Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
1.
Signal Transduct Target Ther ; 9(1): 39, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355690

RESUMEN

Immunostaining in lungs of patients who died with COVID-19 infection showed increased intensity and distribution of chondroitin sulfate and decline in N-acetylgalactostamine-4-sulfatase (Arylsulfatase B; ARSB). To explain these findings, human small airway epithelial cells were exposed to the SARS-CoV-2 spike protein receptor binding domain (SPRBD) and transcriptional mechanisms were investigated. Phospho-p38 MAPK and phospho-SMAD3 increased following exposure to the SPRBD, and their inhibition suppressed the promoter activation of the carbohydrate sulfotransferases CHST15 and CHST11, which contributed to chondroitin sulfate biosynthesis. Decline in ARSB was mediated by phospho-38 MAPK-induced N-terminal Rb phosphorylation and an associated increase in Rb-E2F1 binding and decline in E2F1 binding to the ARSB promoter. The increases in chondroitin sulfotransferases were inhibited when treated with phospho-p38-MAPK inhibitors, SMAD3 (SIS3) inhibitors, as well as antihistamine desloratadine and antibiotic monensin. In the mouse model of carrageenan-induced systemic inflammation, increases in phospho-p38 MAPK and expression of CHST15 and CHST11 and declines in DNA-E2F binding and ARSB expression occurred in the lung, similar to the observed effects in this SPRBD model of COVID-19 infection. Since accumulation of chondroitin sulfates is associated with fibrotic lung conditions and diffuse alveolar damage, increased attention to p38-MAPK inhibition may be beneficial in ameliorating Covid-19 infections.


Asunto(s)
COVID-19 , N-Acetilgalactosamina-4-Sulfatasa , Ratones , Animales , Humanos , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , Sulfatos de Condroitina/farmacología , Sulfatos de Condroitina/química , Sulfatos de Condroitina/metabolismo , Glicoproteína de la Espiga del Coronavirus , Carbohidrato Sulfotransferasas , Enzima Convertidora de Angiotensina 2 , Proteínas Quinasas p38 Activadas por Mitógenos/genética , SARS-CoV-2/metabolismo
2.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166913, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37813168

RESUMEN

In the syngeneic, subcutaneous B16F10 mouse model of malignant melanoma, treatment with exogenous ARSB markedly reduced tumor size and extended survival. In vivo experiments showed that local treatment with exogenous N-acetylgalactosamine-4-sulfatase (Arylsulfatase B; ARSB) led to reduced tumor growth over time (p < 0.0001) and improved the probability of survival up to 21 days (p = 0.0391). Tumor tissue from the treated mice had lower chondroitin 4-sulfate (C4S) content and lower sulfotransferase activity. The free galectin-3 declined, and the SHP2 activity increased, due to altered binding with chondroitin 4-sulfate. These changes induced effects on transcription, which were mediated by Sp1, phospho-ERK1/2, and phospho-p38 MAPK. Reduced mRNA expression of chondroitin sulfate proteoglycan 4 (CSPG4), carbohydrate sulfotransferase 15 (N-acetylgalactosamine 4-sulfate 6-O-sulfotransferase), and matrix metalloproteinases 2 and 9 resulted. Experiments in the human melanoma cell line A375 demonstrated similar responses to exogenous ARSB as in the tumors, and inverse effects followed ARSB siRNA. ARSB, which removes the 4-sulfate group at the non-reducing end of C4S, acts as a tumor suppressor, and treatment with exogenous ARSB impacts on vital cell signaling and reduces the expression of critical genes associated with melanoma progression.


Asunto(s)
Melanoma , N-Acetilgalactosamina-4-Sulfatasa , Neoplasias Cutáneas , Animales , Humanos , Ratones , Sulfatos de Condroitina/metabolismo , Melanoma/tratamiento farmacológico , N-Acetilgalactosamina-4-Sulfatasa/genética , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Melanoma Cutáneo Maligno
3.
JCI Insight ; 8(21)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37751300

RESUMEN

Mucopolysaccharidosis VI (MPS VI) is a rare lysosomal disease arising from impaired function of the enzyme arylsulfatase B (ARSB). This impairment causes aberrant accumulation of dermatan sulfate, a glycosaminoglycan (GAG) abundant in cartilage. While clinical severity varies along with age at first symptom manifestation, MPS VI usually presents early and strongly affects the skeleton. Current enzyme replacement therapy (ERT) does not provide effective treatment for the skeletal manifestations of MPS VI. This lack of efficacy may be due to an inability of ERT to reach affected cells or to the irreversibility of the disease. To address the question of reversibility of skeletal phenotypes, we generated a conditional by inversion (COIN) mouse model of MPS VI, ArsbCOIN/COIN, wherein Arsb is initially null and can be restored to WT using Cre. We restored Arsb at different times during postnatal development, using a tamoxifen-dependent global Cre driver. By restoring Arsb at P7, P21, and P56-P70, we determined that skeletal phenotypes can be fully rescued if Arsb restoration occurs at P7, while only achieving partial rescue at P21 and no significant rescue at P56-P70. This work has highlighted the importance of early intervention in patients with MPS VI to maximize therapeutic impact.


Asunto(s)
Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Ratones , Animales , Humanos , Mucopolisacaridosis VI/tratamiento farmacológico , Mucopolisacaridosis VI/diagnóstico , N-Acetilgalactosamina-4-Sulfatasa/genética , N-Acetilgalactosamina-4-Sulfatasa/uso terapéutico , Fenotipo , Glicosaminoglicanos , Esqueleto
4.
Nanoscale ; 15(21): 9348-9364, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37165691

RESUMEN

Enzyme replacement therapy shows remarkable clinical improvement in treating lysosomal storage disorders. However, this therapeutic approach is hampered by limitations in the delivery of the enzyme to cells and tissues. Therefore, there is an urgent, unmet clinical need to develop new strategies to enhance the enzyme delivery to diseased cells. Graphene-based materials, due to their dimensionality and favourable pattern of interaction with cells, represent a promising platform for the loading and delivery of therapeutic cargo. Herein, the potential use of graphene-based materials, including defect-free graphene with positive or negative surface charge and graphene oxide with different lateral dimensions, was investigated for the delivery of lysosomal enzymes in fibroblasts derived from patients with Mucopolysaccharidosis VI and Pompe disease. We report excellent biocompatibility of all graphene-based materials up to a concentration of 100 µg mL-1 in the cell lines studied. In addition, a noticeable difference in the uptake profile of the materials was observed. Neither type of graphene oxide was taken up by the cells to a significant extent. In contrast, the two types of graphene were efficiently taken up, localizing in the lysosomes. Furthermore, we demonstrate that cationic graphene flakes can be used as carriers for arylsulfatase B enzyme, for the delivery of the lacking enzyme to the lysosomes of Mucopolysaccharidosis VI fibroblasts. Arylsulfatase B complexed with cationic graphene flakes not only retained the enzymatic activity, but also exerted biological effects almost twice as high as arylsulfatase B alone in the clearance of the substrate in Mucopolysaccharidosis VI fibroblasts. This study lays the groundwork for the potential use of graphene-based materials as carriers for enzyme replacement therapy in lysosomal storage disorders.


Asunto(s)
Grafito , Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Humanos , Grafito/metabolismo , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , Mucopolisacaridosis VI/metabolismo , Fibroblastos , Lisosomas/metabolismo
5.
Minerva Pediatr (Torino) ; 75(2): 243-252, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-32748606

RESUMEN

BACKGROUND: Maroteaux-Lamy disease (MPS Type VI) is an autosomal recessive lysosomal storage disorder. Skeletal abnormalities are vast. Early recognition may facilitate timely diagnosis and intervention, leading to improved patient outcomes. The most challenging is when patients manifest a constellation of craniocervical and articular deformities with variable age of onset. METHODS: We collected 15 patients with MPS VI (aged from 6 years-58 years). From within our practice in Pediatric Orthopedics, we present patients with MPS type VI who were found to manifest a diverse and confusing clinical presentation of hip deformities and cervical cord compression. Stem cell transplants were proposed as treatment tool and enzyme replacement therapy has been instituted in some patients. RESULTS: The spectrum of the clinical involvement in our group of patients was supported firstly via the clinical phenotype followed by assessment of the biochemical defect, which has been detected through the deficiency of N-acetylgalactosamine-4-sulfatase (arylsulphatase B) leading to increased excretion of dermatan sulphate. Secondly, through the molecular genetic results, which showed homozygous or compound heterozygous mutation in the ARSB gene on chromosome 5q14. Hip replacements and decompression operations have been performed to restore function and to alleviate pain in the former and life saving procedure in the latter. CONCLUSIONS: The efforts in searching for the etiological diagnosis in patients with skeletal dysplasia/MPSs has not been rewarding as many had anticipated. This emerged from several facts such as improper clinical documentation, missing diagnostic pointers in radiographic interpretations, limited knowledge in skeletal dysplasia and its variants, and the reliance on underpowered studies. Physicians and radiologists are required to appreciate and assess the diverse phenotypic and the radiographic variability of MPS VI. The importance of considering MPS in the differential diagnosis of other forms skeletal dysplasia is mandatory. Finally, we stress that the value of early diagnosis is to overcome dreadful complications.


Asunto(s)
Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Humanos , Edad de Inicio , Mucopolisacaridosis VI/diagnóstico , Mucopolisacaridosis VI/genética , N-Acetilgalactosamina-4-Sulfatasa/química , N-Acetilgalactosamina-4-Sulfatasa/genética , Mutación , Fenotipo
6.
Int J Mol Sci ; 23(21)2022 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-36361933

RESUMEN

The enzyme N-acetylgalactosamine-4-sulfatase (Arylsulfatase B; ARSB) was originally identified as a lysosomal enzyme which was deficient in Mucopolysaccharidosis VI (MPS VI; Maroteaux-Lamy Syndrome). The newly directed attention to the impact of ARSB in human pathobiology indicates a broader, more pervasive effect, encompassing roles as a tumor suppressor, transcriptional mediator, redox switch, and regulator of intracellular and extracellular-cell signaling. By controlling the degradation of chondroitin 4-sulfate and dermatan sulfate by removal or failure to remove the 4-sulfate residue at the non-reducing end of the sulfated glycosaminoglycan chain, ARSB modifies the binding or release of critical molecules into the cell milieu. These molecules, such as galectin-3 and SHP-2, in turn, influence crucial cellular processes and events which determine cell fate. Identification of ARSB at the cell membrane and in the nucleus expands perception of the potential impact of decline in ARSB activity. The regulation of availability of sulfate from chondroitin 4-sulfate and dermatan sulfate may also affect sulfate assimilation and production of vital molecules, including glutathione and cysteine. Increased attention to ARSB in mammalian cells may help to integrate and deepen our understanding of diverse biological phenomenon and to approach human diseases with new insights.


Asunto(s)
Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Humanos , Sulfatos de Condroitina/metabolismo , Dermatán Sulfato , Mucopolisacaridosis VI/genética , Mucopolisacaridosis VI/metabolismo , N-Acetilgalactosamina-4-Sulfatasa/genética , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , Sulfatos
7.
Food Funct ; 13(20): 10558-10573, 2022 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-36156668

RESUMEN

Extensive phase II metabolic reactions (i.e., glucuronidation and sulfation) have resulted in low bioavailability and decreased biological effects of curcumin and quercetin. Compared to glucuronidation, information on the sulfation disposition of curcumin and quercetin is limited. In this study, we identified that BCRP and MRP4 played a critical role in the cellular excretion of curcumin-O-sulfate (C-O-S) and quercetin-O-sulfate (Q-O-S) by integrating chemical inhibition with transporter knock-down experiments. Inhibited excretion of sulfate (C-O-S and Q-O-S) caused significant reductions in cellular O-sulfation of curcumin (a maximal 74.4% reduction) and quercetin (a maximal 76.9% reduction), revealing a strong interplay of sulfation with efflux transport. It was further identified that arylsulfatase B (ARSB) played a crucial role in the regulation of cellular O-sulfation by transporters. ARSB overexpression significantly enhanced the reduction effect of MK-571 on the cellular O-sulfation (fmet) of the model compound (38.8% reduction for curcumin and 44.2% reduction for quercetin). On the contrary, ARSB knockdown could reverse the effect of MK-571 on the O-sulfation disposition of the model compound (29.7% increase for curcumin and 47.3% increase for quercetin). Taken together, ARSB has been proven to be involved in cellular O-sulfation, accounting for transporter-dependent O-sulfation of curcumin and quercetin. A better understanding of the interplay beneath metabolism and transport will contribute to the exact prediction of in vivo drug disposition and drug-drug interactions.


Asunto(s)
Curcumina , N-Acetilgalactosamina-4-Sulfatasa , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Arilsulfotransferasa , Curcumina/farmacología , Células HEK293 , Humanos , Proteínas de Transporte de Membrana , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , Proteínas de Neoplasias/metabolismo , Propionatos , Quercetina , Quinolinas , Sulfatos/metabolismo
9.
Elife ; 112022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35604022

RESUMEN

Sympathetic denervation of the heart following ischemia/reperfusion induced myocardial infarction (MI) is sustained by chondroitin sulfate proteoglycans (CSPGs) in the cardiac scar. Denervation predicts risk of sudden cardiac death in humans. Blocking CSPG signaling restores sympathetic axon outgrowth into the cardiac scar, decreasing arrhythmia susceptibility. Axon growth inhibition by CSPGs can depend on the sulfation status of the glycosaminoglycan (CS-GAG) side chains. Tandem sulfation of CS-GAGs at the 4th (4S) and 6th (6S) positions of n-acetyl-galactosamine inhibits outgrowth in several types of central neurons, but we don't know if sulfation is similarly critical during peripheral nerve regeneration. We asked if CSPG sulfation prevented sympathetic axon outgrowth after MI. Reducing 4S with the 4-sulfatase enzyme Arylsulfatase-B (ARSB) enhanced outgrowth of dissociated rat sympathetic neurons over CSPGs. Likewise, reducing 4S with ARSB restored axon outgrowth from mouse sympathetic ganglia co-cultured with cardiac scar tissue. We quantified enzymes responsible for adding and removing sulfation, and found that CHST15 (4S dependent 6-sulfotransferase) was upregulated, and ARSB was downregulated after MI. This suggests a mechanism for production and maintenance of sulfated CSPGs in the cardiac scar. We decreased 4S,6S CS-GAGs in vivo by transient siRNA knockdown of Chst15 after MI, and found that reducing 4S,6S restored tyrosine hydroxylase (TH) positive sympathetic nerve fibers in the cardiac scar. Reinnervation reduced isoproterenol induced arrhythmias. Our results suggest that modulating CSPG-sulfation after MI may be a therapeutic target to promote sympathetic nerve regeneration in the cardiac scar and reduce post-MI cardiac arrhythmias.


Asunto(s)
Infarto del Miocardio , N-Acetilgalactosamina-4-Sulfatasa , Azufre/metabolismo , Animales , Antígenos , Proteoglicanos Tipo Condroitín Sulfato/química , Cicatriz , Ratones , Regeneración Nerviosa/fisiología , Proteoglicanos , Ratas
10.
Biomaterials ; 284: 121526, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35461098

RESUMEN

Traumatic damage to the spinal cord does not spontaneously heal, often leading to permanent tissue defects. We have shown that injection of imidazole-poly(organophosphazene) hydrogel (I-5) bridges cystic cavities with the newly assembled fibronectin-rich extracellular matrix (ECM). The hydrogel-created ECM contains chondroitin sulfate proteoglycans (CSPGs), collagenous fibrils together with perivascular fibroblasts, and various fibrotic proteins, all of which could hinder axonal growth in the matrix. In an in vitro fibrotic scar model, fibroblasts exhibited enhanced sensitivity to TGF-ß1 when grown on CSPGs. To alleviate the fibrotic microenvironment, the I-5 hydrogel was equipped with an additional function by making a complex with ARSB, a human enzyme degrading CSPGs, via hydrophobic interaction. Delivery of the I-5/ARSB complex significantly diminished the fibrotic ECM components. The complex promoted serotonergic axonal growth into the hydrogel-induced matrix and enhanced serotonergic innervation of the lumbar motor neurons. Regeneration of the propriospinal axons deep into the matrix and to the lumbar spinal cord was robustly increased accompanied by improved locomotor recovery. Therefore, our dual-functional system upgraded the functionality of the hydrogel for spinal cord regeneration by creating ECM to bridge tissue defects and concurrently facilitating axonal connections through the newly assembled ECM.


Asunto(s)
N-Acetilgalactosamina-4-Sulfatasa , Traumatismos de la Médula Espinal , Regeneración de la Medula Espinal , Animales , Axones/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Preparaciones de Acción Retardada/metabolismo , Humanos , Hidrogeles/química , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , N-Acetilgalactosamina-4-Sulfatasa/uso terapéutico , Regeneración Nerviosa/fisiología , Ratas , Ratas Sprague-Dawley , Médula Espinal
11.
Brasília; CONITEC; fev. 2022.
No convencional en Portugués | BRISA/RedTESA | ID: biblio-1444516

RESUMEN

CONDIÇÃO CLÍNICA: Mucopolissacaridose tipo VI (MPS-VI) A MPS-VI, ou Síndrome Maroteaux-Lamy, é uma doença genética rara, causada pela atividade deficiente da enzima Nacetilgalactosamina-4-sulfatase ou arilsulfatase B(ASB), responsável pela degradação do glicosaminoglicano (GAGs) dermatan sulfato (DS), resultando no acúmulo desse componente nos lisossomos de múltiplos tecidos do corpo. Pacientes com a forma rapidamente progressiva apresentam mortalidade precoce no início da fase adulta, geralmente por insuficiência cardiopulmonar. Nas formas com progressão lenta, as morbidades também podem ser significativas com mortalidade em torno da terceira a quinta década de vida.O Protocolo Clínico e Diretrizes Terapêuticas (PCDT) da doença inclui intervenções gerais e específicas. Entre estas, encontram-se o transplante de células-tronco hematopoéticas (TCTH) e a terapia de reposição enzimática com galsulfase. TECNOLOGIA: galsulfase (Naglazyme®) Medicamento que atua como terapia de reposição enzimática específica para pacientes com MPS-VI. A dose recomendada é de 1 mg por kg de peso corporal, administrados uma vez por semana em infusão intravenosa. HISTÓRICO DA INCORPORAÇÃO: Incorporação em dezembro de 2018, condicionada a reavaliação em três anos. Relatório Técnico da Conitec nº 412, de dezembro de 2018. Portaria de incorporação SCTIE/MS nº 83, de 20 de dezembro de 2018. PCDT publicado pela Portaria Conjunta SAS/SCTIE/MS nº 20, de 05 de dezembro de 2019. Inclusão do procedimento na Tabela de Procedimentos, Medicamentos, Órteses/Próteses e Materiais Especiais do SUS pela Portaria SAES/MS nº 1.020, de 22 de outubro de 2020. EVIDÊNCIAS CLÍNICAS: Melhora observada em mortalidade, resistência física (teste de caminhada e subida de escadas), excreção urinária de glicosaminoglicanos, e boa tolerabilidade com o medicamento. Foram incluídos 23 estudos, com pacientes acompanhados por até 15 anos. Cinco estudos foram realizados com a participação de centros ou autores brasileiros. Nenhum ensaio clínico randomizado foi publicado após a incorporação do medicamento. UTILIZAÇÃO DO MEDICAMENTO NO SUS: Início da dispensação em janeiro de 2021, com o atendimento de 183 pacientes até novembro de 2021. Aquisição do medicamento realizada com preço unitário de R$ 4.293,57. Não há informações clínicas disponíveis sobre o desempenho do medicamento no contexto do SUS. No momento da incorporação, previu-se o atendimento a 183 pacientes no primeiro ano de incorporação, com preço proposto de R$ 3.986,60. IMPACTO ORÇAMENTÁRIO OBSERVADO: R$ 96.867.232,77 entre janeiro e novembro de 2021. Previu-se na incorporação um impacto orçamentário de R$ 255.792.054 no primeiro ano. O valor observado foi inferior ao esperado devido ao acesso gradual dos pacientes ao longo dos meses. MONITORAMENTO DO HORIZONTE TECNOLÓGICO: Não foram identificadas tecnologias em fase de pesquisa clínica 3 ou 4 em andamento para o tratamento da MPS-VI. Há três tecnologias em fase 1 e 2 de pesquisa clínica: Odiparcil, de uso oral, em fase 2a; Lysosan™, de administração subcutânea, em fase 1/2; e AAV2/8.TBG.hARSB, administrado por via intravenosa, em fase ½. ESTUDO DE SITUAÇÃO PATENTÁRIA: Foi identificada a patente PI0316039 associada ao medicamento, de titularidade da Biomarin, com vigência até 2023. RECOMENDAÇÃO DA CONITEC: O Plenário da Conitec, em sua 105ª Reunião Ordinária, no dia 10 de fevereiro de 2021, recomendou a continuidade do monitoramento da galsulfase para mucopolissacaridose tipo VI, considerando a implementação recente da tecnologia no SUS.


Asunto(s)
Humanos , Mucopolisacaridosis VI/tratamiento farmacológico , N-Acetilgalactosamina-4-Sulfatasa/uso terapéutico , Brasil , Eficacia , Análisis Costo-Beneficio/economía , Proyectos de Desarrollo Tecnológico e Innovación
12.
Brasília; CONITEC; fev. 2022.
No convencional en Portugués | BRISA/RedTESA | ID: biblio-1452108

RESUMEN

CONDIÇÃO CLÍNICA: Mucopolissacaridose tipo VI (MPS-VI) A MPS-VI, ou Síndrome Maroteaux-Lamy, é uma doença genética rara, causada pela atividade deficiente da enzima Nacetilgalactosamina-4-sulfatase ou arilsulfatase B(ASB), responsável pela degradação do glicosaminoglicano (GAGs) dermatan sulfato (DS), resultando no acúmulo desse componente nos lisossomos de múltiplos tecidos do corpo. Pacientes com a forma rapidamente progressiva apresentam mortalidade precoce no início da fase adulta, geralmente por insuficiência cardiopulmonar. Nas formas com progressão lenta, as morbidades também podem ser significativas com mortalidade em torno da terceira a quinta década de vida.O Protocolo Clínico e Diretrizes Terapêuticas (PCDT) da doença inclui intervenções gerais e específicas. Entre estas, encontram-se o transplante de células-tronco hematopoéticas (TCTH) e a terapia de reposição enzimática com galsulfase. TECNOLOGIA: galsulfase (Naglazyme®) Medicamento que atua como terapia de reposição enzimática específica para pacientes com MPS-VI. A dose recomendada é de 1 mg por kg de peso corporal, administrados uma vez por semana em infusão intravenosa. HISTÓRICO DA INCORPORAÇÃO: Incorporação em dezembro de 2018, condicionada a reavaliação em três anos. Relatório Técnico da Conitec nº 412, de dezembro de 2018. Portaria de incorporação SCTIE/MS nº 83, de 20 de dezembro de 2018. PCDT publicado pela Portaria Conjunta SAS/SCTIE/MS nº 20, de 05 de dezembro de 2019. Inclusão do procedimento na Tabela de Procedimentos, Medicamentos, Órteses/Próteses e Materiais Especiais do SUS pela Portaria SAES/MS nº 1.020, de 22 de outubro de 2020. EVIDÊNCIAS CLÍNICAS: Melhora observada em mortalidade, resistência física (teste de caminhada e subida de escadas), excreção urinária de glicosaminoglicanos, e boa tolerabilidade com o medicamento. Foram incluídos 23 estudos, com pacientes acompanhados por até 15 anos. Cinco estudos foram realizados com a participação de centros ou autores brasileiros. Nenhum ensaio clínico randomizado foi publicado após a incorporação do medicamento. UTILIZAÇÃO DO MEDICAMENTO NO SUS: Início da dispensação em janeiro de 2021, com o atendimento de 183 pacientes até novembro de 2021. Aquisição do medicamento realizada com preço unitário de R$ 4.293,57. Não há informações clínicas disponíveis sobre o desempenho do medicamento no contexto do SUS. No momento da incorporação, previu-se o atendimento a 183 pacientes no primeiro ano de incorporação, com preço proposto de R$ 3.986,60. IMPACTO ORÇAMENTÁRIO OBSERVADO: R$ 96.867.232,77 entre janeiro e novembro de 2021. Previu-se na incorporação um impacto orçamentário de R$ 255.792.054 no primeiro ano. O valor observado foi inferior ao esperado devido ao acesso gradual dos pacientes ao longo dos meses. MONITORAMENTO DO HORIZONTE TECNOLÓGICO: Não foram identificadas tecnologias em fase de pesquisa clínica 3 ou 4 em andamento para o tratamento da MPS-VI. Há três tecnologias em fase 1 e 2 de pesquisa clínica: Odiparcil, de uso oral, em fase 2a; Lysosan™, de administração subcutânea, em fase 1/2; e AAV2/8.TBG.hARSB, administrado por via intravenosa, em fase 1/2. ESTUDO DE SITUAÇÃO PATENTÁRIA: Foi identificada a patente PI0316039 associada ao medicamento, de titularidade da Biomarin, com vigência até 2023. RECOMENDAÇÃO DA CONITEC: O Plenário da Conitec, em sua 105ª Reunião Ordinária, no dia 10 de fevereiro de 2021, recomendou a continuidade do monitoramento da galsulfase para mucopolissacaridose tipo VI, considerando a implementação recente da tecnologia no SUS.


Asunto(s)
Humanos , Mucopolisacaridosis VI/tratamiento farmacológico , N-Acetilgalactosamina-4-Sulfatasa/uso terapéutico , Brasil , Eficacia , Análisis Costo-Beneficio , Proyectos de Desarrollo Tecnológico e Innovación
13.
Iowa Orthop J ; 42(2): 122-127, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36601232

RESUMEN

Background: Mucopolysaccharidoses (MPS) are lysosomal storage disorders characterized by abnormal deposition of glycosaminoglycans (GAGs) in tissues. In type VI MPS, otherwise known as Maroteaux-Lamy syndrome, the defect is in the enzyme N-acetylgalactosamine-4-sulfatase. Thoracolumbar kyphosis results from GAG deposition, leading to incompetence of posterior ligamentous structures as well as poor trunk control. Though neurologic symptoms from canal compression due to deformity and hypertrophy of tissues have been described, occasionally requiring surgical decompression, there has not been a prior report of late onset of symptoms in a previously neurologically intact patient following surgery to correct spine deformity. Methods: The case reviewed is a 14 year old girl with mucopolysaccharidosis type VI underwent anterior release and posterior instrumentation for correction of severe progressive lumbar kyphosis. Postoperatively she developed delayed onset of profound lower extremity weakness and underwent urgent wide laminectomies and resection of thickened ligamentum flavum. At 1 year follow-up, she had near complete neurologic recovery. Conclusion: Patients with mucopolysacchari-doses are at significant risk for neurologic compromise both as part of the natural history of the disease, and as a risk of deformity correction. The surgeon must consider the pathologic thickening of tissues surrounding the spinal cord when planning surgery. Level of Evidence: IV.


Asunto(s)
Cifosis , Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Femenino , Humanos , Adolescente , Mucopolisacaridosis VI/complicaciones , Mucopolisacaridosis VI/cirugía , Cifosis/etiología , Cifosis/cirugía
14.
Mol Genet Metab ; 135(2): 143-153, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34417096

RESUMEN

Maroteaux - Lamy syndrome (mucopolysaccharidosis type VI, MPS VI) is a lysosomal storage disease resulting from insufficient enzymatic activity for degradation of the specific glycosaminoglycans (GAG) chondroitin sulphate (CS) and dermatan sulphate (DS). Among the most pronounced MPS VI clinical manifestations caused by cellular accumulation of excess CS and DS are eye disorders, in particular those that affect the cornea. Ocular manifestations are not treated by the current standard of care, enzyme replacement therapy (ERT), leaving patients with a significant unmet need. Using in vitro and in vivo models, we previously demonstrated the potential of the ß-D-xyloside, odiparcil, as an oral GAG clearance therapy for MPS VI. Here, we characterized the eye phenotypes in MPS VI arylsulfatase B deficient mice (Arsb-) and studied the effects of odiparcil treatment in early and established disease models. Severe levels of opacification and GAG accumulation were detected in the eyes of MPS VI Arsb- mice. Histological examination of MPS VI Arsb- eyes showed an aggregate of corneal phenotypes, including reduction in the corneal epithelium thickness and number of epithelial cell layers, and morphological malformations in the stroma. In addition, colloidal iron staining showed specifically GAG accumulation in the cornea. Orally administered odiparcil markedly reduced GAG accumulation in the eyes of MPS VI Arsb- mice in both disease models and restored the corneal morphology (epithelial layers and stromal structure). In the early disease model of MPS VI, odiparcil partially reduced corneal opacity area, but did not affect opacity area in the established model. Analysis of GAG types accumulating in the MPS VI Arsb- eyes demonstrated major contribution of DS and CS, with some increase in heparan sulphate (HS) as well and all were reduced with odiparcil treatment. Taken together, we further reveal the potential of odiparcil to be an effective therapy for eye phenotypes associated with MPS VI disease.


Asunto(s)
Oftalmopatías/tratamiento farmacológico , Glicósidos , Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Animales , Modelos Animales de Enfermedad , Ojo/patología , Oftalmopatías/genética , Glicósidos/uso terapéutico , Humanos , Ratones , Mucopolisacaridosis VI/tratamiento farmacológico , Mucopolisacaridosis VI/genética , N-Acetilgalactosamina-4-Sulfatasa/genética , Fenotipo
15.
Pathobiology ; 89(2): 81-91, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34788765

RESUMEN

INTRODUCTION: The potential role of accumulation of chondroitin sulfates (CSs) in the pathobiology of COVID-19 has not been examined. Accumulation may occur by increased synthesis or by decline in activity of the enzyme arylsulfatase B (ARSB; N-acetylgalactosamine-4-sulfatase) which requires oxygen for activity. METHODS: Immunostaining of lung tissue from 28 patients who died due to COVID-19 infection was performed for CS, ARSB, and carbohydrate sulfotransferase (CHST)15. Measurements of mRNA expression of CHST15 and CHST11, sulfotransferase activity, and total sulfated glycosaminoglycans (GAGs) were determined in human vascular smooth muscle cells following angiotensin (Ang) II treatment. RESULTS: CS immunostaining showed increase in intensity and distribution, and immunostaining of ARSB was diminished in COVID-19 compared to normal lung tissue. CHST15 immunostaining was prominent in vascular smooth muscle cells associated with diffuse alveolar damage due to COVID-19 or other causes. Expression of CHST15 and CHST11 which are required for synthesis of CSE and chondroitin 4-sulfate, total sulfated GAGs, and sulfotransferase activity was significantly increased following AngII exposure in vascular smooth muscle cells. Expression of Interleukin-6 (IL-6), a mediator of cytokine storm in COVID-19, was inversely associated with ARSB expression. DISCUSSION/CONCLUSION: Decline in ARSB and resulting increases in CS may contribute to the pathobiology of COVID-19, as IL-6 does. Increased expression of CHSTs following activation of Ang-converting enzyme 2 may lead to buildup of CSs.


Asunto(s)
COVID-19 , N-Acetilgalactosamina-4-Sulfatasa , Insuficiencia Respiratoria , Sulfatos de Condroitina/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Glicoproteínas de Membrana , N-Acetilgalactosamina-4-Sulfatasa/genética , N-Acetilgalactosamina-4-Sulfatasa/metabolismo , Sulfotransferasas
16.
J Inherit Metab Dis ; 45(2): 340-352, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34910312

RESUMEN

Mucopolysaccharidosis (MPS) disorders are a group of rare, progressive lysosomal storage diseases characterized by the accumulation of glycosaminoglycans (GAGs) and classified according to the deficient enzyme. Enzyme replacement therapy (ERT) of MPS VI has limited effects on ophthalmic, cardiovascular, and skeletal systems. Odiparcil is an orally available small molecule that results in the synthesis of odiparcil-linked GAGs facilitating their excretion and reducing cellular and tissue GAG accumulation. Improve MPS treatment was a Phase 2a study of the safety, pharmacokinetics/pharmacodynamics, and efficacy of two doses of odiparcil in patients with MPS VI. The core study was a 26-week, randomized, double-blind, placebo-controlled trial in patients receiving ERT and an open-label, noncomparative, single-dose cohort not receiving ERT. Patients aged ≥ 16 years receiving ERT were randomized to odiparcil 250 or 500 mg twice daily or placebo. Patients without ERT received odiparcil 500 mg twice daily. Of 20 patients enrolled, 13 (65.0%) completed the study. Odiparcil increased total urine GAGs (uGAGs), chondroitin sulfate, and dermatan sulfate concentrations. A linear increase in uGAG levels and odiparcil exposure occurred with increased odiparcil dose. Odiparcil demonstrated a good safety and tolerability profile. Individual analyses found more improvements in pain, corneal clouding, cardiac, vascular, and respiratory functions in the odiparcil groups vs placebo. This study confirmed the mechanism of action and established the safety of odiparcil with clinical beneficial effects after only a short treatment duration in an advanced stage of disease. Further assessment of odiparcil in younger patients is needed.


Asunto(s)
Mucopolisacaridosis , Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Terapia de Reemplazo Enzimático/métodos , Glicosaminoglicanos , Glicósidos/uso terapéutico , Humanos , Mucopolisacaridosis VI/tratamiento farmacológico , N-Acetilgalactosamina-4-Sulfatasa/uso terapéutico
17.
Int J Mol Sci ; 22(24)2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34948256

RESUMEN

Mucopolysaccharidosis type VI, or Maroteaux-Lamy syndrome, is a rare, autosomal recessive genetic disease, mainly affecting the pediatric age group. The disease is due to pathogenic variants of the ARSB gene, coding for the lysosomal hydrolase N-acetylgalactosamine 4-sulfatase (arylsulfatase B, ASB). The enzyme deficit causes a pathological accumulation of the undegraded glycosaminoglycans dermatan-sulphate and chondroitin-sulphate, natural substrates of ASB activity. Intracellular and extracellular deposits progressively take to a pathological scenario, often severe, involving most organ-systems and generally starting from the osteoarticular apparatus. Neurocognitive and behavioral abilities, commonly described as maintained, have been actually investigated by few studies. The disease, first described in 1963, has a reported prevalence between 0.36 and 1.3 per 100,000 live births across the continents. With this paper, we wish to contribute an updated overview of the disease from the clinical, diagnostic, and therapeutic sides. The numerous in vitro and in vivo preclinical studies conducted in the last 10-15 years to dissect the disease pathogenesis, the efficacy of the available therapeutic treatment (enzyme replacement therapy), as well as new therapies under study are here described. This review also highlights the need to identify new disease biomarkers, potentially speeding up the diagnostic process and the monitoring of therapeutic efficacy.


Asunto(s)
Mucopolisacaridosis VI/genética , Mucopolisacaridosis VI/fisiopatología , Sulfatos de Condroitina/uso terapéutico , Terapia de Reemplazo Enzimático , Glicosaminoglicanos/uso terapéutico , Humanos , Mucopolisacaridosis VI/terapia , N-Acetilgalactosamina-4-Sulfatasa/genética
18.
Orphanet J Rare Dis ; 16(1): 500, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857033

RESUMEN

BACKGROUND: Research about pediatric patients' perspective on mucopolysaccharidosis type VI (MPS VI) and its impact on daily life is limited. We aimed to identify the disease concepts of interest that most impact function and day-to-day life of pediatric patients with MPS VI, and to consider clinical outcome assessments (COAs) that may potentially measure meaningful improvements in these concepts. METHODS: Potential focus group participants were identified by the National MPS Society (USA) and invited to participate if they self-reported a clinician-provided diagnosis of MPS VI and were 4 to 18 years, receiving enzyme replacement therapy (ERT), and available to attend a 1-day focus group with their caregiver in Dallas, TX, USA. The focus group consisted of a series of polling and open-ended concept elicitation questions and a cognitive debriefing session. The discussion was audio recorded, transcribed verbatim, and analyzed to identify disease concepts of interest and functional impacts most relevant to participants. RESULTS: Overall, caregivers (n = 9) and patients with MPS VI (n = 9) endorsed that although their children/they receive ERT, residual symptoms exist and impact health-related quality of life. The key disease concepts of interest identified were impaired mobility, upper extremity and fine motor deficits, pain, and fatigue. Pain was unanimously reported by all patients across many areas of the body and impacted daily activity. Key disease concepts were mapped to a selection of pediatric COAs including generic measures such as PROMIS®, PODCI, CHAQ, and PedsQL™. Caregivers endorsed the relevance of PODCI and PROMIS Upper Extremity, Mobility, and Pain items and all patients completed the NIH Toolbox Pegboard Dexterity Test. Additional COAs that aligned with the disease concepts included range of motion, the 2- and 6-min walk tests, timed stair climbs, Bruininks-Oseretsky Test of Motor Proficiency, 2nd edition, grip strength, pain visual analog scale, and the Faces Pain Scale-Revised. CONCLUSION: An MPS VI focus group of pediatric patients and their caregivers identified impaired mobility, upper extremity and fine motor deficits, pain, and fatigue as key disease concepts of interest. These disease concepts were mapped to existing pediatric COAs, which were provided to the group for endorsement of their relevance.


Asunto(s)
Actividades Cotidianas , Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Calidad de Vida , Cuidadores , Niño , Terapia de Reemplazo Enzimático , Grupos Focales , Humanos , Trastornos de la Destreza Motora/etiología , Mucopolisacaridosis VI/complicaciones , Mucopolisacaridosis VI/tratamiento farmacológico , Dolor/etiología , Resultado del Tratamiento
19.
Orphanet J Rare Dis ; 16(1): 438, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34666789

RESUMEN

BACKGROUND: The objective of this study was to describe clinical manifestations and events of patients with mucopolysaccharidosis (MPS) VI in Turkey who are treated with galsulfase enzyme replacement therapy (ERT). Clinical data of 14 children with MPS VI who were followed up at the Department of Pediatrics of the Gazi University Faculty of Medicine in Ankara, Turkey were retrospectively collected from the patients' medical records. Patients were selected based on availability of a pre-ERT baseline and follow-up clinical data for a similar period of time (1.9-3.2 years). Event data (occurrence of acute clinical events, onset of chronic events, surgeries) collected during hospital visits and telemedicine were available for up to 10 years after initiation of ERT (2.5-10 years). RESULTS: Age at initiation of ERT ranged from 2.8 to 15.8 years (mean age 7.5 years). All patients presented with reduced endurance and skeletal abnormalities (dysostosis multiplex) on radiography. Other common clinical manifestations were cardiac valve disease (N = 13), short stature (N = 11), cranial abnormalities on MRI (N = 10), spinal abnormalities on MRI (N = 7), and mild cognitive impairment (N = 6). School attendance was generally poor, and several patients had urinary incontinence. After 1.9 to 3.2 years of ERT, most patients showed improvements in endurance in the 6-min walk test and 3-min stair climb tests; the frequency of urinary incontinence decreased. ERT did not seem to prevent progression of cardiac valve disease, eye disorders, hearing loss, or bone disease. Long-term event-based data showed a high incidence of respiratory tract infections, adenotonsillectomy/adenoidectomy, reduced sleep quality, sleep apnea, and depression before initiation of ERT. The number of events tended to remain stable or decrease in all patients over 2.5-10 years follow-up. However, the nature of the events shifted over time, with a reduction in the frequency of respiratory tract infections and sleep problems and an increase in ophthalmologic events, ear tube insertions, and depression. CONCLUSIONS: This case series shows the high disease burden of the MPS VI population in Turkey and provides a unique insight into their clinical journey based on real-life clinical and event-based data collected before and after initiation of ERT.


Asunto(s)
Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Adolescente , Niño , Preescolar , Terapia de Reemplazo Enzimático , Humanos , Mucopolisacaridosis VI/tratamiento farmacológico , Radiografía , Estudios Retrospectivos , Turquía
20.
Cochrane Database Syst Rev ; 9: CD009806, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34533215

RESUMEN

BACKGROUND: Mucopolysaccharidosis type VI (MPS VI) or Maroteaux-Lamy syndrome is a rare genetic disorder caused by the deficiency of arylsulphatase B. The resultant accumulation of dermatan sulphate causes lysosomal damage. The clinical symptoms are related to skeletal dysplasia (i.e. short stature and degenerative joint disease). Other manifestations include cardiac disease, impaired pulmonary function, ophthalmological complications, hepatosplenomegaly, sinusitis, otitis, hearing loss and sleep apnea. Intellectual impairment is generally absent. Clinical manifestation is typically by two or three years of age; however, slowly progressive cases may not present until adulthood. Enzyme replacement therapy (ERT) with galsulfase is considered a new approach for treating MPS VI. OBJECTIVES: To evaluate the effectiveness and safety of treating MPS VI by ERT with galsulfase compared to other interventions, placebo or no intervention. SEARCH METHODS: Eletronic searches were performed on the Cystic Fibrosis and Genetic Disorders Group's Inborn Errors of Metabolism Trials Register. Date of the latest search:  09 June 2021. Further searches of the following databases were also performed: CENTRAL, MEDLINE, LILACS, the Journal of Inherited Metabolic Disease, the World Health Organization International Clinical Trials Registry Platform and ClinicalTrials.gov. Date of the latest search: 20 August 2021. SELECTION CRITERIA: Randomized and quasi-randomized controlled clinical studies of ERT with galsulfase compared to other interventions or placebo. DATA COLLECTION AND ANALYSIS: Two authors independently screened the studies, assessed the risk of bias, extracted data and assessed the certainty of the the evidence using the GRADE criteria. MAIN RESULTS: One study was included involving 39 participants who received either ERT with galsulfase (recombinant human arylsulphatase B) or placebo. This small study was considered overall to have an unclear risk of bias in relation to the design and implementation of the study, since the authors did not report how both the allocation generation and concealment were performed. Given the very low certainty of the evidence, we are uncertain whether at 24 weeks there was a difference between groups in relation to the 12-minute walk test, mean difference (MD) of 92.00 meters (95% confidence interval (CI) 11.00 to 172.00), or the three-minute stair climb, MD 5.70 (95% CI -0.10 to 11.50). In relation to respiratory tests, we are uncertain whether galsulfase makes any difference as compared to placebo in forced vital capacity in litres (FVC (L) (absolute change in baseline), given the very low certainty of the evidence. Cardiac function was not reported in the included study. We found that galsulfase, as compared to placebo, may decrease urinary glycosaminoglycan levels at 24 weeks, MD -227.00 (95% CI -264.00 to -190.00) (low-certainty evidence). We are uncertain whether there are differences between the galsulfase and placebo groups in relation to adverse events (very low-certainty evidence). In general, the dose of galsulfase was well tolerated and there were no differences between groups. These events include drug-related adverse events, serious and severe adverse events, those during infusion, drug-related adverse events during infusion, and deaths. More infusion-related reactions were observed in the galsulfase group and were managed with interruption or slowing of infusion rate or administration of antihistamines or corticosteroids drugs. No deaths occurred during the study.   AUTHORS' CONCLUSIONS: The results of this review are based only on one small study (a 24-week randomised phase of the study and prior to the open-label extension). We are uncertain whether galsulfase is more effective than placebo, for treating people with MPS VI, in relation to the 12-minute walk test or the three-minute stair climb, as the certainty of the evidence has been assessed as very low. We found that galsulfase may reduce urinary glycosaminoglycans levels. We are also uncertain whether there are any differences between treatment groups in relation to cardiac or pulmonary functions, liver or spleen volume, overnight apnea-hypopnea, height and weight, quality of life and adverse effects. Further studies are needed to obtain more information on the long-term effectiveness and safety of ERT with galsulfase.


Asunto(s)
Mucopolisacaridosis VI , N-Acetilgalactosamina-4-Sulfatasa , Adulto , Terapia de Reemplazo Enzimático , Humanos , Mucopolisacaridosis VI/tratamiento farmacológico , Calidad de Vida , Proteínas Recombinantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...