Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 67(12): 9857-9868, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38842931

ABSTRACT

Increasing research efforts focus on exploiting antibodies to inhibit the amyloid formation of neurodegenerative proteins. Nevertheless, it is challenging to discover antibodies that inhibit this process in a specific manner. Using ribosome display, we screened for synthetic single-domain antibodies, i.e., sybodies, of the P1 region of α-synuclein (residues 36-42), a protein that forms amyloid in Parkinson's disease and multiple-system atrophy. Hits were assessed for direct binding to a P1 peptide and the inhibition of amyloid formation. We discovered a sybody, named αSP1, that inhibits amyloid formation of α-synuclein at substoichiometric concentrations in a specific manner, even within highly crowded heterogeneous mixtures. Fluorescence resonance energy transfer-based binding assays and seeding experiments with and without αSP1 further demonstrate the importance of the P1 region for both primary and secondary nucleation mechanisms of amyloid assembly.


Subject(s)
Amyloid , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/antagonists & inhibitors , Humans , Amyloid/metabolism , Amyloid/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Protein Binding
2.
Int J Mol Sci ; 22(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923609

ABSTRACT

Neurodegenerative disorders are a highly prevalent class of diseases, whose pathological mechanisms start before the appearance of any clear symptoms. This fact has prompted scientists to search for biomarkers that could aid early treatment. These currently incurable pathologies share the presence of aberrant aggregates called amyloids in the nervous system, which are composed of specific proteins. In this review, we discuss how these proteins, their conformations and modifications could be exploited as biomarkers for diagnostic purposes. We focus on proteins that are associated with the most prevalent neurodegenerative disorders, including Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, and frontotemporal dementia. We also describe current challenges in detection, the most recent techniques with diagnostic potentials and possible future developments in diagnosis.


Subject(s)
Amyloid/metabolism , Neurodegenerative Diseases/metabolism , Protein Aggregation, Pathological/metabolism , Amyloid/chemistry , Amyloid/genetics , Animals , Biomarkers/metabolism , Humans , Neurodegenerative Diseases/pathology , Protein Aggregation, Pathological/pathology
3.
Sci Rep ; 11(1): 6101, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33731745

ABSTRACT

Individual cells exhibit specific proliferative responses to changes in microenvironmental conditions. Whether such potential is constrained by the cell density throughout the growth process is however unclear. Here, we identify a theoretical framework that captures how the information encoded in the initial density of cancer cell populations impacts their growth profile. By following the growth of hundreds of populations of cancer cells, we found that the time they need to adapt to the environment decreases as the initial cell density increases. Moreover, the population growth rate shows a maximum at intermediate initial densities. With the support of a mathematical model, we show that the observed interdependence of adaptation time and growth rate is significantly at odds both with standard logistic growth models and with the Monod-like function that governs the dependence of the growth rate on nutrient levels. Our results (i) uncover and quantify a previously unnoticed heterogeneity in the growth dynamics of cancer cell populations; (ii) unveil how population growth may be affected by single-cell adaptation times; (iii) contribute to our understanding of the clinically-observed dependence of the primary and metastatic tumor take rates on the initial density of implanted cancer cells.


Subject(s)
Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , Humans , Jurkat Cells , Neoplasm Metastasis
4.
J Mol Biol ; 429(12): 1800-1816, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28501587

ABSTRACT

In previous work, we have generated the engineered Escherichia coli strains SuptoxD and SuptoxR, which upon co-expression of the effector genes djlA or rraA, respectively, are capable of suppressing the cytotoxicity caused by membrane protein (MP) overexpression and of producing dramatically enhanced yields for a variety of recombinant MPs of both prokaryotic and eukaryotic origin. Here, we investigated the functional requirements for DnaJ-like protein A (DjlA)- and regulator of ribonuclease activity A (RraA)-mediated enhancement of recombinant MP production in these strains and show that: (i) DjlA and RraA act independently, that is, the beneficial effects of each protein on recombinant MP production occur through a mechanism that does not involve the other, and in a non-additive manner; (ii) full-length and membrane-bound DjlA is required for exerting its beneficial effects on recombinant MP production in E. coli SuptoxD; (iii) the MP production-promoting properties of DjlA in SuptoxD involve the action of the molecular chaperone DnaK but do not rely on the activation of the regulation of capsular synthesis response, a well-established consequence of djlA overexpression; (iv) the observed RraA-mediated effects in E. coli SuptoxR involve the ribonucleolytic activity of RNase E, but not that of its paralogous ribonuclease RNase G; and (v) DjlA and RraA are unique among similar E. coli proteins in their ability to promote bacterial recombinant MP production. These observations provide important clues about the molecular requirements for suppressed toxicity and enhanced MP accumulation in SuptoxD/SuptoxR and will guide future studies aiming to decipher the exact mechanism of DjlA- and RraA-mediated enhancement of recombinant MP production in these strains.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , HSP40 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Recombinant Proteins/metabolism , Bacterial Proteins , Escherichia coli/genetics , Escherichia coli Proteins/genetics , HSP40 Heat-Shock Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/toxicity , Metabolic Engineering , Recombinant Proteins/genetics , Recombinant Proteins/toxicity
5.
ACS Synth Biol ; 6(2): 284-300, 2017 02 17.
Article in English | MEDLINE | ID: mdl-27797488

ABSTRACT

Membrane proteins perform critical cellular functions in all living organisms and constitute major targets for drug discovery. Escherichia coli has been the most popular overexpression host for membrane protein biochemical/structural studies. Bacterial production of recombinant membrane proteins, however, is typically hampered by poor cellular accumulation and severe toxicity for the host, which leads to low final biomass and minute volumetric yields. In this work, we aimed to rewire the E. coli protein-producing machinery to withstand the toxicity caused by membrane protein overexpression in order to generate engineered bacterial strains with the ability to achieve high-level membrane protein production. To achieve this, we searched for bacterial genes whose coexpression can suppress membrane protein-induced toxicity and identified two highly potent effectors: the membrane-bound DnaK cochaperone DjlA, and the inhibitor of the mRNA-degrading activity of the E. coli RNase E, RraA. E. coli strains coexpressing either djlA or rraA, termed SuptoxD and SuptoxR, respectively, accumulated markedly higher levels of final biomass and produced dramatically enhanced yields for a variety of prokaryotic and eukaryotic recombinant membrane proteins. In all tested cases, either SuptoxD, or SuptoxR, or both, outperformed the capabilities of commercial strains frequently utilized for recombinant membrane protein production purposes.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Membrane Proteins/genetics , Recombinant Proteins/genetics , Biomass , Endoribonucleases/genetics , Gene Expression Regulation, Bacterial/genetics , Genes, Bacterial/genetics , HSP40 Heat-Shock Proteins/genetics , RNA Stability/genetics , RNA, Messenger/genetics
6.
Microb Biotechnol ; 3(4): 403-11, 2010 Jul.
Article in English | MEDLINE | ID: mdl-21255339

ABSTRACT

The bacterium Escherichia coli is the most widely used expression host for overexpression trials of membrane proteins. Usually, different strains, culture conditions and expression regimes are screened for to identify the optimal overexpression strategy. However, yields are often not satisfactory, especially for eukaryotic membrane proteins. This has initiated a revolution of membrane protein overexpression in bacteria. Recent studies have shown that it is feasible to (i) engineer or select for E. coli strains with strongly improved membrane protein overexpression characteristics, (ii) use bacteria other than E. coli for the expression of membrane proteins, (iii) engineer or select for membrane protein variants that retain functionality but express better than the wild-type protein, and (iv) express membrane proteins using E. coli-based cell-free systems.


Subject(s)
Bacteria/metabolism , Gene Expression , Membrane Proteins/biosynthesis , Bacteria/genetics , Biotechnology/methods , Cell-Free System , Genetic Engineering/methods , Membrane Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...