Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
FEBS J ; 290(6): 1398-1419, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35122394

RESUMO

Light chain amyloidosis (AL) is a systemic disease in which abnormally proliferating plasma cells secrete large amounts of mutated antibody light chains (LCs) that eventually form fibrils. The fibrils are deposited in various organs, most often in the heart and kidney, and impair their function. The prognosis for patients diagnosed with AL is generally poor. The disease is set apart from other amyloidoses by the huge number of patient-specific mutations in the disease-causing and fibril-forming protein. The molecular mechanisms that drive the aggregation of mutated LCs into fibrils have been enigmatic, which hindered the development of efficient diagnostics and therapies. In this review, we summarize our current knowledge on AL amyloidosis and discuss open issues.


Assuntos
Amiloidose , Humanos , Amiloidose/genética , Amiloidose/metabolismo , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/metabolismo , Prognóstico , Plasmócitos/metabolismo , Anticorpos , Amiloide/genética , Amiloide/metabolismo
2.
Nat Commun ; 12(1): 6516, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34764275

RESUMO

In antibody light chain (AL) amyloidosis, overproduced light chain (LC) fragments accumulate as fibrils in organs and tissues of patients. In vitro, AL fibril formation is a slow process, characterized by a pronounced lag phase. The events occurring during this lag phase are largely unknown. We have dissected the lag phase of a patient-derived LC truncation and identified structural transitions that precede fibril formation. The process starts with partial unfolding of the VL domain and the formation of small amounts of dimers. This is a prerequisite for the formation of an ensemble of oligomers, which are the precursors of fibrils. During oligomerization, the hydrophobic core of the LC domain rearranges which leads to changes in solvent accessibility and rigidity. Structural transitions from an anti-parallel to a parallel ß-sheet secondary structure occur in the oligomers prior to amyloid formation. Together, our results reveal a rate-limiting multi-step mechanism of structural transitions prior to fibril formation in AL amyloidosis, which offers, in the long run, opportunities for therapeutic intervention.


Assuntos
Amiloide/metabolismo , Amiloidose de Cadeia Leve de Imunoglobulina/metabolismo , Amiloide/química , Estrutura Secundária de Proteína , Espectroscopia de Infravermelho com Transformada de Fourier
3.
Nat Commun ; 12(1): 6697, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795272

RESUMO

Hsp26 is a small heat shock protein (sHsp) from S. cerevisiae. Its chaperone activity is activated by oligomer dissociation at heat shock temperatures. Hsp26 contains 9 phosphorylation sites in different structural elements. Our analysis of phospho-mimetic mutations shows that phosphorylation activates Hsp26 at permissive temperatures. The cryo-EM structure of the Hsp26 40mer revealed contacts between the conserved core domain of Hsp26 and the so-called thermosensor domain in the N-terminal part of the protein, which are targeted by phosphorylation. Furthermore, several phosphorylation sites in the C-terminal extension, which link subunits within the oligomer, are sensitive to the introduction of negative charges. In all cases, the intrinsic inhibition of chaperone activity is relieved and the N-terminal domain becomes accessible for substrate protein binding. The weakening of domain interactions within and between subunits by phosphorylation to activate the chaperone activity in response to proteotoxic stresses independent of heat stress could be a general regulation principle of sHsps.


Assuntos
Proteínas de Choque Térmico/metabolismo , Multimerização Proteica , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Sítios de Ligação/genética , Dicroísmo Circular , Microscopia Crioeletrônica , Transferência Ressonante de Energia de Fluorescência , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Resposta ao Choque Térmico , Modelos Moleculares , Mutação , Fosforilação , Ligação Proteica , Conformação Proteica , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/ultraestrutura , Espectrometria de Massas em Tandem , Temperatura
4.
J Biol Chem ; 296: 100334, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33508322

RESUMO

Systemic light chain (AL) amyloidosis is a fatal protein misfolding disease in which excessive secretion, misfolding, and subsequent aggregation of free antibody light chains eventually lead to deposition of amyloid plaques in various organs. Patient-specific mutations in the antibody VL domain are closely linked to the disease, but the molecular mechanisms by which certain mutations induce misfolding and amyloid aggregation of antibody domains are still poorly understood. Here, we compare a patient VL domain with its nonamyloidogenic germline counterpart and show that, out of the five mutations present, two of them strongly destabilize the protein and induce amyloid fibril formation. Surprisingly, the decisive, disease-causing mutations are located in the highly variable complementarity determining regions (CDRs) but exhibit a strong impact on the dynamics of conserved core regions of the patient VL domain. This effect seems to be based on a deviation from the canonical CDR structures of CDR2 and CDR3 induced by the substitutions. The amyloid-driving mutations are not necessarily involved in propagating fibril formation by providing specific side chain interactions within the fibril structure. Rather, they destabilize the VL domain in a specific way, increasing the dynamics of framework regions, which can then change their conformation to form the fibril core. These findings reveal unexpected influences of CDR-framework interactions on antibody architecture, stability, and amyloid propensity.


Assuntos
Amiloide/ultraestrutura , Regiões Determinantes de Complementaridade/genética , Amiloidose de Cadeia Leve de Imunoglobulina/genética , Placa Amiloide/genética , Sequência de Aminoácidos/genética , Amiloide/genética , Amiloide/imunologia , Proteínas Amiloidogênicas/genética , Proteínas Amiloidogênicas/imunologia , Proteínas Amiloidogênicas/ultraestrutura , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/ultraestrutura , Humanos , Amiloidose de Cadeia Leve de Imunoglobulina/imunologia , Amiloidose de Cadeia Leve de Imunoglobulina/metabolismo , Mutação/genética , Placa Amiloide/imunologia , Placa Amiloide/patologia , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/imunologia , Agregação Patológica de Proteínas/patologia , Conformação Proteica , Dobramento de Proteína
5.
J Mol Biol ; 432(23): 6187-6199, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33058870

RESUMO

In antibody light chain amyloidosis (AL), mutant light chains (LCs) or their variable domains (VLs) form fibrils, which accumulate in organs and lead to their failure. The molecular mechanism of this disease is still poorly understood. One of the key open issues is whether the mutant VLs and LCs differ in fibril formation. We addressed this question studying the effects of the VL mutations S20N and R61A within the isolated VL domain and in the full-length LC scaffold. Both VL variants readily form fibrils. Here, we find that in the LC context, the S20N variant is protected from fibril formation while for LC R61A fibril formation is even accelerated compared to VL R61A. Our analyses revealed that the partially unfolded state of the VL R61A domain destabilizes the CL domain by non-native interactions, in turn leading to a further unfolding of the VL domain. In contrast, the folded mutant VL S20N and VL wt form native interactions with CL. These are beneficial for LC stability and promote amyloid resistance. Thus the effects of specific mutations on the VL fold can have opposing effects on LC domain interactions, stability and amyloidogenicity.


Assuntos
Amiloide/genética , Proteínas Amiloidogênicas/genética , Cadeias Leves de Imunoglobulina/imunologia , Agregação Patológica de Proteínas/genética , Sequência de Aminoácidos/genética , Amiloide/imunologia , Proteínas Amiloidogênicas/imunologia , Amiloidose/genética , Amiloidose/imunologia , Humanos , Cadeias Leves de Imunoglobulina/genética , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Agregação Patológica de Proteínas/imunologia , Conformação Proteica
6.
Elife ; 92020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32151314

RESUMO

In systemic light chain amyloidosis, an overexpressed antibody light chain (LC) forms fibrils which deposit in organs and cause their failure. While it is well-established that mutations in the LC's VL domain are important prerequisites, the mechanisms which render a patient LC amyloidogenic are ill-defined. In this study, we performed an in-depth analysis of the factors and mutations responsible for the pathogenic transformation of a patient-derived λ LC, by recombinantly expressing variants in E. coli. We show that proteolytic cleavage of the patient LC resulting in an isolated VL domain is essential for fibril formation. Out of 11 mutations in the patient VL, only one, a leucine to valine mutation, is responsible for fibril formation. It disrupts a hydrophobic network rendering the C-terminal segment of VL more dynamic and decreasing domain stability. Thus, the combination of proteolytic cleavage and the destabilizing mutation trigger conformational changes that turn the LC pathogenic.


Amyloid light chain amyloidosis, shortened to AL amyloidosis, is a rare and often fatal disease. It is caused by a disorder of the bone marrow. Usually, cells in the bone marrow produce Y-shaped proteins called antibodies to fight infections. In AL amyloidosis, these cells release too much of the short arm of the antibody, known as its light chain, and the light chains also carry mutations. The antibodies are no longer able to assemble properly, and instead misfold and form structures, known as amyloid fibrils. The fibrils build up outside the cells, gradually causing damage to tissues and organs that can lead to life-threatening organ failure. Due to the rareness of the disease, diagnosis is often overlooked and delayed. People experience widely varying symptoms, depending on the organs affected. Also, given the diversity of antibodies people make, every person with AL amyloidosis has a variety of mutations implicated in their disease. It is thought that mutations in the antibody light chain make it unstable and prone to misfolding, but it remains unclear which specific mutations trigger a cascade of amyloid fibril formation. Now, Kazman et al. have pinpointed the exact mechanism in one case of the disease. First, tissue biopsies from a woman with advanced AL amyloidosis were analyzed, and the defunct antibody light chain was isolated. Eleven mutations were identified in the antibody light chain, only one of which was found to be responsible for the formation of the harmful fibrils. The next step was to determine how this one small change was so damaging. The experiments showed that after the antibody light chain was cut in two, a process that happens naturally in the body, this single mutation transforms it into a protein capable of causing disease. In this 'bedside to lab bench' study, Kazman et al. have succeeded in determining the molecular origin of one case of AL amyloidosis. The results have also shown that the instability of antibodies due to mutation does not alone explain the formation of amyloid fibrils in this disease and that the cutting of this protein in two is also important. It is hoped that, in the long run, this work will lead to new diagnostics and treatment options for people with AL amyloidosis.


Assuntos
Amiloidose/patologia , Predisposição Genética para Doença , Cadeias Leves de Imunoglobulina/genética , Placa Amiloide/patologia , Sequência de Aminoácidos , Evolução Fatal , Feminino , Humanos , Pessoa de Meia-Idade , Modelos Moleculares , Mutação , Conformação Proteica , Dobramento de Proteína
7.
Heliyon ; 5(6): e01860, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31194064

RESUMO

The application of essential oils as antimicrobials is a current subject of research and a promising approach in terms of natural food preservation. Due to the diversity of EO producing plant genera and the inconsistent use of susceptibility testing methods, information on the antibacterial potency of many EO varieties is fragmentary. This study was performed to assess the minimal inhibitory concentrations (MIC) of 179 EO samples from 86 plant varieties, using a single method approach, excluding emulsifying agents. MICs were acquired in a broth microdilution assay, using a dispersion based approach to incorporate EOs in a concentration range of 6400 to 50 µg/ml. Staphylococcus aureus and Escherichia coli were used as model bacteria. At concentrations below 400 µg/ml S. aureus was inhibited by 30, E. coli by 12 EO varieties. Azadirachta indica (50 µg/ml vs. S. aureus) and Litsea cubeba (50 µg/ml vs. S. aureus, 200 µg/ml vs. E. coli) essential oils were identified as promising new antimicrobial EO candidates with significant antimicrobial activity against the two foodborne pathogenic bacteria.

8.
J Mol Biol ; 430(24): 4925-4940, 2018 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-30414962

RESUMO

The antibody light chain (LC) consists of two domains and is essential for antigen binding in mature immunoglobulins. The two domains are connected by a highly conserved linker that comprises the structurally important Arg108 residue. In antibody light chain (AL) amyloidosis, a severe protein amyloid disease, the LC and its N-terminal variable domain (VL) convert to fibrils deposited in the tissues causing organ failure. Understanding the factors shaping the architecture of the LC is important for basic science, biotechnology and for deciphering the principles that lead to fibril formation. In this study, we examined the structure and properties of LC variants with a mutated or extended linker. We show that under destabilizing conditions, the linker modulates the amyloidogenicity of the LC. The fibril formation propensity of LC linker variants and their susceptibility to proteolysis directly correlate implying an interplay between the two LC domains. Using NMR and residual dipolar coupling-based simulations, we found that the linker residue Arg108 is a key factor regulating the relative orientation of the VL and CL domains, keeping them in a bent and dense, but still flexible conformation. Thus, inter-domain contacts and the relative orientation of VL and CL to each other are of major importance for maintaining the structural integrity of the full-length LC.


Assuntos
Cadeias Leves de Imunoglobulina/química , Cadeias Leves de Imunoglobulina/metabolismo , Mutação , Agregação Patológica de Proteínas/metabolismo , Amiloide/química , Amiloide/genética , Amiloide/metabolismo , Arginina/metabolismo , Sítios de Ligação , Humanos , Cadeias Leves de Imunoglobulina/genética , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Domínios Proteicos , Proteólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA