Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
BMC Infect Dis ; 24(1): 613, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902610

ABSTRACT

INTRODUCTION: Since the mpox outbreak in 2022, it was unclear if and how often infections with mpox virus (MPXV) were clinically inapparent, i.e. not presenting to clinical care with mpox symptoms. Moreover, it was hypothesized that MPXV circulated in the affected communities before the outbreak was officially detected. METHODS: We retrospectively tested rectal and urethral swabs, and pooled samples for presence of MPXV. Samples were obtained from routine STI testing of three anonymous Community Based Voluntary Counselling and Testing (CBVCT) centres in Berlin, in 2022 and 2023. Testing results were linked to anonymously provided behavioural data. RESULTS: Overall, 9,053 samples from 6,600 client visits were included. Clinically inapparent MPXV infections were detectable in 1.1% of the samples. We did not find MPXV infections in the month before the first cases appeared in Berlin or between October 2022 and January 2023 when case numbers were low in Germany. However, during the outbreak period in 2022, we found clinically inapparent MPXV infections among 2.2% of the clients and during summer/autumn 2023 among 0.3%. The number of condomless anal/vaginal intercourse partners within the previous 6 months and PrEP use were identified as predictors of clinically inapparent MPXV infection. CONCLUSION: Clinically inapparent MPXV infections occurred during the mpox outbreak in Berlin in 2022 and post-outbreak in summer/autumn 2023. Unrecognized MPXV circulation in Berlin before the recognition of the outbreak in May 2022 appears unlikely. However, low-level sustained circulation of clinically inapparent MPXV infections need to be acknowledged in mpox prevention strategies.


Subject(s)
Counseling , Humans , Male , Adult , Berlin/epidemiology , Retrospective Studies , Female , Disease Outbreaks , Middle Aged , Young Adult , Germany/epidemiology
2.
Exp Clin Endocrinol Diabetes ; 130(2): 77-84, 2022 Feb.
Article in English | MEDLINE | ID: mdl-32615613

ABSTRACT

AIM/HYPOTHESIS: It was the aim to prospectively study regimes of "preventive" carbohydrate administration to avoid major reduction in plasma glucose during physical activity. METHODS: 24 patients with type 1 diabetes (age 41±12 years; 11 women, 13 men; BMI 26.5±4.7 kg/m2; HbA1c 9.1±1.5%; insulin dose 0.64±0.22 IU/kg body weight and day) participated in one experiment without physical activity and in three experiments with a 4 km, 60 min hike starting at 2 p.m.. No "preventive" carbohydrates, 2×10 g or 2×20 g carbohydrates (muesli bars) were taken when starting and after 30 min (randomized order). Plasma glucose was determined. RESULTS: Within 30 min after starting physical activity, plasma glucose fell by approximately 70 mg/dl, making additional carbohydrate intake necessary in 70% of the subjects. This drop was not prevented by any regimens of "preventive" carbohydrate intake. After the nadir, plasma glucose rose faster after the 2×20 g carbohydrate regime (the largest amount tested; p=0.0036). With "preventive" administration of carbohydrates, significantly (p<0.05) less additional "therapeutic" carbohydrates needed to be administered in 6 h following the initiation of the hike. CONCLUSIONS/INTERPRETATION: In conclusion, in the setting of 2 h postprandial exercise in type 1 diabetes, preventive carbohydrate supplementation alone will not completely eliminate the risk of brisk falls in plasma glucose concentrations or hypoglycaemic episodes. Else, higher amounts or repeated administration of carbohydrates may be necessary.


Subject(s)
Diabetes Mellitus, Type 1/diet therapy , Dietary Carbohydrates/pharmacology , Exercise , Hypoglycemia/prevention & control , Adult , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Dietary Carbohydrates/administration & dosage , Exercise/physiology , Female , Humans , Hypoglycemia/blood , Male , Middle Aged , Prospective Studies
3.
Prog Mol Biol Transl Sci ; 156: 1-62, 2018.
Article in English | MEDLINE | ID: mdl-29747811

ABSTRACT

Gangliosides (GGs) are cell type-specific sialic acid-containing glycosphingolipids (GSLs), which are enriched in mammalian brain. Defects in GSL metabolism cause fatal human diseases. GSLs are composed of a hydrophilic oligosaccharide linked in 1-O-position to a hydrophobic ceramide anchor, which itself is composed of a long-chain amino alcohol, the sphingoid base, and an amide-bound acyl chain. Biosynthesis of mammalian GGs and other GSLs starts with the formation of their hydrophobic ceramide anchor in the endoplasmic reticulum, followed by sequential glycosylation reactions along the secretory pathway, mainly at the luminal surface of Golgi and trans-Golgi network (TGN) membranes. Few membrane-anchored and often promiscuous glycosyltransferases allow the formation of cell type-specific glycolipid patterns in a combinatorial process. Inherited defects of these transferases therefore affect not only single structures but defined glycolipid series. GGs and other GSLs are thereafter transported by an exocytotic membrane flow to the plasma membrane where they are expressed in cell type-specific patterns, which can be modified by metabolic reactions at or near the cellular surface. Endocytosed (glyco)sphingolipids are degraded, together with other membrane lipids in a stepwise fashion by endolysosomal enzymes with the help of small lipid-binding proteins, the sphingolipid activator proteins (SAPs), at the surface of intraluminal lysosomal vesicles. Inherited defects in a sphingolipid-degrading enzyme or SAP cause the accumulation of the corresponding lipid substrates. Endolysosomal GSL degradation is strongly modified by the lipid components of the organelle microenvironments.


Subject(s)
Gangliosides/metabolism , Metabolic Diseases/physiopathology , Animals , Health Status , Humans
4.
Mol Biol Cell ; 29(4): 396-407, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29282281

ABSTRACT

Inherited peroxisomal biogenesis disorders (PBDs) are characterized by the absence of functional peroxisomes. They are caused by mutations of peroxisomal biogenesis factors encoded by Pex genes, and result in childhood lethality. Owing to the many metabolic functions fulfilled by peroxisomes, PBD pathology is complex and incompletely understood. Besides accumulation of peroxisomal educts (like very-long-chain fatty acids [VLCFAs] or branched-chain fatty acids) and lack of products (like bile acids or plasmalogens), many peroxisomal defects lead to detrimental mitochondrial abnormalities for unknown reasons. We generated Pex19 Drosophila mutants, which recapitulate the hallmarks of PBDs, like absence of peroxisomes, reduced viability, neurodegeneration, mitochondrial abnormalities, and accumulation of VLCFAs. We present a model of hepatocyte nuclear factor 4 (Hnf4)-induced lipotoxicity and accumulation of free fatty acids as the cause for mitochondrial damage in consequence of peroxisome loss in Pex19 mutants. Hyperactive Hnf4 signaling leads to up-regulation of lipase 3 and enzymes for mitochondrial ß-oxidation. This results in enhanced lipolysis, elevated concentrations of free fatty acids, maximal ß-oxidation, and mitochondrial abnormalities. Increased acid lipase expression and accumulation of free fatty acids are also present in a Pex19-deficient patient skin fibroblast line, suggesting the conservation of key aspects of our findings.


Subject(s)
Lipolysis/genetics , Membrane Proteins/genetics , Mitochondria/metabolism , Peroxisomal Disorders/genetics , Peroxisomes/metabolism , Animals , Disease Models, Animal , Drosophila , Fatty Acids/metabolism , Humans , Mutation , Peroxisomal Disorders/diagnosis
5.
Dis Model Mech ; 10(6): 737-750, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28389479

ABSTRACT

Sphingolipidoses are inherited diseases belonging to the class of lysosomal storage diseases (LSDs), which are characterized by the accumulation of indigestible material in the lysosome caused by specific defects in the lysosomal degradation machinery. While some LSDs can be efficiently treated by enzyme replacement therapy (ERT), this is not possible if the nervous system is affected due to the presence of the blood-brain barrier. Sphingolipidoses in particular often present as severe, untreatable forms of LSDs with massive sphingolipid and membrane accumulation in lysosomes, neurodegeneration and very short life expectancy. The digestion of intralumenal membranes within lysosomes is facilitated by lysosomal sphingolipid activator proteins (saposins), which are cleaved from a prosaposin precursor. Prosaposin mutations cause some of the severest forms of sphingolipidoses, and are associated with perinatal lethality in mice, hampering studies on disease progression. We identify the Drosophila prosaposin orthologue Saposin-related (Sap-r) as a key regulator of lysosomal lipid homeostasis in the fly. Its mutation leads to a typical spingolipidosis phenotype with an enlarged endolysosomal compartment and sphingolipid accumulation as shown by mass spectrometry and thin layer chromatography. Sap-r mutants show reduced viability with ∼50% survival to adulthood, allowing us to study progressive neurodegeneration and analyze their lipid profile in young and aged flies. Additionally, we observe a defect in sterol homeostasis with local sterol depletion at the plasma membrane. Furthermore, we find that autophagy is increased, resulting in the accumulation of mitochondria in lysosomes, concomitant with increased oxidative stress. Together, we establish Drosophila Sap-r mutants as a lysosomal storage disease model suitable for studying the age-dependent progression of lysosomal dysfunction associated with lipid accumulation and the resulting pathological signaling events.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Lysosomal Storage Diseases/genetics , Mutation/genetics , Saposins/genetics , Sphingolipidoses/genetics , Acids/metabolism , Animals , Cell Membrane/metabolism , Disease Models, Animal , Drosophila Proteins/metabolism , Lysosomal Storage Diseases/physiopathology , Lysosomes/metabolism , Lysosomes/ultrastructure , Motor Activity , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Oxidative Stress , Phospholipids/metabolism , Protein Transport , Sequence Homology, Amino Acid , Sphingolipidoses/physiopathology , Sphingolipids/metabolism , Sterols/metabolism , Subcellular Fractions/metabolism
6.
Biochim Biophys Acta ; 1841(5): 799-810, 2014 May.
Article in English | MEDLINE | ID: mdl-24184515

ABSTRACT

Endocytosed (glyco)sphingolipids are degraded, together with other membrane lipids in a stepwise fashion by endolysosomal enzymes with the help of small lipid binding proteins, the sphingolipid activator proteins (SAPs), at the surface of intraluminal lysosomal vesicles. Inherited defects in a sphingolipid-degrading enzyme or SAP cause the accumulation of the corresponding lipid substrates, including cytotoxic lysosphingolipids, such as galactosylsphingosine and glucosylsphingosine, and lead to a sphingolipidosis. Analysis of patients with prosaposin deficiency revealed the accumulation of intra-endolysosmal vesicles and membrane structures (IM). Feeding of prosaposin reverses the storage, suggesting inner membrane structures as platforms of sphingolipid degradation. Water soluble enzymes can hardly attack sphingolipids embedded in the membrane of inner endolysosomal vesicles. The degradation of sphingolipids with few sugar residues therefore requires the help of the SAPs, and is strongly stimulated by anionic membrane lipids. IMs are rich in anionic bis(monoacylglycero)phosphate (BMP). This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.


Subject(s)
Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Lysosomes/metabolism , Lysosomes/pathology , Sphingolipids/metabolism , Animals , Humans , Sphingolipid Activator Proteins/metabolism
7.
Lancet Neurol ; 10(6): 509-19, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21565557

ABSTRACT

BACKGROUND: Human retinal pigment epithelial (RPE) cells produce levodopa and their transplantation into the striatum might improve continuity of administration compared with that achieved with oral levodopa. We aimed to assess the safety, tolerability, and efficacy of transplantation of microcarrier-bound human RPE cells versus a sham surgery control in patients with advanced Parkinson's disease. METHODS: In this randomised, double-blind study eligible patients were aged 36-70 years, had been symptomatic for at least 5 years, were in Hoehn and Yahr stage 3-4 and had unified Parkinson's disease rating scale (UPDRS) motor scores of 38-70 when off medication (off state), and had symptoms that responded to oral levodopa but were insufficiently controlled by optimised pharmacotherapy. Randomisation was done in a 1:1 ratio. Only the neurosurgical team was aware of treatment assignments. During stereotactic transplantation around 325,000 cells per side were injected into the postcommissural putamen; sham surgery patients received partial burr holes. The primary efficacy endpoint was change in UPDRS off-state motor score at 12 months. This study is registered with ClinicalTrials.gov, number NCT00206687. FINDINGS: Of 71 enrolled patients, 35 underwent cell transplantation and 36 sham surgery. Change in mean motor scores did not differ significantly between groups (-10.5 [SD 10.26] for transplantation vs -10.1 [SD 12.26] for sham surgery, p=0.9). The overall rate of adverse events was similar in the two study groups, although the number attributable to surgery or RPE cells (mostly neurological or psychiatric) was higher in transplant recipients. Two and seven patients died in the sham surgery and transplantation group, respectively; one death in the latter group was possibly related to surgery or RPE cells. INTERPRETATION: Transplantation of human RPE cells provided no antiparkinsonian benefits compared with sham surgery. FUNDING: Bayer HealthCare AG.


Subject(s)
Corpus Striatum/surgery , Drug Carriers , Epithelial Cells/transplantation , Parkinson Disease/surgery , Placebos/therapeutic use , Retinal Pigment Epithelium/cytology , Adult , Aged , Double-Blind Method , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Levodopa/metabolism , Levodopa/therapeutic use , Male , Middle Aged , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Treatment Outcome
8.
Article in English | MEDLINE | ID: mdl-21502308

ABSTRACT

Lysosomal lipid storage diseases, or lipidoses, are inherited metabolic disorders in which typically lipids accumulate in cells and tissues. Complex lipids, such as glycosphingolipids, are constitutively degraded within the endolysosomal system by soluble hydrolytic enzymes with the help of lipid binding proteins in a sequential manner. Because of a functionally impaired hydrolase or auxiliary protein, their lipid substrates cannot be degraded, accumulate in the lysosome, and slowly spread to other intracellular membranes. In Niemann-Pick type C disease, cholesterol transport is impaired and unesterified cholesterol accumulates in the late endosome. In most lysosomal lipid storage diseases, the accumulation of one or few lipids leads to the coprecipitation of other hydrophobic substances in the endolysosomal system, such as lipids and proteins, causing a "traffic jam." This can impair lysosomal function, such as delivery of nutrients through the endolysosomal system, leading to a state of cellular starvation. Therapeutic approaches are currently restricted to mild forms of diseases with significant residual catabolic activities and without brain involvement.


Subject(s)
Lipid Metabolism , Lipidoses/metabolism , Lysosomal Storage Diseases/metabolism , Lysosomes/physiology , Biological Transport , Carrier Proteins/metabolism , Carrier Proteins/physiology , Endocytosis , Humans , Intracellular Membranes/metabolism , Lipidoses/pathology , Lysosomal Storage Diseases/pathology , Lysosomes/metabolism , Models, Biological , Sphingolipids/chemistry , Sphingolipids/metabolism , Sphingolipids/physiology
9.
Biochim Biophys Acta ; 1793(4): 674-83, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19014978

ABSTRACT

Cellular membranes enter the lysosomal compartment by endocytosis, phagocytosis, or autophagy. Within the lysosomal compartment, membrane components of complex structure are degraded into their building blocks. These are able to leave the lysosome and can then be utilized for the resynthesis of complex molecules or can be further degraded. Constitutive degradation of membranes occurs on the surface of intra-endosomal and intra-lysosomal membrane structures. Many integral membrane proteins are sorted to the inner membranes of endosomes and lysosome after ubiquitinylation. In the lysosome, proteins are degraded by proteolytic enzymes, the cathepsins. Phospholipids originating from lipoproteins or cellular membranes are degraded by phospholipases. Water-soluble glycosidases sequentially cleave off the terminal carbohydrate residues of glycoproteins, glycosaminoglycans, and glycosphingolipids. For glycosphingolipids with short oligosaccharide chains, the additional presence of membrane-active lysosomal lipid-binding proteins is required. The presence of lipid-binding proteins overcomes the phase problem of water soluble enzymes and lipid substrates by transferring the substrate to the degrading enzyme or by solubilizing the internal membranes. The lipid composition of intra-lysosomal vesicles differs from that of the plasma membrane. To allow at least glycosphingolipid degradation by hydrolases and activator proteins, the cholesterol content of these intraorganellar membranes decreases during endocytosis and the concentration of bis(monoacylglycero)phosphate, a stimulator of sphingolipid degradation, increases. A considerable part of our current knowledge about mechanism and biochemistry of lysosomal lipid degradation is derived from a class of human diseases, the sphingolipidoses, which are caused by inherited defects within sphingolipid and glycosphingolipid catabolism.


Subject(s)
Biochemistry , Intracellular Membranes/metabolism , Lipid Metabolism , Lysosomes/metabolism , Animals , Glycosphingolipids/chemistry , Glycosphingolipids/metabolism , Humans
10.
Biol Chem ; 388(12): 1333-43, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18020949

ABSTRACT

Human acid ceramidase catalyzes the last step of lysosomal sphingolipid degradation, the hydrolysis of ceramide to sphingosine and free fatty acid. Inherited deficiency of acid ceramidase activity leads to Farber disease (Farber lipogranulomatosis). In this study, we describe the overexpression and processing of recombinant human acid ceramidase in Sf21 insect cells, its purification and characterization. Infection of Sf21 cells with a recombinant baculovirus encoding acid ceramidase precursor led to a mixture of human acid ceramidase precursor and mature enzyme secreted into the medium. Acidification of the cell culture supernatant to pH 4.2-4.3 triggered the processing of the precursor and resulted in a homogeneous sample of mature human acid ceramidase. The enzyme was purified by chromatography on Concanavalin A Sepharose and Octyl Sepharose yielding 1 mg purified protein per liter of supernatant. The recombinant enzyme was deglycosylated with peptide N-glycosidase F and the main component of the released oligosaccharides was identified as GlcNAc(2)(Fuc)Man(3) by electrospray mass spectrometry. Apparently, five of the six potential N-glycosylation sites were used. Tryptic digestion of the functional recombinant enzyme and matrix-assisted laser desorption/ionization time-of-flight- and electrospray ionization-mass spectrometry analysis of the resulting peptides indicated disulfide bridges between C10-C319, C122-C271 and C367-C371.


Subject(s)
Galactosylgalactosylglucosylceramidase/biosynthesis , Galactosylgalactosylglucosylceramidase/chemistry , Animals , Baculoviridae/chemistry , Baculoviridae/genetics , Blotting, Western , Cell Line , Culture Media , Cysteine/chemistry , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Galactosylgalactosylglucosylceramidase/pharmacokinetics , Glycosylation , Humans , Hydrogen-Ion Concentration , Insecta , Kinetics , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/metabolism , Mass Spectrometry , Oligosaccharides/chemistry , Oxidation-Reduction , Peptides/chemistry , Plasmids/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin/chemistry
11.
Biol Chem ; 384(9): 1293-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14515991

ABSTRACT

Acid sphingomyelinase (A-SMase, EC 3.1.4.12) catalyzes the lysosomal degradation of sphingomyelin to phosphorylcholine and ceramide. Inherited deficiencies of acid sphingomyelinase activity result in various clinical forms of Niemann-Pick disease, which are characterised by massive lysosomal accumulation of sphingomyelin. Sphingomyelin hydrolysis by both, acid sphingomyelinase and membrane-associated neutral sphingomyelinase, plays also an important role in cellular signaling systems regulating proliferation, apoptosis and differentiation. Here, we present a potent and selective novel inhibitor of A-SMase, L-alpha-phosphatidyl-D-myo-inositol-3,5-bisphosphate (PtdIns3,5P2), a naturally occurring substance detected in mammalian, plant and yeast cells. The inhibition constant Ki for the new A-SMase inhibitor PtdIns3,5P2 is 0.53 microM as determined in a micellar assay system with radiolabeled sphingomyelin as substrate and recombinant human A-SMase purified from insect cells. Even at concentrations of up to 50 microM, PtdIns3,5P2 neither decreased plasma membrane-associated, magnesium-dependent neutral sphingomyelinase activity, nor was it an inhibitor of the lysosomal hydrolases beta-hexosaminidase A and acid ceramidase. Other phosphoinositides tested had no or a much weaker effect on acid sphingomyelinase. Different inositol-bisphosphates were studied to elucidate structure-activity relationships for A-SMase inhibition. Our investigations provide an insight into the structural features required for selective, efficient inhibition of acid sphingomyelinase and may also be used as starting point for the development of new potent A-SMase inhibitors optimised for diverse applications.


Subject(s)
Phosphatidylinositol Phosphates/pharmacology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Kinetics , Magnesium , Membrane Proteins/drug effects , Phosphatidylinositol Phosphates/chemistry , Recombinant Proteins , Structure-Activity Relationship , Substrate Specificity
12.
J Biol Chem ; 278(35): 32978-86, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12815059

ABSTRACT

Human acid ceramidase was overexpressed in Chinese hamster ovary cells by amplification of the transfected, full-length cDNA. The majority of the overexpressed enzyme was secreted into the culture media and purified to apparent homogeneity. The purified protein contained the same 13-(alpha) and 40 (beta)-kDa subunits as human acid ceramidase from natural sources, had an acidic pH optimum (4.5), and followed normal Michaelis-Menten kinetics using 14C- and BODIPY-labeled C12-ceramide as substrates. Deglycosylation studies showed that the recombinant enzyme contained mostly "high mannose" type oligosaccharides and that two distinct beta-subunits were present. Amino acid sequencing of these subunit polypeptides revealed a single N terminus, suggesting that the approximately 2-4-kDa molecular mass difference was likely due to C-terminal processing. The purified enzyme also catalyzed ceramide synthesis in vitro using 14C-labeled C12 fatty acid and sphingosine as substrates. Surprisingly, we found that media from the overexpressing hamster cells had increased acid sphingomyelinase activity and that this activity could be co-precipitated with acid ceramidase using anti-ceramidase antibodies. Overexpression of acid ceramidase in normal human skin fibroblasts also led to enhanced acid sphingomyelinase secretion, but this was not observed in Niemann-Pick disease cells. RNA studies showed that this increased activity was not due to overexpression of the endogenous acid sphingomyelinase gene. Uptake studies using mouse macrophages revealed rapid internalization of the acid ceramidase activity from the hamster cell media but not acid sphingomyelinase. These studies provide new insights into acid ceramidase and the related lipid hydrolase, acid sphingomyelinase.


Subject(s)
Galactosylgalactosylglucosylceramidase/chemistry , Galactosylgalactosylglucosylceramidase/isolation & purification , Sphingomyelin Phosphodiesterase/chemistry , Adenoviridae/genetics , Amidohydrolases/pharmacology , Animals , Blotting, Northern , CHO Cells , Catalysis , Chromatography, Gel , Concanavalin A/chemistry , Cricetinae , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Glycosylation , Hexosaminidases/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics , Lipids , Macrophages/metabolism , Mice , Mice, Knockout , Neuraminidase/pharmacology , Oligosaccharides/chemistry , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase , Precipitin Tests , Protein Structure, Tertiary , RNA/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sepharose/chemistry , Skin/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...