighted as attractive therapeutic strategies.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Brain Neoplasms/drug therapy , Decorin/metabolism , Glioblastoma/drug therapy , Humans , Tumor Microenvironment
2.
Am J Med Genet A ; 179(8): 1652-1664, 2019 08.
Article in English | MEDLINE | ID: mdl-31218820

ABSTRACT

Frank-Ter Haar syndrome (FTHS), Winchester syndrome (WS), and multicentric osteolysis, nodulosis, and arthropathy (MONA) are ultra-rare multisystem disorders characterized by craniofacial malformations, reduced bone density, skeletal and cardiac anomalies, and dermal fibrosis. These autosomal recessive syndromes are caused by homozygous mutation or deletion of respectively SH3PXD2B (SH3 and PX Domains 2B), MMP14 (matrix metalloproteinase 14), or MMP2. Here, we give an overview of the clinical features of 63 previously reported patients with an SH3PXD2B, MMP14, or MMP2 mutation, demonstrating considerable clinical overlap between FTHS, WS, and MONA. Interestingly, the protein products of SH3PXD2B, MMP14, and MMP2 directly cooperate in collagen remodeling. We review animal models for these three disorders that accurately reflect the major clinical features and likewise show significant phenotypical similarity with each other. Furthermore, they demonstrate that defective collagen remodeling is central in the underlying pathology. As such, we propose a nosological revision, placing these SH3PXD2B, MMP14, and MMP2 related syndromes in a novel "defective collagen-remodelling spectrum (DECORS)". In our opinion, this revised nosology better reflects the central role for impaired collagen remodeling, a potential target for pharmaceutical intervention.


Subject(s)
Collagen/genetics , Hajdu-Cheney Syndrome/diagnosis , Hajdu-Cheney Syndrome/genetics , Mutation , Phenotype , Alleles , Animals , Collagen/chemistry , Gene Knockdown Techniques , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism
3.
Exp Dermatol ; 28(10): 1106-1113, 2019 10.
Article in English | MEDLINE | ID: mdl-29570224

ABSTRACT

Erythrokeratodermia variabilis et progressiva (EKV-P) is caused by mutations in either the GJB3 (Cx31) or GJB4 genes (Cx30.3). We identified a rare GJB3 missense mutation, c.134G>A (p.G45E), in two unrelated patients and investigated its cellular characteristics. Expression of Cx31G45E-GFP caused previously undescribed changes within HeLa cells and HaCaT cells, a model human keratinocyte cell line. Cx31WT-GFP localised to the plasma membrane, but expression of Cx31G45E-GFP caused vacuolar expansion of the endoplasmic reticulum (ER), the mutant protein accumulated within the ER membrane and disassembly of the microtubular network occurred. No ER stress responses were evoked. Cx31WT-myc-myc-6xHis and Cx31G45E-GFP co-immunoprecipitated, indicative of heteromeric interaction, but co-expression with Cx31WT-mCherry, Cx26 or Cx30.3 did not mitigate the phenotype. Cx31 and Cx31G45E both co-immunoprecipitated with Cx43, indicating the ability to form heteromeric connexons. WT-Cx31 and Cx43 assembled into large gap junction plaques at points of cell-to-cell contact; Cx31G45E restricted the ability of Cx43 to reach the plasma membrane in both HaCaT cells and HeLa cells stably expressing Cx43 where the proteins strongly co-localised with the vacolourised ER. Cell viability assays identified an increase in cell death in cells expressing Cx31G45E-GFP, which FACS analysis determined was necrotic. Blocking connexin channel function with 18α-glycyrrhetinic acid did not completely rescue necrosis or prevent propidium iodide uptake, suggesting that expression of Cx31G45E-GFP damages the cellular membrane independent of its channel function. Our data suggest that entrapment of Cx43 and necrotic cell death in the epidermis could underlie the EKV skin phenotype.


Subject(s)
Connexins/genetics , Erythrokeratodermia Variabilis/genetics , Mutation, Missense , Cell Death , Cell Membrane/drug effects , Cells, Cultured , Connexin 43/biosynthesis , Connexin 43/genetics , Endoplasmic Reticulum/ultrastructure , Epidermis/pathology , Erythrokeratodermia Variabilis/pathology , Genes, Dominant , Genetic Association Studies , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , HeLa Cells , Humans , Keratinocytes , Necrosis , Protein Transport
4.
Hum Mol Genet ; 27(16): 2775-2788, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29741626

ABSTRACT

Winchester syndrome (WS, MIM #277950) is an extremely rare autosomal recessive skeletal dysplasia characterized by progressive joint destruction and osteolysis. To date, only one missense mutation in MMP14, encoding the membrane-bound matrix metalloprotease 14, has been reported in WS patients. Here, we report a novel hypomorphic MMP14 p.Arg111His (R111H) allele, associated with a mitigated form of WS. Functional analysis demonstrated that this mutation, in contrast to previously reported human and murine MMP14 mutations, does not affect MMP14's transport to the cell membrane. Instead, it partially impairs MMP14's proteolytic activity. This residual activity likely accounts for the mitigated phenotype observed in our patients. Based on our observations as well as previously published data, we hypothesize that MMP14's catalytic activity is the prime determinant of disease severity. Given the limitations of our in vitro assays in addressing the consequences of MMP14 dysfunction, we generated a novel mmp14a/b knockout zebrafish model. The fish accurately reflected key aspects of the WS phenotype including craniofacial malformations, kyphosis, short-stature and reduced bone density owing to defective collagen remodeling. Notably, the zebrafish model will be a valuable tool for developing novel therapeutic approaches to a devastating bone disorder.


Subject(s)
Abnormalities, Multiple/genetics , Contracture/genetics , Corneal Opacity/genetics , Craniofacial Abnormalities/genetics , Growth Disorders/genetics , Matrix Metalloproteinase 14/genetics , Osteolysis/genetics , Osteoporosis/genetics , Abnormalities, Multiple/physiopathology , Alleles , Animals , Catalytic Domain/genetics , Contracture/physiopathology , Corneal Opacity/physiopathology , Craniofacial Abnormalities/physiopathology , Gene Knockout Techniques , Growth Disorders/physiopathology , Humans , Mice , Osteolysis/physiopathology , Osteoporosis/physiopathology , Phenotype , Zebrafish
5.
Autophagy ; 10(10): 1749-60, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25126726

ABSTRACT

Birt-Hogg-Dubé (BHD) syndrome is a rare autosomal dominant condition caused by mutations in the FLCN gene and characterized by benign hair follicle tumors, pneumothorax, and renal cancer. Folliculin (FLCN), the protein product of the FLCN gene, is a poorly characterized tumor suppressor protein, currently linked to multiple cellular pathways. Autophagy maintains cellular homeostasis by removing damaged organelles and macromolecules. Although the autophagy kinase ULK1 drives autophagy, the underlying mechanisms are still being unraveled and few ULK1 substrates have been identified to date. Here, we identify that loss of FLCN moderately impairs basal autophagic flux, while re-expression of FLCN rescues autophagy. We reveal that the FLCN complex is regulated by ULK1 and elucidate 3 novel phosphorylation sites (Ser406, Ser537, and Ser542) within FLCN, which are induced by ULK1 overexpression. In addition, our findings demonstrate that FLCN interacts with a second integral component of the autophagy machinery, GABA(A) receptor-associated protein (GABARAP). The FLCN-GABARAP association is modulated by the presence of either folliculin-interacting protein (FNIP)-1 or FNIP2 and further regulated by ULK1. As observed by elevation of GABARAP, sequestome 1 (SQSTM1) and microtubule-associated protein 1 light chain 3 (MAP1LC3B) in chromophobe and clear cell tumors from a BHD patient, we found that autophagy is impaired in BHD-associated renal tumors. Consequently, this work reveals a novel facet of autophagy regulation by ULK1 and substantially contributes to our understanding of FLCN function by linking it directly to autophagy through GABARAP and ULK1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Autophagy-Related Protein-1 Homolog , Birt-Hogg-Dube Syndrome/metabolism , Birt-Hogg-Dube Syndrome/pathology , Carrier Proteins/metabolism , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Binding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/deficiency , Sequestosome-1 Protein , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/deficiency
6.
J Clin Invest ; 124(6): 2640-50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24762438

ABSTRACT

The Warburg effect is a tumorigenic metabolic adaptation process characterized by augmented aerobic glycolysis, which enhances cellular bioenergetics. In normal cells, energy homeostasis is controlled by AMPK; however, its role in cancer is not understood, as both AMPK-dependent tumor-promoting and -inhibiting functions were reported. Upon stress, energy levels are maintained by increased mitochondrial biogenesis and glycolysis, controlled by transcriptional coactivator PGC-1α and HIF, respectively. In normoxia, AMPK induces PGC-1α, but how HIF is activated is unclear. Germline mutations in the gene encoding the tumor suppressor folliculin (FLCN) lead to Birt-Hogg-Dubé (BHD) syndrome, which is associated with an increased cancer risk. FLCN was identified as an AMPK binding partner, and we evaluated its role with respect to AMPK-dependent energy functions. We revealed that loss of FLCN constitutively activates AMPK, resulting in PGC-1α-mediated mitochondrial biogenesis and increased ROS production. ROS induced HIF transcriptional activity and drove Warburg metabolic reprogramming, coupling AMPK-dependent mitochondrial biogenesis to HIF-dependent metabolic changes. This reprogramming stimulated cellular bioenergetics and conferred a HIF-dependent tumorigenic advantage in FLCN-negative cancer cells. Moreover, this pathway is conserved in a BHD-derived tumor. These results indicate that FLCN inhibits tumorigenesis by preventing AMPK-dependent HIF activation and the subsequent Warburg metabolic transformation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Birt-Hogg-Dube Syndrome/etiology , Birt-Hogg-Dube Syndrome/genetics , Birt-Hogg-Dube Syndrome/metabolism , Cell Line , Cell Transformation, Neoplastic , Energy Metabolism , Hypoxia-Inducible Factor 1/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
7.
Hum Mol Genet ; 22(21): 4383-97, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23784378

ABSTRACT

Birt-Hogg-Dubé (BHD) syndrome is an autosomal dominant disorder where patients are predisposed to kidney cancer, lung and kidney cysts and benign skin tumors. BHD is caused by heterozygous mutations affecting folliculin (FLCN), a conserved protein that is considered a tumor suppressor. Previous research has uncovered multiple roles for FLCN in cellular physiology, yet it remains unclear how these translate to BHD lesions. Since BHD manifests hallmark characteristics of ciliopathies, we speculated that FLCN might also have a ciliary role. Our data indicate that FLCN localizes to motile and non-motile cilia, centrosomes and the mitotic spindle. Alteration of FLCN levels can cause changes to the onset of ciliogenesis, without abrogating it. In three-dimensional culture, abnormal expression of FLCN disrupts polarized growth of kidney cells and deregulates canonical Wnt signalling. Our findings further suggest that BHD-causing FLCN mutants may retain partial functionality. Thus, several BHD symptoms may be due to abnormal levels of FLCN rather than its complete loss and accordingly, we show expression of mutant FLCN in a BHD-associated renal carcinoma. We propose that BHD is a novel ciliopathy, its symptoms at least partly due to abnormal ciliogenesis and canonical Wnt signalling.


Subject(s)
Birt-Hogg-Dube Syndrome/physiopathology , Cilia/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Base Sequence , Birt-Hogg-Dube Syndrome/genetics , Cell Line , Cell Polarity , Cell Proliferation , Centrosome/physiology , Cilia/pathology , Humans , Kidney/physiology , Microtubules/physiology , Molecular Sequence Data , Sequence Analysis, DNA , Wnt Signaling Pathway
8.
Fam Cancer ; 12(3): 381-5, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23307118

ABSTRACT

Birt-Hogg-Dubé syndrome (MIM #135150) is characterized by the development of benign skin tumours called fibrofolliculomas, pulmonary cysts that may lead to pneumothorax and a high risk of developing kidney cancer. BHD is caused by mutations affecting the highly conserved protein folliculin (FLCN), which probably has a role in intracellular transport. Most of the research effort directed towards BHD has focused on understanding how loss of FLCN causes kidney cancer. The cutaneous manifestations have received comparatively little attention. Although understandable, it is unfortunate, as the fibrofolliculomas are highly accessible and thus potentially are an excellent system for trying to understand the basic pathobiology of BHD. Also, patients can be very much burdened by the cosmetic consequences of having hundreds of facial skin tumours. Our lack of insight into what drives fibrofolliculoma growth translates into a very limited therapeutic arsenal. Thus, paying attention to fibrofolliculomas has both basic science and practical benefits. In this review, we will discuss the state of the art regarding our understanding of fibrofolliculoma pathogenesis and indicate future directions for research.


Subject(s)
Birt-Hogg-Dube Syndrome/complications , Skin Diseases/etiology , Humans , Skin Diseases/diagnosis , Skin Diseases/therapy
9.
Fam Cancer ; 12(3): 373-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23264078

ABSTRACT

Birt-Hogg-Dubé syndrome (BHD) is an autosomal dominant condition due to germline FLCN (folliculin) mutations, characterized by skin fibrofolliculomas, lung cysts, pneumothorax and renal cancer. We identified a de novo FLCN mutation, c.499C>T (p.Gln167X), in a patient who presented with spontaneous pneumothorax. Subsequently, typical skin features and asymptomatic renal cancer were diagnosed. Probably, de novo FLCN mutations are rare. However, they may be under-diagnosed if BHD is not considered in sporadic patients who present with one or more of the syndromic features. Genetic and immunohistochemical analysis of the renal tumour indicated features compatible with a tumour suppressor role of FLCN. The finding that mutant FLCN was expressed in the tumour might indicate residual functionality of mutant FLCN, a notion which will be explored in future studies.


Subject(s)
Germ-Line Mutation/genetics , Kidney Neoplasms/genetics , Pneumothorax/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Adult , Humans , Immunoenzyme Techniques , Kidney Neoplasms/diagnosis , Magnetic Resonance Imaging , Male , Pneumothorax/diagnosis , Prognosis , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...