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










Database
Language
Publication year range
1.
Metab Eng ; 81: 157-166, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38081506

ABSTRACT

Rare diseases are, despite their name, collectively common and millions of people are affected daily of conditions where treatment often is unavailable. Sulfatases are a large family of activating enzymes related to several of these diseases. Heritable genetic variations in sulfatases may lead to impaired activity and a reduced macromolecular breakdown within the lysosome, with several severe and lethal conditions as a consequence. While therapeutic options are scarce, treatment for some sulfatase deficiencies by recombinant enzyme replacement are available. The recombinant production of such sulfatases suffers greatly from both low product activity and yield, further limiting accessibility for patient groups. To mitigate the low product activity, we have investigated cellular properties through computational evaluation of cultures with varying media conditions and comparison of two CHO clones with different levels of one active sulfatase variant. Transcriptome analysis identified 18 genes in secretory pathways correlating with increased sulfatase production. Experimental validation by upregulation of a set of three key genes improved the specific enzymatic activity at varying degree up to 150-fold in another sulfatase variant, broadcasting general production benefits. We also identified a correlation between product mRNA levels and sulfatase activity that generated an increase in sulfatase activity when expressed with a weaker promoter. Furthermore, we suggest that our proposed workflow for resolving bottlenecks in cellular machineries, to be useful for improvements of cell factories for other biologics as well.


Subject(s)
Sulfatases , Humans , Sulfatases/genetics , Sulfatases/metabolism
2.
Neuroscience ; 405: 92-102, 2019 05 01.
Article in English | MEDLINE | ID: mdl-29101080

ABSTRACT

Brain injury is associated with neuroinflammation, and microglia are key players in this process. Microglia can acquire pro-inflammatory or anti-inflammatory properties, but how this affects neural stem cells (NSCs) remains controversial. Here, NSCs were grown in conditioned media collected from either non-stimulated microglia, or microglia stimulated with pro- or anti-inflammatory agents. NSC survival, proliferation, migration, and differentiation were investigated thereafter. We found that NSCs kept in conditioned medium from the anti-inflammatory microglial subtype had better survival, increased migration, and lower astrocytic differentiation compared to NSCs grown in conditioned medium collected from the pro-inflammatory subtype. Finally, we found that NSCs differentiated in microglial conditioned media generated cells expressing the pro-inflammatory chemokine CCL2, most pronounced when differentiated in medium from the pro-inflammatory microglia subtype. Our results show that microglial subtypes regulate NSCs differently and induce generation of cells with inflammatory properties.


Subject(s)
Cytokines/metabolism , Microglia/physiology , Neural Stem Cells/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Astrocytes/physiology , Cell Differentiation/drug effects , Cell Movement/drug effects , Culture Media, Conditioned , Cytokines/biosynthesis , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
3.
PLoS One ; 12(10): e0185777, 2017.
Article in English | MEDLINE | ID: mdl-28985224

ABSTRACT

The accumulation of amyloid beta (Aß) peptide (Amyloid cascade hypothesis), an APP protein cleavage product, is a leading hypothesis in the etiology of Alzheimer's disease (AD). In order to identify additional AD risk genes, we performed targeted sequencing and rare variant burden association study for nine candidate genes involved in the amyloid metabolism in 1886 AD cases and 1700 controls. We identified a significant variant burden association for the gene encoding caspase-8, CASP8 (p = 8.6x10-5). For two CASP8 variants, p.K148R and p.I298V, the association remained significant in a combined sample of 10,820 cases and 8,881 controls. For both variants we performed bioinformatics structural, expression and enzymatic activity studies and obtained evidence for loss of function effects. In addition to their role in amyloid processing, caspase-8 and its downstream effector caspase-3 are involved in synaptic plasticity, learning, memory and control of microglia pro-inflammatory activation and associated neurotoxicity, indicating additional mechanisms that might contribute to AD. As caspase inhibition has been proposed as a mechanism for AD treatment, our finding that AD-associated CASP8 variants reduce caspase function calls for caution and is an impetus for further studies on the role of caspases in AD and other neurodegenerative diseases.


Subject(s)
Alleles , Alzheimer Disease/genetics , Caspase 8/genetics , Genetic Variation , Alzheimer Disease/metabolism , Case-Control Studies , Caspase 8/metabolism , Cell Line, Tumor , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Neurons/metabolism
4.
Nat Immunol ; 17(11): 1282-1290, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27618552

ABSTRACT

Glioma cells recruit and exploit microglia (the resident immune cells of the brain) for their proliferation and invasion ability. The underlying molecular mechanism used by glioma cells to transform microglia into a tumor-supporting phenotype has remained elusive. We found that glioma-induced microglia conversion was coupled to a reduction in the basal activity of microglial caspase-3 and increased S-nitrosylation of mitochondria-associated caspase-3 through inhibition of thioredoxin-2 activity, and that inhibition of caspase-3 regulated microglial tumor-supporting function. Furthermore, we identified the activity of nitric oxide synthase 2 (NOS2, also known as iNOS) originating from the glioma cells as a driving stimulus in the control of microglial caspase-3 activity. Repression of glioma NOS2 expression in vivo led to a reduction in both microglia recruitment and tumor expansion, whereas depletion of microglial caspase-3 gene promoted tumor growth. Our results provide evidence that inhibition of the denitrosylation of S-nitrosylated procaspase-3 mediated by the redox protein Trx2 is a part of the microglial pro-tumoral activation pathway initiated by glioma cancer cells.


Subject(s)
Caspase 3/metabolism , Glioma/metabolism , Glioma/pathology , Microglia/metabolism , Phenotype , Animals , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Enzyme Activation , Gene Knockdown Techniques , Glioma/immunology , Heterografts , Humans , Male , Mice , Microglia/immunology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Thioredoxins/metabolism , Tumor Burden
5.
Acta Neuropathol Commun ; 4(1): 92, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27566702

ABSTRACT

Ischemic stroke (caused by thrombosis, embolism or vasoconstriction) lead to the recruitment and activation of immune cells including resident microglia and infiltrating peripheral macrophages, which contribute to an inflammatory response involved in regulation of the neuronal damage. We showed earlier that upon pro-inflammatory stimuli, the orderly activation of caspase-8 and caspase-3/7 regulates microglia activation through a protein kinase C-δ dependent pathway. Here, we present in vivo evidence for the activation of caspase-8 and caspase-3 in microglia/macrophages in post-mortem tissue from human ischemic stroke subjects. Indeed, CD68-positive microglia/macrophages in the ischemic peri-infarct area exhibited significant expression of the cleaved and active form of caspase-8 and caspase-3. The temporal and spatial activation of caspase-8 was further investigated in a permanent middle cerebral artery occlusion mouse model of ischemic stroke. Increasing levels of active caspase-8 was found in Iba1-positive cells over time in the peri-infarct area, at 6, 24 and 48 h after artery occlusion. Analysis of post-mortem brain tissue from human subject who suffered two stroke events, referred as recent and old stroke, revealed that expression of cleaved caspase-8 and -3 in CD68-positive cells could only be found in the recent stroke area. Analysis of cleaved caspase-8 and -3 expressions in a panel of human stroke cases arranged upon days-after stroke and age-matched controls suggested that the expression of these caspases correlated with the time of onset of stroke. Collectively, these data illustrate the temporal and spatial activation of caspase-8 and -3 in microglia/macrophages occurring upon ischemic stroke and suggest that the expression of these caspases could be used in neuropathological diagnostic work.


Subject(s)
Brain Ischemia/enzymology , Brain/enzymology , Caspase 8/metabolism , Myeloid Cells/enzymology , Stroke/enzymology , Acute Disease , Aged , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/pathology , Brain Ischemia/pathology , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Humans , Male , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/enzymology , Microglia/pathology , Myeloid Cells/pathology , Stroke/pathology , Time Factors
6.
Oncotarget ; 7(25): 37456-37470, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27250033

ABSTRACT

In septic patients, the onset of septic shock occurs due to the over-activation of monocytes. We tested the therapeutic potential of directly targeting innate immune cell activation to limit the cytokine storm and downstream phases. We initially investigated whether caspase-8 could be an appropriate target given it has recently been shown to be involved in microglial activation. We found that LPS caused a mild increase in caspase-8 activity and that the caspase-8 inhibitor IETD-fmk partially decreased monocyte activation. Furthermore, caspase-8 inhibition induced necroptotic cell death of activated monocytes. Despite inducing necroptosis, caspase-8 inhibition reduced LPS-induced expression and release of IL-1ß and IL-10. Thus, blocking monocyte activation has positive effects on both the pro and anti-inflammatory phases of septic shock. We also found that in primary mouse monocytes, caspase-8 inhibition did not reduce LPS-induced activation or induce necroptosis. On the other hand, broad caspase inhibitors, which have already been shown to improve survival in mouse models of sepsis, achieved both. Thus, given that monocyte activation can be regulated in humans via the inhibition of a single caspase, we propose that the therapeutic use of caspase-8 inhibitors could represent a more selective alternative that blocks both phases of septic shock at the source.


Subject(s)
Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Monocytes/enzymology , Monocytes/immunology , Shock, Septic/prevention & control , Animals , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Shock, Septic/enzymology , Shock, Septic/immunology
7.
Aging (Albany NY) ; 7(9): 673-89, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26405176

ABSTRACT

Increasing evidence involves sustained pro-inflammatory microglia activation in the pathogenesis of different neurodegenerative diseases, particularly Parkinson's disease (PD). We recently uncovered a completely novel and unexpected role for caspase-8 and its downstream substrates caspase-3/7 in the control of microglia activation and associated neurotoxicity to dopaminergic cells. To demonstrate the genetic evidence, mice bearing a floxed allele ofCASP8 were crossed onto a transgenic line expressing Cre under the control of Lysozyme 2 gene. Analysis of caspase-8 gene deletion in brain microglia demonstrated a high efficiency in activated but not in resident microglia. Mice were challenged with lipopolysaccharide, a potent inducer of microglia activation, or with MPTP, which promotes specific dopaminergic cell damage and consequent reactive microgliosis. In neither of these models, CASP8 deletion appeared to affect the overall number of microglia expressing the pan specific microglia marker, Iba1. In contrast, CD16/CD32 expression, a microglial pro-inflammatory marker, was found to be negatively affected upon CASP8 deletion. Expression of additional proinflammatory markers were also found to be reduced in response to lipopolysaccharide. Of importance, reduced pro-inflammatory microglia activation was accompanied by a significant protection of the nigro-striatal dopaminergic system in the MPTP mouse model of PD.


Subject(s)
Caspase 8/genetics , Inflammation/pathology , MPTP Poisoning/genetics , Microglia/pathology , Myeloid Cells/enzymology , Animals , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Dopaminergic Neurons , Gliosis/pathology , Lipopolysaccharides/pharmacology , MPTP Poisoning/pathology , Mice , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Muramidase/genetics , Muramidase/metabolism
8.
Methods Mol Biol ; 1041: 11-6, 2013.
Article in English | MEDLINE | ID: mdl-23813364

ABSTRACT

Despite the fact that microglia cells were first described almost a century ago, microglia-derived immortalized cell lines have only been established in the last two decades. One should be aware of their limitations but also of their advantages. Cell lines offer a potentially powerful tool to investigate some functional aspects of microglia. Cell culturing of human and murine microglia cell lines will be described in this chapter. It includes a presentation of equipment needed, cell culture medium and supplements, cell culture monitoring, and a protocol describing the steps for subculturing of microglia cell lines.


Subject(s)
Cell Culture Techniques/methods , Microglia/cytology , Animals , Cell Line , Cells, Cultured , Humans , Mice
9.
Oncotarget ; 4(2): 289-97, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23470527

ABSTRACT

The TP73 gene, a member of the p53 family, due to the use of different promoters and alternative splicing, is transcribed into different isoforms with contrasting attributes and which contribute to its functional diversity. Considerable efforts are made to identify the functional diversity of the p73 splicing variants during tumorigenesis. TAp73α and TAp73ß isoforms have been shown to differentially regulate cell cycle progression, differentiation and apoptosis. Interestingly, a particular increase in expression of the TAp73 isoform, in favor of the α splicing variant, has been reported in multiple tumour types. Here, we report a distinctive role for TAp73ß isoform in the control of cell migration and invasion. In fact, TAp73ß- dependent induction of p57(Kip2) expression accounted for inhibitory effects on the actin cytoskeleton dynamics and thereby cancer cell motility. In contrast, TAp73α is not able to induce p57(Kip2) expression, and exhibits a positive effect on actin cytoskeleton dynamics as well as cell migration and invasion. In conclusion, the inhibitory effect on cell migration and invasion of TAp73ß would qualify this distinct p73 isoform as tumor suppressor gene. In contrast, the promoting effect of TAp73α on cell motility and invasion strengthens the potential oncogenic activities of this p73 isoform.


Subject(s)
Actin Cytoskeleton/physiology , Cell Movement/physiology , Cyclin-Dependent Kinase Inhibitor p57/physiology , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Tumor Suppressor Proteins/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Isoforms , Transcriptional Activation , Transfection , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...