Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 32(14): 2347-2356, 2023 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-37162351

RESUMEN

Medium-chain acyl-CoA dehydrogenase (MCAD) deficiency is the most common inherited disorder of mitochondrial fatty acid ß-oxidation (FAO) in humans. Patients exhibit clinical episodes often associated with fasting. Symptoms include hypoketotic hypoglycemia and Reye-like episodes. With limited treatment options, we explored the use of human MCAD (hMCAD) mRNA in fibroblasts from patients with MCAD deficiency to provide functional MCAD protein and reverse the metabolic block. Transfection of hMCAD mRNA into MCAD- deficient patient cells resulted in an increased MCAD protein that localized to mitochondria, concomitant with increased enzyme activity in cell extracts. The therapeutic hMCAD mRNA-lipid nanoparticle (LNP) formulation was also tested in vivo in Acadm-/- mice. Administration of multiple intravenous doses of the hMCAD mRNA-LNP complex (LNP-MCAD) into Acadm-/- mice produced a significant level of MCAD protein with increased enzyme activity in liver, heart and skeletal muscle homogenates. Treated Acadm-/- mice were more resistant to cold stress and had decreased plasma levels of medium-chain acylcarnitines compared to untreated animals. Furthermore, hepatic steatosis in the liver from treated Acadm-/- mice was reduced compared to untreated ones. Results from this study support the potential therapeutic value of hMCAD mRNA-LNP complex treatment for MCAD deficiency.


Asunto(s)
Acil-CoA Deshidrogenasas , Fibroblastos , Humanos , Ratones , Animales , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasa/metabolismo , ARN Mensajero/genética , Modelos Animales de Enfermedad , Fibroblastos/metabolismo
2.
Ann Rheum Dis ; 82(3): 365-373, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36600187

RESUMEN

OBJECTIVES: To mine the serum proteome of patients with systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH) and to detect biomarkers that may assist in earlier and more effective diagnosis and treatment. METHODS: Patients with limited cutaneous SSc, no extensive interstitial lung disease and no PAH-specific therapy were included. They were classified as cases if they had PAH confirmed by right heart catheterisation (RHC) and serum collected on the same day as RHC; and as controls if they had no clinical evidence of PAH. RESULTS: Patients were mostly middle-aged females with anticentromere-associated SSc. Among 1129 proteins assessed by a high-throughput proteomic assay (SOMAscan), only 2 were differentially expressed and correlated significantly with pulmonary vascular resistance (PVR) in SSc-PAH patients (n=15): chemerin (ρ=0.62, p=0.01) and SET (ρ=0.62, p=0.01). To validate these results, serum levels of chemerin were measured by ELISA in an independent cohort. Chemerin levels were confirmed to be significantly higher (p=0.01) and correlate with PVR (ρ=0.42, p=0.04) in SSc-PAH patients (n=24). Chemerin mRNA expression was detected in fibroblasts, pulmonary artery smooth muscle cells (PA-SMCs)/pericytes and mesothelial cells in SSc-PAH lungs by single-cell RNA-sequencing. Confocal immunofluorescence revealed increased expression of a chemerin receptor, CMKLR1, on SSc-PAH PA-SMCs. SSc-PAH serum seemed to induce higher PA-SMC proliferation than serum from SSc patients without PAH. This difference appeared neutralised when adding the CMKLR1 inhibitor α-NETA. CONCLUSION: Chemerin seems an interesting surrogate biomarker for PVR in SSc-PAH. Increased chemerin serum levels and CMKLR1 expression by PA-SMCs may contribute to SSc-PAH pathogenesis by inducing PA-SMC proliferation.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Esclerodermia Sistémica , Persona de Mediana Edad , Femenino , Humanos , Hipertensión Pulmonar/etiología , Proteoma , Proteómica , Hipertensión Arterial Pulmonar/etiología , Hemodinámica , Biomarcadores , Esclerodermia Sistémica/complicaciones
3.
Mol Genet Metab ; 138(1): 106982, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36580829

RESUMEN

Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is an inborn error of long chain fatty acid ß-oxidation (FAO) with limited treatment options. Patients present with heterogeneous clinical phenotypes affecting predominantly heart, liver, and skeletal muscle. While VLCAD deficiency is a systemic disease, restoration of liver FAO has the potential to improve symptoms more broadly due to increased total body ATP production and reduced accumulation of potentially toxic metabolites. We explored the use of synthetic human VLCAD (hVLCAD) mRNA and lipid nanoparticle encapsulated hVLCAD mRNA (LNP-VLCAD) to generate functional VLCAD enzyme in patient fibroblasts derived from VLCAD deficient patients, mouse embryonic fibroblasts, hepatocytes isolated from VLCAD knockout (Acadvl-/-) mice, and Acadvl-/- mice to reverse the metabolic effects of the deficiency. Transfection of all cell types with hVLCAD mRNA resulted in high level expression of protein that localized to mitochondria with increased enzyme activity. Intravenous administration of LNP-VLCAD to Acadvl-/- mice produced a significant amount of VLCAD protein in liver, which declined over a week. Treated Acadvl-/- mice showed reduced hepatic steatosis, were more resistant to cold stress, and accumulated less toxic metabolites in blood than untreated animals. Results from this study support the potential for hVLCAD mRNA for treatment of VLCAD deficiency.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga , Errores Innatos del Metabolismo Lipídico , Humanos , Animales , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/terapia
4.
J Inherit Metab Dis ; 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-38044746

RESUMEN

Argininosuccinate lyase (ASL) is integral to the urea cycle detoxifying neurotoxic ammonia and the nitric oxide (NO) biosynthesis cycle. Inherited ASL deficiency causes argininosuccinic aciduria (ASA), a rare disease with hyperammonemia and NO deficiency. Patients present with developmental delay, epilepsy and movement disorder, associated with NO-mediated downregulation of central catecholamine biosynthesis. A neurodegenerative phenotype has been proposed in ASA. To better characterise this neurodegenerative phenotype in ASA, we conducted a retrospective study in six paediatric and adult metabolic centres in the UK in 2022. We identified 60 patients and specifically looked for neurodegeneration-related symptoms: movement disorder such as ataxia, tremor and dystonia, hypotonia/fatigue and abnormal behaviour. We analysed neuroimaging with diffusion tensor imaging (DTI) magnetic resonance imaging (MRI) in an individual with ASA with movement disorders. We assessed conventional and DTI MRI alongside single photon emission computer tomography (SPECT) with dopamine analogue radionuclide 123 I-ioflupane, in Asl-deficient mice treated by hASL mRNA with normalised ureagenesis. Movement disorders in ASA appear in the second and third decades of life, becoming more prevalent with ageing and independent from the age of onset of hyperammonemia. Neuroimaging can show abnormal DTI features affecting both grey and white matter, preferentially basal ganglia. ASA mouse model with normalised ureagenesis did not recapitulate these DTI findings and showed normal 123 I-ioflupane SPECT and cerebral dopamine metabolomics. Altogether these findings support the pathophysiology of a late-onset movement disorder with cell-autonomous functional central catecholamine dysregulation but without or limited neurodegeneration of dopaminergic neurons, making these symptoms amenable to targeted therapy.

5.
Am J Pathol ; 186(10): 2650-64, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27565038

RESUMEN

Tissue injury triggers the activation and differentiation of multiple cell types to minimize damage and initiate repair processes. In systemic sclerosis, these repair processes appear to run unchecked, leading to aberrant remodeling and fibrosis of the skin and multiple internal organs, yet the fundamental pathological defect remains unknown. We describe herein a transition wherein the abundant CD34(+) dermal fibroblasts present in healthy human skin disappear in the skin of systemic sclerosis patients, and CD34(-), podoplanin(+), and CD90(+) fibroblasts appear. This transition is limited to the upper dermis in several inflammatory skin diseases, yet in systemic sclerosis, it can occur in all regions of the dermis. In vitro, primary dermal fibroblasts readily express podoplanin in response to the inflammatory stimuli tumor necrosis factor and IL-1ß. Furthermore, we show that on acute skin injury in both human and murine settings, this transition occurs quickly, consistent with a response to inflammatory signaling. Transitioned fibroblasts partially resemble the cells that form the reticular networks in organized lymphoid tissues, potentially linking two areas of fibroblast research. These results allow for the visualization and quantification of a basic stage of fibroblast differentiation in inflammatory and fibrotic diseases in the skin.


Asunto(s)
Fibrosis/patología , Glicoproteínas de Membrana/metabolismo , Esclerodermia Sistémica/patología , Antígenos Thy-1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Diferenciación Celular , Dermis/inmunología , Dermis/patología , Femenino , Fibroblastos/inmunología , Fibroblastos/patología , Fibrosis/inmunología , Humanos , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Esclerodermia Sistémica/inmunología , Piel/inmunología , Piel/patología , Factor de Necrosis Tumoral alfa/metabolismo
6.
Exp Dermatol ; 24(2): 133-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25425469

RESUMEN

Toll-like receptor 9 (TLR9) drives innate immune responses after recognition of foreign or endogenous DNA containing unmethylated CpG motifs. DNA-mediated TLR9 activation is highly implicated in the pathogenesis of several autoimmune skin diseases, yet its contribution to the inflammation seen in these diseases remains unclear. In this study, TLR9 ligand, CpGB DNA, was administered to mice via a subcutaneous osmotic pump with treatment lasting 1 or 4 weeks. Gene expression and immunofluorescence analyses were used to determine chemokine expression and cell recruitment in the skin surrounding the pump outlet. CpGB DNA skin treatment dramatically induced a marked influx of CD11b+ F4/80+ macrophages, increasing over 4 weeks of treatment, and induction of IFNγ and TNFα expression. Chemokines, CCL2, CCL4, CCL5, CXCL9 and CXCL10, were highly induced in CpGB DNA-treated skin, although abrogation of these signalling pathways individually did not alter macrophage accumulation. Flow cytometry analysis showed that TLR9 activation in the skin increased circulating CD11b+ CD115+ Ly6C(hi) inflammatory monocytes following 1 week of CpGB DNA treatment. Additionally, skin-resident CD11b+ cells were found to initially take up subcutaneous CpGB DNA and propagate the subsequent immune response. Using diphtheria toxin-induced monocyte depletion mouse model, gene expression analysis demonstrated that CD11b+ cells are responsible for the CpGB DNA-induced cytokine and chemokine response. Overall, these data demonstrate that chronic TLR9 activation induces a specific inflammatory response, ultimately leading to a striking and selective accumulation of macrophages in the skin.


Asunto(s)
Islas de CpG , Activación de Macrófagos , Macrófagos/metabolismo , Piel/metabolismo , Administración Cutánea , Animales , Antígeno CD11b/metabolismo , Quimiocinas/metabolismo , ADN , Toxina Diftérica/química , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inmunohistoquímica , Inflamación/metabolismo , Interferón gamma/metabolismo , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Monocitos/citología , Ósmosis , Piel/citología , Receptor Toll-Like 9/metabolismo
7.
Nucleic Acids Res ; 40(21): e164, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22879378

RESUMEN

Life abounds with genetic variations writ in sequences that are often only a few hundred nucleotides long. Rapid detection of these variations for identification of genetic diseases, pathogens and organisms has become the mainstay of molecular science and medicine. This report describes a new, highly informative closed-tube polymerase chain reaction (PCR) strategy for analysis of both known and unknown sequence variations. It combines efficient quantitative amplification of single-stranded DNA targets through LATE-PCR with sets of Lights-On/Lights-Off probes that hybridize to their target sequences over a broad temperature range. Contiguous pairs of Lights-On/Lights-Off probes of the same fluorescent color are used to scan hundreds of nucleotides for the presence of mutations. Sets of probes in different colors can be combined in the same tube to analyze even longer single-stranded targets. Each set of hybridized Lights-On/Lights-Off probes generates a composite fluorescent contour, which is mathematically converted to a sequence-specific fluorescent signature. The versatility and broad utility of this new technology is illustrated in this report by characterization of variant sequences in three different DNA targets: the rpoB gene of Mycobacterium tuberculosis, a sequence in the mitochondrial cytochrome C oxidase subunit 1 gene of nematodes and the V3 hypervariable region of the bacterial 16 s ribosomal RNA gene. We anticipate widespread use of these technologies for diagnostics, species identification and basic research.


Asunto(s)
ADN/química , Colorantes Fluorescentes/química , Variación Genética , Sondas de Ácido Nucleico/química , Reacción en Cadena de la Polimerasa/métodos , Secuencia de Bases , Conformación de Ácido Nucleico , Nucleótidos/química , Temperatura
8.
Vet Rec ; 194(11): i-ii, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38819847

RESUMEN

Lisa Rice is south area supervisor for Pet Blood Bank, having first gained experience in small animal practice then training to take blood donations and organise donation sessions when she joined the organisation.


Asunto(s)
Técnicos de Animales , Animales , Perros , Humanos , Técnicos de Animales/educación , Bancos de Sangre , Reino Unido , Medicina Veterinaria/organización & administración
9.
Nat Commun ; 15(1): 3804, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714648

RESUMEN

Messenger RNA (mRNA) therapeutics delivered via lipid nanoparticles hold the potential to treat metabolic diseases caused by protein deficiency, including propionic acidemia (PA), methylmalonic acidemia (MMA), and phenylketonuria (PKU). Herein we report results from multiple independent preclinical studies of mRNA-3927 (an investigational treatment for PA), mRNA-3705 (an investigational treatment for MMA), and mRNA-3210 (an investigational treatment for PKU) in murine models of each disease. All 3 mRNA therapeutics exhibited pharmacokinetic/pharmacodynamic (PK/PD) responses in their respective murine model by driving mRNA, protein, and/or protein activity responses, as well as by decreasing levels of the relevant biomarker(s) when compared to control-treated animals. These preclinical data were then used to develop translational PK/PD models, which were scaled allometrically to humans to predict starting doses for first-in-human clinical studies for each disease. The predicted first-in-human doses for mRNA-3927, mRNA-3705, and mRNA-3210 were determined to be 0.3, 0.1, and 0.4 mg/kg, respectively.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Modelos Animales de Enfermedad , Fenilcetonurias , Acidemia Propiónica , ARN Mensajero , Acidemia Propiónica/genética , Acidemia Propiónica/terapia , Acidemia Propiónica/tratamiento farmacológico , Animales , Fenilcetonurias/genética , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/terapia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Ratones , Humanos , Masculino , Femenino , Nanopartículas/química , Ratones Endogámicos C57BL , Liposomas
10.
Sci Transl Med ; 16(729): eadh1334, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38198573

RESUMEN

The urea cycle enzyme argininosuccinate lyase (ASL) enables the clearance of neurotoxic ammonia and the biosynthesis of arginine. Patients with ASL deficiency present with argininosuccinic aciduria, an inherited metabolic disease with hyperammonemia and a systemic phenotype coinciding with neurocognitive impairment and chronic liver disease. Here, we describe the dysregulation of glutathione biosynthesis and upstream cysteine utilization in ASL-deficient patients and mice using targeted metabolomics and in vivo positron emission tomography (PET) imaging using (S)-4-(3-18F-fluoropropyl)-l-glutamate ([18F]FSPG). Up-regulation of cysteine metabolism contrasted with glutathione depletion and down-regulated antioxidant pathways. To assess hepatic glutathione dysregulation and liver disease, we present [18F]FSPG PET as a noninvasive diagnostic tool to monitor therapeutic response in argininosuccinic aciduria. Human hASL mRNA encapsulated in lipid nanoparticles improved glutathione metabolism and chronic liver disease. In addition, hASL mRNA therapy corrected and rescued the neonatal and adult Asl-deficient mouse phenotypes, respectively, enhancing ureagenesis. These findings provide mechanistic insights in liver glutathione metabolism and support clinical translation of mRNA therapy for argininosuccinic aciduria.


Asunto(s)
Aciduria Argininosuccínica , Hepatopatías , Adulto , Humanos , Animales , Ratones , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/terapia , Cisteína , Glutatión , Metabolómica
11.
F1000Res ; 12: 1580, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38618017

RESUMEN

Background: In academic research and the pharmaceutical industry, in vitro cell lines and in vivo animal models are considered as gold standards in modelling diseases and assessing therapeutic efficacy. However, both models have intrinsic limitations, whilst the use of precision-cut tissue slices can bridge the gap between these mainstream models. Precision-cut tissue slices combine the advantage of high reproducibility, studying all cell sub-types whilst preserving the tissue matrix and extracellular architecture, thereby closely mimicking a mini-organ. This approach can be used to replicate the biological phenotype of liver monogenic diseases using mouse models. Methods: Here, we describe an optimised and easy-to-implement protocol for the culture of sections from mouse livers, enabling its use as a reliable ex-vivo model to assess the therapeutic screening of inherited metabolic diseases. Results: We show that precision-cut liver sections can be a reliable model for recapitulating the biological phenotype of inherited metabolic diseases, exemplified by common urea cycle defects such as citrullinemia type 1 and argininosuccinic aciduria, caused by argininosuccinic synthase (ASS1) and argininosuccinic lyase (ASL) deficiencies respectively. Conclusions: Therapeutic response to gene therapy such as messenger RNA replacement delivered via lipid nanoparticles can be monitored, demonstrating that precision-cut liver sections can be used as a preclinical screening tool to assess therapeutic response and toxicity in monogenic liver diseases.


Asunto(s)
Hepatopatías , Enfermedades Metabólicas , Animales , Ratones , Reproducibilidad de los Resultados , Hepatopatías/genética , Hepatopatías/terapia , Fenotipo
13.
Nat Commun ; 12(1): 3090, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035281

RESUMEN

Glycogen Storage Disease 1a (GSD1a) is a rare, inherited metabolic disorder caused by deficiency of glucose 6-phosphatase (G6Pase-α). G6Pase-α is critical for maintaining interprandial euglycemia. GSD1a patients exhibit life-threatening hypoglycemia and long-term liver complications including hepatocellular adenomas (HCAs) and carcinomas (HCCs). There is no treatment for GSD1a and the current standard-of-care for managing hypoglycemia (Glycosade®/modified cornstarch) fails to prevent HCA/HCC risk. Therapeutic modalities such as enzyme replacement therapy and gene therapy are not ideal options for patients due to challenges in drug-delivery, efficacy, and safety. To develop a new treatment for GSD1a capable of addressing both the life-threatening hypoglycemia and HCA/HCC risk, we encapsulated engineered mRNAs encoding human G6Pase-α in lipid nanoparticles. We demonstrate the efficacy and safety of our approach in a preclinical murine model that phenotypically resembles the human condition, thus presenting a potential therapy that could have a significant therapeutic impact on the treatment of GSD1a.


Asunto(s)
Modelos Animales de Enfermedad , Terapia Genética/métodos , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno/terapia , ARN Mensajero/genética , Animales , Línea Celular Tumoral , Citocinas/sangre , Citocinas/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno/patología , Células HeLa , Humanos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Mensajero/administración & dosificación , ARN Mensajero/química , Resultado del Tratamiento , Triglicéridos/metabolismo
14.
Sci Rep ; 10(1): 7052, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32341402

RESUMEN

Alpha 1-antitrypsin (AAT) deficiency arises from an inherited mutation in the SERPINA1 gene. The disease causes damage in the liver where the majority of the AAT protein is produced. Lack of functioning circulating AAT protein also causes uninhibited elastolytic activity in the lungs leading to AAT deficiency-related emphysema. The only therapy apart from liver transplantation is augmentation with human AAT protein pooled from sera, which is only reserved for patients with advanced lung disease caused by severe AAT deficiency. We tested modified mRNA encoding human AAT in primary human hepatocytes in culture, including hepatocytes from AAT deficient patients. Both expression and functional activity were investigated. Secreted AAT protein increased from 1,14 to 3,43 µg/ml in media from primary human hepatocytes following mRNA treatment as investigated by ELISA and western blot. The translated protein showed activity and protease inhibitory function as measured by elastase activity assay. Also, mRNA formulation in lipid nanoparticles was assessed for systemic delivery in both wild type mice and the NSG-PiZ transgenic mouse model of AAT deficiency. Systemic intravenous delivery of modified mRNA led to hepatic uptake and translation into a functioning protein in mice. These data support the use of systemic mRNA therapy as a potential treatment for AAT deficiency.


Asunto(s)
ARN Mensajero/metabolismo , Deficiencia de alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/terapia , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Nanopartículas/química , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/fisiología
15.
Front Immunol ; 9: 2191, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30327649

RESUMEN

Objective: The mechanisms that lead to endothelial cell (EC) injury and propagate the vasculopathy in Systemic Sclerosis (SSc) are not well understood. Using single cell RNA sequencing (scRNA-seq), our goal was to identify EC markers and signature pathways associated with vascular injury in SSc skin. Methods: We implemented single cell sorting and subsequent RNA sequencing of cells isolated from SSc and healthy control skin. We used t-distributed stochastic neighbor embedding (t-SNE) to identify the various cell types. We performed pathway analysis using Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA). Finally, we independently verified distinct markers using immunohistochemistry on skin biopsies and qPCR in primary ECs from SSc and healthy skin. Results: By combining the t-SNE analysis with the expression of known EC markers, we positively identified ECs among the sorted cells. Subsequently, we examined the differential expression profile between the ECs from healthy and SSc skin. Using GSEA and IPA analysis, we demonstrated that the SSc endothelial cell expression profile is enriched in processes associated with extracellular matrix generation, negative regulation of angiogenesis and epithelial-to-mesenchymal transition. Two of the top differentially expressed genes, HSPG2 and APLNR, were independently verified using immunohistochemistry staining and real-time qPCR analysis. Conclusion: ScRNA-seq, differential gene expression and pathway analysis revealed that ECs from SSc patients show a discrete pattern of gene expression associated with vascular injury and activation, extracellular matrix generation and negative regulation of angiogenesis. HSPG2 and APLNR were identified as two of the top markers of EC injury in SSc.


Asunto(s)
Receptores de Apelina/genética , Receptores de Apelina/metabolismo , Células Endoteliales/metabolismo , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Secuencia de Bases , Biomarcadores/metabolismo , Biopsia , Activación de Complemento , Transición Epitelial-Mesenquimal/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Expresión Génica , Humanos , Inmunohistoquímica , Metaloproteinasas de la Matriz/metabolismo , Neovascularización Patológica/metabolismo , Esclerodermia Sistémica/patología , Análisis de Secuencia de ARN , Lesiones del Sistema Vascular/patología
16.
J Scleroderma Relat Disord ; 3(3): 242-248, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30498788

RESUMEN

OBJECTIVE: Pulmonary arterial hypertension (PAH) and interstitial lung disease (ILD) are major causes of mortality in systemic sclerosis (SSc). We used a previously identified microarray biomarker to determine if SSc-PAH and SSc-ILD patients demonstrate distinct gene expression profiles. METHODS: PBMCs were collected from healthy controls (n=10), SSc (SSc) patients without pulmonary hypertension [SSc-noPAH, n=39], and SSc-PAH patients (n=21; mPAP≥25, PCWP≤15, PVR≥3WU) diagnosed by right heart catheterization (RHC). SSc-ILD patients were defined as those with evidence of fibrosis on chest CT and significant restriction (FVC<70% predicted, n = 11). SSc-PAH biomarker included 69 genes selected by unbiased statistical screening of 3 publicly available microarray studies. RNA levels were measured by Nanostring. Gene expression levels that were significantly correlated with PAH (multiple statistical measures) were chosen as inputs into a forward selection logistic regression model. RESULTS: When ILD patients were included (n=64), 4 genes (S100P, CD8B1, CCL2, TIMP1) and male sex predicted PAH with a high level of accuracy (AUC = 0.83). Without ILD patients (n=53), 2 genes (THBS1, CD8B1) and male sex predicted PAH with a high level of accuracy (AUC = 0.80). When examining SSc patients with borderline elevated pulmonary pressures (mPAP = 21-24 mmHg), gene expression changes closely resembled the SSc-PAH group, except for THBS1. CONCLUSION: SSc-PAH and SSc-ILD have similar, but distinct, gene expression profiles. Many gene expression changes occur early in the disease course, potentially allowing for early detection. THBS1 appears to be an important mediator in the development of PAH-predominant phenotype. Further prospective investigation is warranted.

17.
Arthritis Res Ther ; 20(1): 185, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-30115106

RESUMEN

BACKGROUND: Systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH) is one of the leading causes of death in SSc. Identification of a serum-based proteomic diagnostic biomarker for SSc-PAH would allow for rapid non-invasive screening and could positively impact patient survival. Identification and validation of novel proteins could potentially facilitate the identification of SSc-PAH, and might also point to important protein mediators in pathogenesis. METHODS: Thirteen treatment-naïve SSc-PAH patients had serum collected at time of diagnosis and were used as the discovery cohort for the protein-expression biomarker. Two proteins, Midkine and Follistatin-like 3 (FSTL3) were then validated by enzyme-linked immunosorbent assays. Midkine and FSTL3 were tested in combination to identify SSc-PAH and were validated in two independent cohorts of SSc-PAH (n = 23, n = 11). RESULTS: Eighty-two proteins were found to be differentially regulated in SSc-PAH sera. Two proteins (Midkine and FSTL3) were also shown to be elevated in publicly available data and their expression was evaluated in independent cohorts. In the validation cohorts, the combination of Midkine and FSTL3 had an area under the receiver operating characteristic curve (AUC) of 0.85 and 0.92 with respective corresponding measures of sensitivity of 76% and 91%, and specificity measures of 76% and 80%. CONCLUSIONS: These findings indicate that there is a clear delineation between overall protein expression in sera from SSc patients and those with SSc-PAH. The combination of Midkine and FSTL3 can serve as an SSc-PAH biomarker and are potential drug targets for this rare disease population.


Asunto(s)
Biomarcadores/sangre , Proteínas Relacionadas con la Folistatina/sangre , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/etiología , Midkina/sangre , Esclerodermia Sistémica/complicaciones , Anciano , Anciano de 80 o más Años , Diagnóstico Precoz , Femenino , Humanos , Hipertensión Pulmonar/sangre , Masculino , Persona de Mediana Edad , Esclerodermia Sistémica/sangre , Sensibilidad y Especificidad
18.
Arthritis Rheumatol ; 70(6): 912-919, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29858547

RESUMEN

OBJECTIVE: At present, there are no clinical or laboratory measures that accurately forecast the progression of skin fibrosis and organ involvement in patients with systemic sclerosis (SSc). The goal of this study was to identify skin biomarkers that could be prognostic for the progression of skin fibrosis in patients with early diffuse cutaneous SSc (dcSSc). METHODS: We analyzed clinical data and gene expression in skin biopsy samples from 38 placebo-treated patients, part of the Roche Safety and Efficacy of Subcutaneous Tocilizumab in Adults with Systemic Sclerosis (FASSCINATE) phase II study of tocilizumab in SSc. RNA samples were analyzed using nCounter. A trajectory model based on a modified Rodnan skin thickness score was used to describe 3 skin disease trajectories over time. We examined the association of skin gene expression with skin score trajectory groups, by chi-square test. Logistic regression was used to examine the prognostic power of each gene identified. RESULTS: We found that placebo-treated patients with high expression of messenger RNA for CD14, SERPINE1, IL13RA1, CTGF, and OSMR at baseline were more likely to have progressive skin score trajectories. We also found that those genes were prognostic for the risk of skin progression and that IL13RA1, OSMR, and SERPINE1 performed the best. CONCLUSION: Skin gene expression of biomarkers associated with macrophages (CD14, IL13RA1) and transforming growth factor ß activation (SERPINE1, CTGF, OSMR) are prognostic for progressive skin disease in patients with dcSSc. These biomarkers may provide guidance in decision-making about which patients should be considered for aggressive therapies and/or for clinical trials.


Asunto(s)
Expresión Génica , Macrófagos/metabolismo , ARN Mensajero/metabolismo , Esclerodermia Difusa/genética , Piel/citología , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/uso terapéutico , Ensayos Clínicos Fase II como Asunto , Factor de Crecimiento del Tejido Conjuntivo/genética , Progresión de la Enfermedad , Método Doble Ciego , Femenino , Fibrosis , Marcadores Genéticos/genética , Humanos , Subunidad alfa1 del Receptor de Interleucina-13/genética , Receptores de Lipopolisacáridos/genética , Masculino , Persona de Mediana Edad , Subunidad beta del Receptor de Oncostatina M/genética , Inhibidor 1 de Activador Plasminogénico/genética , Pronóstico , Ensayos Clínicos Controlados Aleatorios como Asunto , Esclerodermia Difusa/tratamiento farmacológico , Esclerodermia Difusa/patología , Índice de Severidad de la Enfermedad , Piel/patología , Adulto Joven
20.
J Invest Dermatol ; 137(1): 62-70, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27640094

RESUMEN

In this study we systematically investigated alterations in the serum proteome of patients with diffuse cutaneous systemic sclerosis and identified differentially expressed proteins that correlated with disease severity. Our goal was to identify a combination of serum proteins that would provide a biological measure for the extent of skin disease and that could be combined into a longitudinal pharmacodynamic biomarker. We found that 16% of the sera proteins analyzed by SOMAscan aptamer technology, from two cohorts of patients with diffuse cutaneous systemic sclerosis, were identified as differentially regulated between diffuse cutaneous systemic sclerosis and controls and correlated with modified Rodnan skin score. This dataset showed tumor necrosis factor-α, IFN-γ, transforming growth factor-ß, and IL-13 as potential upstream regulators of the serum protein patterns in the sera of patients with diffuse cutaneous systemic sclerosis. By ELISA, two analytes (ST2 and Spondin-1) best described longitudinal change in modified Rodnan skin score, using linear mixed models. This model was then validated in three independent cohorts. In this study we discovered a large array of proteins not previously associated with systemic sclerosis that provide insight into pathogenesis and potential targets for therapeutic intervention. Furthermore, we show that two of these proteins can be combined to form a robust longitudinal biomarker that might be used in clinical trials to assess changes in diffuse cutaneous systemic sclerosis skin disease over time.


Asunto(s)
Citocinas/metabolismo , Proteoma/metabolismo , Esclerodermia Difusa/sangre , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Modelos Lineales , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Pruebas de Farmacogenómica , Reproducibilidad de los Resultados , Esclerodermia Difusa/tratamiento farmacológico , Esclerodermia Difusa/fisiopatología , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA