Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 107(6): 1044-1061, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33159882

RESUMO

Heparan sulfate belongs to the group of glycosaminoglycans (GAGs), highly sulfated linear polysaccharides. Heparan sulfate 2-O-sulfotransferase 1 (HS2ST1) is one of several specialized enzymes required for heparan sulfate synthesis and catalyzes the transfer of the sulfate groups to the sugar moiety of heparan sulfate. We report bi-allelic pathogenic variants in HS2ST1 in four individuals from three unrelated families. Affected individuals showed facial dysmorphism with coarse face, upslanted palpebral fissures, broad nasal tip, and wide mouth, developmental delay and/or intellectual disability, corpus callosum agenesis or hypoplasia, flexion contractures, brachydactyly of hands and feet with broad fingertips and toes, and uni- or bilateral renal agenesis in three individuals. HS2ST1 variants cause a reduction in HS2ST1 mRNA and decreased or absent heparan sulfate 2-O-sulfotransferase 1 in two of three fibroblast cell lines derived from affected individuals. The heparan sulfate synthesized by the individual 1 cell line lacks 2-O-sulfated domains but had an increase in N- and 6-O-sulfated domains demonstrating functional impairment of the HS2ST1. As heparan sulfate modulates FGF-mediated signaling, we found a significantly decreased activation of the MAP kinases ERK1/2 in FGF-2-stimulated cell lines of affected individuals that could be restored by addition of heparin, a GAG similar to heparan sulfate. Focal adhesions in FGF-2-stimulated fibroblasts of affected individuals concentrated at the cell periphery. Our data demonstrate that a heparan sulfate synthesis deficit causes a recognizable syndrome and emphasize a role for 2-O-sulfated heparan sulfate in human neuronal, skeletal, and renal development.


Assuntos
Osso e Ossos/anormalidades , Corpo Caloso/patologia , Deficiências do Desenvolvimento/genética , Rim/anormalidades , Sulfotransferases/genética , Adolescente , Alelos , Biópsia , Criança , Pré-Escolar , Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Saúde da Família , Feminino , Fibroblastos/metabolismo , Variação Genética , Heparitina Sulfato/metabolismo , Humanos , Ácido Idurônico/farmacologia , Recém-Nascido , Masculino , Linhagem , Fenótipo , Síndrome , Anormalidades Urogenitais/genética
2.
Int J Mol Sci ; 24(4)2023 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-36835644

RESUMO

In vivo models of acute myeloid leukemia (AML) are low throughput, and standard liquid culture models fail to recapitulate the mechanical and biochemical properties of the extracellular matrix-rich protective bone marrow niche that contributes to drug resistance. Candidate drug discovery in AML requires advanced synthetic platforms to improve our understanding of the impact of mechanical cues on drug sensitivity in AML. By use of a synthetic, self-assembling peptide hydrogel (SAPH) of modifiable stiffness and composition, a 3D model of the bone marrow niche to screen repurposed FDA-approved drugs has been developed and utilized. AML cell proliferation was dependent on SAPH stiffness, which was optimized to facilitate colony growth. Three candidate FDA-approved drugs were initially screened against the THP-1 cell line and mAF9 primary cells in liquid culture, and EC50 values were used to inform drug sensitivity assays in the peptide hydrogel models. Salinomycin demonstrated efficacy in both an 'early-stage' model in which treatment was added shortly after initiation of AML cell encapsulation, and an 'established' model in which time-encapsulated cells had started to form colonies. Sensitivity to Vidofludimus treatment was not observed in the hydrogel models, and Atorvastatin demonstrated increased sensitivity in the 'established' compared to the 'early-stage' model. AML patient samples were equally sensitive to Salinomycin in the 3D hydrogels and partially sensitive to Atorvastatin. Together, this confirms that AML cell sensitivity is drug- and context-specific and that advanced synthetic platforms for higher throughput are valuable tools for pre-clinical evaluation of candidate anti-AML drugs.


Assuntos
Hidrogéis , Leucemia Mieloide Aguda , Humanos , Hidrogéis/uso terapêutico , Atorvastatina/uso terapêutico , Leucemia Mieloide Aguda/metabolismo , Medula Óssea/metabolismo , Peptídeos/uso terapêutico
3.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34209670

RESUMO

The glycosaminoglycan, heparan sulphate (HS), orchestrates many developmental processes. Yet its biological role has not yet fully been elucidated. Small molecule chemical inhibitors can be used to perturb HS function and these compounds provide cheap alternatives to genetic manipulation methods. However, existing chemical inhibition methods for HS also interfere with chondroitin sulphate (CS), complicating data interpretation of HS function. Herein, a simple method for the selective inhibition of HS biosynthesis is described. Using endogenous metabolic sugar pathways, Ac4GalNAz produces UDP-GlcNAz, which can target HS synthesis. Cell treatment with Ac4GalNAz resulted in defective chain elongation of the polymer and decreased HS expression. Conversely, no adverse effect on CS production was observed. The inhibition was transient and dose-dependent, affording rescue of HS expression after removal of the unnatural azido sugar. The utility of inhibition is demonstrated in cell culture and in whole organisms, demonstrating that this small molecule can be used as a tool for HS inhibition in biological systems.


Assuntos
Vias Biossintéticas/efeitos dos fármacos , Sulfatos de Condroitina/biossíntese , Heparitina Sulfato/biossíntese , Animais , Células CHO , Metabolismo dos Carboidratos/efeitos dos fármacos , Sulfatos de Condroitina/química , Cricetulus , Descoberta de Drogas , Glicosaminoglicanos/biossíntese , Heparitina Sulfato/química
4.
J Biol Chem ; 291(36): 18600-18607, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27387504

RESUMO

Analysis of heparan sulfate synthesized by HEK 293 cells overexpressing murine NDST1 and/or NDST2 demonstrated that the amount of heparan sulfate was increased in NDST2- but not in NDST1-overexpressing cells. Altered transcript expression of genes encoding other biosynthetic enzymes or proteoglycan core proteins could not account for the observed changes. However, the role of NDST2 in regulating the amount of heparan sulfate synthesized was confirmed by analyzing heparan sulfate content in tissues isolated from Ndst2(-/-) mice, which contained reduced levels of the polysaccharide. Detailed disaccharide composition analysis showed no major structural difference between heparan sulfate from control and Ndst2(-/-) tissues, with the exception of heparan sulfate from spleen where the relative amount of trisulfated disaccharides was lowered in the absence of NDST2. In vivo transcript expression levels of the heparan sulfate-polymerizing enzymes Ext1 and Ext2 were also largely unaffected by NDST2 levels, pointing to a mode of regulation other than increased gene transcription. Size estimation of heparan sulfate polysaccharide chains indicated that increased chain lengths in NDST2-overexpressing cells alone could explain the increased heparan sulfate content. A model is discussed where NDST2-specific substrate modification stimulates elongation resulting in increased heparan sulfate chain length.


Assuntos
Amidoidrolases/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Heparitina Sulfato/biossíntese , Modelos Biológicos , Sulfotransferases/biossíntese , Transcrição Gênica/fisiologia , Amidoidrolases/genética , Animais , Células HEK293 , Heparitina Sulfato/genética , Humanos , Camundongos , Camundongos Knockout , N-Acetilglucosaminiltransferases/biossíntese , N-Acetilglucosaminiltransferases/genética , Sulfotransferases/genética
5.
J Cell Sci ; 127(Pt 1): 158-71, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24190885

RESUMO

Here, we show that epithelial-mesenchymal status influences how cells deposit extracellular matrix. Retinal pigmented epithelial (RPE) cells that expressed high levels of E-cadherin and had cell-cell junctions rich in zona occludens (ZO)-1, ß-catenin and heparan sulfate, required syndecan-4 but not fibronectin or protein kinase C α (PKCα) to assemble extracellular matrix (fibrillin microfibrils and perlecan). In contrast, RPE cells that strongly expressed mesenchymal smooth muscle α-actin but little ZO-1 or E-cadherin, required fibronectin (like fibroblasts) and PKCα, but not syndecan-4. Integrins α5ß1 and/or α8ß1 and actomyosin tension were common requirements for microfibril deposition, as was heparan sulfate biosynthesis. TGFß, which stimulates epithelial-mesenchymal transition, altered gene expression and overcame the dependency on syndecan-4 for microfibril deposition in epithelial RPE cells, whereas blocking cadherin interactions disrupted microfibril deposition. Renal podocytes had a transitional phenotype with pericellular ß-catenin but little ZO-1; they required syndecan-4 and fibronectin for efficient microfibril deposition. Thus, epithelial-mesenchymal status modulates microfibril deposition.


Assuntos
Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Microfibrilas/metabolismo , Proteínas dos Microfilamentos/genética , Actinas/genética , Actinas/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular , Células Epiteliais/ultraestrutura , Feminino , Fibrilina-1 , Fibrilinas , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Fibronectinas/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica , Heparitina Sulfato/metabolismo , Humanos , Integrinas/genética , Integrinas/metabolismo , Junções Intercelulares/metabolismo , Junções Intercelulares/ultraestrutura , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/ultraestrutura , Microfibrilas/ultraestrutura , Proteínas dos Microfilamentos/metabolismo , Especificidade de Órgãos , Podócitos/metabolismo , Podócitos/ultraestrutura , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/ultraestrutura , Sindecana-4/genética , Sindecana-4/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
J Biol Chem ; 289(52): 36194-203, 2014 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-25359774

RESUMO

Mucopolysaccharidosis I Hurler (MPSI-H) is a pediatric lysosomal storage disease caused by genetic deficiencies in IDUA, coding for α-l-iduronidase. Idua(-/-) mice share similar clinical pathology with patients, including the accumulation of the undegraded glycosaminoglycans (GAGs) heparan sulfate (HS), and dermatan sulfate (DS), progressive neurodegeneration, and dysostosis multiplex. Hematopoietic stem cell transplantation (HSCT) is the most effective treatment for Hurler patients, but reduced intensity conditioning is a risk factor in transplantation, suggesting an underlying defect in hematopoietic cell engraftment. HS is a co-receptor in the CXCL12/CXCR4 axis of hematopoietic stem and progenitor cell (HSPC) migration to the bone marrow (BM), but the effect of HS alterations on HSPC migration, or the functional role of HS in MPSI-H are unknown. We demonstrate defective WT HSPC engraftment and migration in Idua(-/-) recipient BM, particularly under reduced intensity conditioning. Both intra- but especially extracellular Idua(-/-) BM HS was significantly increased and abnormally sulfated. Soluble heparinase-sensitive GAGs from Idua(-/-) BM and specifically 2-O-sulfated HS, elevated in Idua(-/-) BM, both inhibited CXCL12-mediated WT HSPC transwell migration, while DS had no effect. Thus we have shown that excess overly sulfated extracellular HS binds, and sequesters CXCL12, limiting hematopoietic migration and providing a potential mechanism for the limited scope of HSCT in Hurler disease.


Assuntos
Movimento Celular , Células-Tronco Hematopoéticas/fisiologia , Heparitina Sulfato/farmacologia , Mucopolissacaridose I/terapia , Animais , Medula Óssea/patologia , Quimiocina CXCL12/metabolismo , Sobrevivência de Enxerto , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nicho de Células-Tronco
7.
Ann Rheum Dis ; 74(6): 1249-56, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24442880

RESUMO

OBJECTIVES: Leri's pleonosteosis (LP) is an autosomal dominant rheumatic condition characterised by flexion contractures of the interphalangeal joints, limited motion of multiple joints, and short broad metacarpals, metatarsals and phalanges. Scleroderma-like skin thickening can be seen in some individuals with LP. We undertook a study to characterise the phenotype of LP and identify its genetic basis. METHODS AND RESULTS: Whole-genome single-nucleotide polymorphism genotyping in two families with LP defined microduplications of chromosome 8q22.1 as the cause of this condition. Expression analysis of dermal fibroblasts from affected individuals showed overexpression of two genes, GDF6 and SDC2, within the duplicated region, leading to dysregulation of genes that encode proteins of the extracellular matrix and downstream players in the transforming growth factor (TGF)-ß pathway. Western blot analysis revealed markedly decreased inhibitory SMAD6 levels in patients with LP. Furthermore, in a cohort of 330 systemic sclerosis cases, we show that the minor allele of a missense SDC2 variant, p.Ser71Thr, could confer protection against disease (p<1×10(-5)). CONCLUSIONS: Our work identifies the genetic cause of LP in these two families, demonstrates the phenotypic range of the condition, implicates dysregulation of extracellular matrix homoeostasis genes in its pathogenesis, and highlights the link between TGF-ß/SMAD signalling, growth/differentiation factor 6 and syndecan-2. We propose that LP is an additional member of the growing 'TGF-ß-pathies' group of musculoskeletal disorders, which includes Myhre syndrome, acromicric dysplasia, geleophysic dysplasias, Weill-Marchesani syndromes and stiff skin syndrome. Identification of a systemic sclerosis-protective SDC2 variant lays the foundation for exploration of the role of syndecan-2 in systemic sclerosis in the future.


Assuntos
Cromossomos Humanos Par 8/genética , Duplicação Gênica , Fator 6 de Diferenciação de Crescimento/genética , Deformidades Congênitas da Mão/genética , Artropatias/congênito , Ossificação Heterotópica/genética , Escleroderma Sistêmico/genética , Sindecana-2/genética , Adulto , Idoso , Pré-Escolar , Matriz Extracelular/metabolismo , Fácies , Feminino , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Fator 6 de Diferenciação de Crescimento/metabolismo , Deformidades Congênitas da Mão/metabolismo , Deformidades Congênitas da Mão/fisiopatologia , Humanos , Lactente , Artropatias/genética , Artropatias/metabolismo , Artropatias/fisiopatologia , Masculino , Pessoa de Meia-Idade , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/fisiopatologia , Fenótipo , Transdução de Sinais , Sindecana-2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adulto Jovem
8.
Stem Cells ; 32(11): 2869-79, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25074424

RESUMO

Mouse embryonic stem cells (mESCs) and epiblast stem cells represent the naïve and primed pluripotent states, respectively. These cells self-renew via distinct signaling pathways and can transition between the two states in the presence of appropriate growth factors. Manipulation of signaling pathways has therefore allowed the isolation of novel pluripotent cell types such as Fibroblast growth factor, Activin and BIO-derived stem cells and IESCs. However, the effect of cell seeding density on pluripotency remains unexplored. In this study, we have examined whether mESCs can epigenetically regulate E-cadherin to enter a primed-like state in response to low cell seeding density. We show that low density seeding in the absence of leukaemia inhibitory factor (LIF) induces decreased apoptosis and maintenance of pluripotency via Activin/Nodal, concomitant with loss of E-cadherin, Signal transducer and activator of transcription phosphorylation, and chimera-forming ability. These cells, E-cadherin negative proliferating stem cells (ENPSCs) can be reverted to a naïve phenotype by addition of LIF or forced E-cadherin expression. However, prolonged culture of ENPSCs without LIF leads to methylation of the E-cadherin promoter (ENPSC(M)), which cannot be reversed by LIF supplementation, and increased histone H3K27 and decreased H3K4 trimethylation. Transcript analysis of ENPSC(M) revealed a primed-like phenotype and their differentiation leads to enrichment of neuroectoderm cells. The generation of ENPSCs is similar to tumorigenesis as ENPSCs exhibit transcript alterations associated with neoplasia, hyperplasia, carcinoma, and metastasis. We therefore describe a novel cell model to elucidate the role of E-cadherin in pluripotency and to investigate epigenetic regulation of this gene during mESC differentiation and tumor metastasis.


Assuntos
Caderinas/metabolismo , Diferenciação Celular/fisiologia , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/citologia , Regiões Promotoras Genéticas , Transdução de Sinais/fisiologia , Animais , Separação Celular , Células Cultivadas , Epigênese Genética/efeitos dos fármacos , Humanos , Fator Inibidor de Leucemia/metabolismo , Camundongos da Linhagem 129 , Células-Tronco Pluripotentes/metabolismo
9.
J Biol Chem ; 288(8): 5530-8, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23235146

RESUMO

As our understanding of what guides the behavior of multi- and pluripotent stem cells deepens, so too does our ability to utilize certain cues to manipulate their behavior and maximize their therapeutic potential. Engineered, biologically functionalized materials have the capacity to influence stem cell behavior through a powerful combination of biological, mechanical, and topographical cues. Here, we present the development of a novel electrospun scaffold, functionalized with glycosaminoglycans (GAGs) ionically immobilized onto the fiber surface. Bound GAGs retained the ability to interact with GAG-binding molecules and, crucially, presented GAG sulfation motifs fundamental to mediating stem cell behavior. Bound GAG proved to be biologically active, rescuing the neural differentiation capacity of heparan sulfate-deficient mouse embryonic stem cells and functioning in concert with FGF4 to facilitate the formation of extensive neural processes across the scaffold surface. The combination of GAGs with electrospun scaffolds creates a biomaterial with potent applicability for the propagation and effective differentiation of pluripotent stem cells.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Heparitina Sulfato/metabolismo , Alilamina/química , Animais , Materiais Biocompatíveis/química , Diferenciação Celular , Células Cultivadas , Dissacarídeos/química , Epitopos/química , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Transgênicos , Oligossacarídeos/química , Biblioteca de Peptídeos , Polímeros/química , Regeneração , Medicina Regenerativa/métodos
10.
Biochem Soc Trans ; 42(3): 607-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24849226

RESUMO

The field of regenerative medicine offers tantalizing hope for the repair and replacement of damaged organs and tissues, with the ultimate goal of restoring normal tissue function. This field represents an enormous range of biological, chemical and biophysical technologies that harness the restorative properties of living materials, especially human cells, to produce new molecular and cellular medicines, diagnostics, devices and healthcare research tools. The goal of this Biochemical Society Annual Symposium was to explore the key biochemical determinants of tissue regeneration, and we highlight the contribution of biochemistry to this emerging field of regenerative medicine.


Assuntos
Bioquímica , Medicina Regenerativa , Humanos
11.
Biochem Soc Trans ; 42(3): 689-95, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24849238

RESUMO

Differentiation and subsequent specialization of every cell within an organism is an intricate interwoven process. A complex network of signalling pathways eventually leads to the specification of a multitude of different cell types able to function co-operatively. HS (heparan sulfate) is a highly sulfated linear polysaccharide that resides at the pericellular cell-matrix interface where it dictates the binding and activity of a large number of proteins, including growth factors and morphogens such as members of the FGF (fibroblast growth factor) and BMP (bone morphogenetic protein) families. Embryonic stem cells derived from mice with mutations in components of the HS biosynthetic pathway provide an opportunity to dissect the contribution of HS to signalling pathways critical for regulating stem cell maintenance and differentiation. In addition to improving our understanding of signalling mechanisms, this knowledge enables the selection of exogenous HS saccharides to improve the efficiency and selectivity of directed differentiation protocols, offering a cost-effective alternative to high concentrations of expensive growth factors to drive differentiation towards a particular therapeutically relevant cell type.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Glicosaminoglicanos/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Desenvolvimento Embrionário , Glicosaminoglicanos/farmacologia , Humanos , Camundongos
12.
Biochem Soc Trans ; 42(3): 703-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24849240

RESUMO

Most research strategies for cartilage tissue engineering use extended culture with complex media loaded with costly GFs (growth factors) to drive tissue assembly and yet they result in the production of cartilage with inferior mechanical and structural properties compared with the natural tissue. Recent evidence suggests that GAGs (glycosaminoglycans) incorporated into tissue engineering scaffolds can sequester and/or activate GFs and thereby more effectively mimic the natural ECM (extracellular matrix). Such approaches may have potential for the improvement of cartilage engineering. However, natural GAGs are structurally complex and heterogeneous, making structure-function relationships hard to determine and clinical translation difficult. Importantly, subfractions of GAGs with specific chain lengths and sulfation patterns have been shown to activate key signalling processes during stem cell differentiation. In addition, recently, GAGs have been bound to synthetic biomaterials, such as electrospun scaffolds and hydrogels, in biologically active conformations, and methods to purify and select affinity-matched GAGs for specific GFs have also been developed. The identification and use of specific GAG moieties to promote chondrogenesis is therefore an exciting new avenue of research. Combining these with synthetic biomaterials may allow a more effective mimicry of the natural ECM, reduction in the need for expensive GFs, and perhaps the deposition of an articular cartilage-like matrix in a clinically relevant manner.


Assuntos
Cartilagem/fisiologia , Linhagem da Célula , Glicosaminoglicanos/fisiologia , Regeneração , Cartilagem/citologia , Cartilagem/metabolismo , Glicosaminoglicanos/metabolismo , Humanos
13.
Mol Ther ; 21(10): 1938-49, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23748415

RESUMO

Mucopolysaccharidosis type IIIA (MPSIIIA) is a lysosomal storage disorder caused by mutations in N-sulfoglucosamine sulfohydrolase (SGSH), resulting in heparan sulfate (HS) accumulation and progressive neurodegeneration. There are no treatments. We previously demonstrated improved neuropathology in MPSIIIA mice using lentiviral vectors (LVs) overexpressing SGSH in wild-type (WT) hematopoietic stem cell (HSC) transplants (HSCTs), achieved via donor monocyte/microglial engraftment in the brain. However, neurological disease was not corrected using LVs in autologous MPSIIIA HSCTs. To improve brain expression via monocyte/microglial specificity, LVs expressing enhanced green fluorescent protein (eGFP) under ubiquitous phosphoglycerate kinase (PGK) or myeloid-specific promoters were compared in transplanted HSCs. LV-CD11b-GFP gave significantly higher monocyte/B-cell eGFP expression than LV-PGK-GFP or LV-CD18-GFP after 6 months. Subsequently, autologous MPSIIIA HSCs were transduced with either LV-PGK-coSGSH or LV-CD11b-coSGSH vectors expressing codon-optimized SGSH and transplanted into MPSIIIA mice. Eight months after HSCT, LV-PGK-coSGSH vectors produced bone marrow SGSH (576% normal activity) similar to LV-CD11b-coSGSH (473%), but LV-CD11b-coSGSH had significantly higher brain expression (11 versus 7%), demonstrating improved brain specificity. LV-CD11b-coSGSH normalized MPSIIIA behavior, brain HS, GM2 ganglioside, and neuroinflammation to WT levels, whereas LV-PGK-coSGSH partly corrected neuropathology but not behavior. We demonstrate compelling evidence of neurological disease correction using autologous myeloid driven lentiviral-HSC gene therapy in MPSIIIA mice.


Assuntos
Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Hidrolases/genética , Hidrolases/metabolismo , Microglia/fisiologia , Mucopolissacaridose III/terapia , Animais , Encéfalo/enzimologia , Antígeno CD11b/genética , Linhagem Celular , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Lentivirus/genética , Leucócitos/metabolismo , Lisossomos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/enzimologia , Mucopolissacaridose III/metabolismo , Mucopolissacaridose III/patologia , Células Mieloides/enzimologia , Células Mieloides/fisiologia , Especificidade de Órgãos , Regiões Promotoras Genéticas
14.
Cell Rep ; 43(1): 113668, 2024 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-38198277

RESUMO

Perlecan (HSPG2), a heparan sulfate proteoglycan similar to agrin, is key for extracellular matrix (ECM) maturation and stabilization. Although crucial for cardiac development, its role remains elusive. We show that perlecan expression increases as cardiomyocytes mature in vivo and during human pluripotent stem cell differentiation to cardiomyocytes (hPSC-CMs). Perlecan-haploinsuffient hPSCs (HSPG2+/-) differentiate efficiently, but late-stage CMs have structural, contractile, metabolic, and ECM gene dysregulation. In keeping with this, late-stage HSPG2+/- hPSC-CMs have immature features, including reduced ⍺-actinin expression and increased glycolytic metabolism and proliferation. Moreover, perlecan-haploinsuffient engineered heart tissues have reduced tissue thickness and force generation. Conversely, hPSC-CMs grown on a perlecan-peptide substrate are enlarged and display increased nucleation, typical of hypertrophic growth. Together, perlecan appears to play the opposite role of agrin, promoting cellular maturation rather than hyperplasia and proliferation. Perlecan signaling is likely mediated via its binding to the dystroglycan complex. Targeting perlecan-dependent signaling may help reverse the phenotypic switch common to heart failure.


Assuntos
Agrina , Proteoglicanas de Heparan Sulfato , Humanos , Proteoglicanas de Heparan Sulfato/genética , Proteoglicanas de Heparan Sulfato/metabolismo , Agrina/metabolismo , Miócitos Cardíacos/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo
15.
Stem Cells ; 30(9): 1842-51, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22696497

RESUMO

We have recently shown that loss of E-cadherin in mouse embryonic stem cells (mESCs) results in significant alterations to both the transcriptome and hierarchy of pluripotency-associated signaling pathways. Here, we show that E-cadherin promotes kruppel-like factor 4 (Klf4) and Nanog transcript and protein expression in mESCs via STAT3 phosphorylation and that ß-catenin, and its binding region in E-cadherin, is required for this function. To further investigate the role of E-cadherin in leukemia inhibitory factor (LIF)-dependent pluripotency, E-cadherin null (Ecad(-/-)) mESCs were cultured in LIF/bone morphogenetic protein supplemented medium. Under these conditions, Ecad(-/-) mESCs exhibited partial restoration of cell-cell contact and STAT3 phosphorylation and upregulated Klf4, Nanog, and N-cadherin transcripts and protein. Abrogation of N-cadherin using an inhibitory peptide caused loss of phospho STAT3, Klf4, and Nanog in these cells, demonstrating that N-cadherin supports LIF-dependent pluripotency in this context. We therefore identify a novel molecular mechanism linking E- and N-cadherin to the core circuitry of pluripotency in mESCs. This mechanism may explain the recently documented role of E-cadherin in efficient induced pluripotent stem cell reprogramming.


Assuntos
Caderinas/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/biossíntese , Fator de Transcrição STAT3/metabolismo , Animais , Diferenciação Celular , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Proteína Homeobox Nanog , Fosforilação , Transdução de Sinais , Transfecção
16.
In Vitro Model ; 2(3-4): 99-111, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37808200

RESUMO

Purpose: This 3D in vitro cancer model for propagation of patient-derived cells, using a synthetic self-assembling peptide gel, allows the formation of a fully characterised, tailorable tumour microenvironment. Unlike many existing 3D cancer models, the peptide gel is inert, apart from molecules and motifs deliberately added or produced by cells within the model. Methods: Breast cancer patient-derived xenografts (PDXs) were disaggregated and embedded in a peptide hydrogel. Growth was monitored by microscopic examination and at intervals, cells were extracted from the gels and passaged on into fresh gels. Passaged cells were assessed by qPCR and immunostaining techniques for the retention of characteristic markers. Results: Breast cancer PDXs were shown to be capable of expansion over four or more passages in the peptide gel. Contaminating mouse cells were found to be rapidly removed by successive passages. The resulting human cells were shown to be compatible with a range of common assays useful for assessing survival, growth and maintenance of heterogeneity. Conclusions: Based on these findings, the hydrogel has the potential to provide an effective and practical breast cancer model for the passage of PDXs which will have the added benefits of being relatively cheap, fully-defined and free from the use of animals or animal products. Encapsulated cells will require further validation to confirm the maintenance of cell heterogeneity, genotypes and phenotypes across passage, but with further development, including the addition of bespoke cell and matrix components of the tumour microenvironment, there is clear potential to model other cancer types. Supplementary Information: The online version contains supplementary material available at 10.1007/s44164-023-00048-x.

17.
Commun Biol ; 6(1): 463, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-37117487

RESUMO

Biomechanical cues from the extracellular matrix (ECM) are essential for directing many cellular processes, from normal development and repair, to disease progression. To better understand cell-matrix interactions, we have developed a new instrument named 'OptoRheo' that combines light sheet fluorescence microscopy with particle tracking microrheology. OptoRheo lets us image cells in 3D as they proliferate over several days while simultaneously sensing the mechanical properties of the surrounding extracellular and pericellular matrix at a sub-cellular length scale. OptoRheo can be used in two operational modalities (with and without an optical trap) to extend the dynamic range of microrheology measurements. We corroborated this by characterising the ECM surrounding live breast cancer cells in two distinct culture systems, cell clusters in 3D hydrogels and spheroids in suspension culture. This cutting-edge instrument will transform the exploration of drug transport through complex cell culture matrices and optimise the design of the next-generation of disease models.


Assuntos
Matriz Extracelular , Hidrogéis , Microscopia de Fluorescência , Comunicação Celular
18.
Acta Neuropathol Commun ; 11(1): 6, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36631900

RESUMO

The most common malignant brain tumour in children, medulloblastoma (MB), is subdivided into four clinically relevant molecular subgroups, although targeted therapy options informed by understanding of different cellular features are lacking. Here, by comparing the most aggressive subgroup (Group 3) with the intermediate (SHH) subgroup, we identify crucial differences in tumour heterogeneity, including unique metabolism-driven subpopulations in Group 3 and matrix-producing subpopulations in SHH. To analyse tumour heterogeneity, we profiled individual tumour nodules at the cellular level in 3D MB hydrogel models, which recapitulate subgroup specific phenotypes, by single cell RNA sequencing (scRNAseq) and 3D OrbiTrap Secondary Ion Mass Spectrometry (3D OrbiSIMS) imaging. In addition to identifying known metabolites characteristic of MB, we observed intra- and internodular heterogeneity and identified subgroup-specific tumour subpopulations. We showed that extracellular matrix factors and adhesion pathways defined unique SHH subpopulations, and made up a distinct shell-like structure of sulphur-containing species, comprising a combination of small leucine-rich proteoglycans (SLRPs) including the collagen organiser lumican. In contrast, the Group 3 tumour model was characterized by multiple subpopulations with greatly enhanced oxidative phosphorylation and tricarboxylic acid (TCA) cycle activity. Extensive TCA cycle metabolite measurements revealed very high levels of succinate and fumarate with malate levels almost undetectable particularly in Group 3 tumour models. In patients, high fumarate levels (NMR spectroscopy) alongside activated stress response pathways and high Nuclear Factor Erythroid 2-Related Factor 2 (NRF2; gene expression analyses) were associated with poorer survival. Based on these findings we predicted and confirmed that NRF2 inhibition increased sensitivity to vincristine in a long-term 3D drug treatment assay of Group 3 MB. Thus, by combining scRNAseq and 3D OrbiSIMS in a relevant model system we were able to define MB subgroup heterogeneity at the single cell level and elucidate new druggable biomarkers for aggressive Group 3 and low-risk SHH MB.


Assuntos
Biomarcadores Tumorais , Neoplasias Cerebelares , Proteínas Hedgehog , Meduloblastoma , Humanos , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Proteínas Hedgehog/metabolismo , Hidrogéis/uso terapêutico , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Fator 2 Relacionado a NF-E2 , Análise de Célula Única , RNA-Seq
19.
J Biol Chem ; 286(8): 6241-52, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21148566

RESUMO

Heparan sulfate proteoglycans (HSPG) encompass some of the most abundant macromolecules on the surface of almost every cell type. Heparan sulfate (HS) chains provide a key interaction surface for the binding of numerous proteins such as growth factors and morphogens, helping to define the ability of a cell to respond selectively to environmental cues. The specificity of HS-protein interactions are governed predominantly by the order and positioning of sulfate groups, with distinct cell types expressing unique sets of HS epitopes. Embryos deficient in HS-synthesis (Ext1(-/-)) exhibit pre-gastrulation lethality and lack recognizable organized mesoderm and extraembryonic tissues. Here we demonstrate that embryonic stem cells (ESCs) derived from Ext1(-/-) embryos are unable to differentiate into hematopoietic lineages, instead retaining ESC marker expression throughout embryoid body (EB) culture. However hematopoietic differentiation can be restored by the addition of soluble heparin. Consistent with specific size and composition requirements for HS:growth factor signaling, chains measuring at least 12 saccharides were required for partial rescue of hematopoiesis with longer chains (18 saccharides or more) required for complete rescue. Critically N- and 6-O-sulfate groups were essential for rescue. Heparin addition restored the activity of multiple signaling pathways including bone morphogenic protein (BMP) with activation of phospho-SMADs re-established by the addition of heparin. Heparin addition to wild-type cultures also altered the outcome of differentiation, promoting hematopoiesis at low concentrations, yet inhibiting blood formation at high concentrations. Thus altering the levels of HS and HS sulfation within differentiating ESC cultures provides an attractive and accessible mechanism for influencing cell fate.


Assuntos
Anticoagulantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Heparina/farmacologia , Heparitina Sulfato/farmacologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Células-Tronco Embrionárias/citologia , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Knockout , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo
20.
J Biol Chem ; 286(43): 37515-24, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21873421

RESUMO

Mucopolysaccharide (MPS) diseases are characterized by accumulation of glycosaminoglycans (GAGs) due to deficiencies in lysosomal enzymes responsible for GAG breakdown. Using a murine model of MPSI Hurler (MPSIH), we have quantified the heparan sulfate (HS) accumulation resulting from α-l-iduronidase (Idua) deficiency. HS levels were significantly increased in liver and brain tissue from 12-week-old Idua(-/-) mice by 87- and 20-fold, respectively. In addition, HS chains were shown to contain significantly increased N-, 2-O-, and 6-O-sulfation. Disaccharide compositional analyses also uncovered an HS disaccharide uniquely enriched in MPSIH, representing the terminal iduronic acid residue capping the non-reducing end of the HS chain, where no further degradation can occur in the absence of Idua. Critically, we identified that excess HS, some of which is colocalized to the Golgi secretory pathway, acts as a positive regulator of HS-sulfation, increasing the N-sulfotransferase activity of HS-modifying N-deacetylase/N-sulfotransferase enzymes. This mechanism may have severe implications during disease progression but, now identified, could help direct improved therapeutic strategies.


Assuntos
Complexo de Golgi/metabolismo , Heparitina Sulfato/metabolismo , Iduronidase , Mucopolissacaridose I/enzimologia , Sulfotransferases/metabolismo , Animais , Modelos Animais de Doenças , Complexo de Golgi/genética , Heparitina Sulfato/genética , Humanos , Ácido Idurônico/metabolismo , Camundongos , Camundongos Knockout , Mucopolissacaridose I/genética , Sulfotransferases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA