Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(4): 107174, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499153

ABSTRACT

AL amyloidosis is a life-threatening disease caused by deposition of immunoglobulin light chains. While the mechanisms underlying light chains amyloidogenesis in vivo remain unclear, several studies have highlighted the role that tissue environment and structural amyloidogenicity of individual light chains have in the disease pathogenesis. AL natural deposits contain both full-length light chains and fragments encompassing the variable domain (VL) as well as different length segments of the constant region (CL), thus highlighting the relevance that proteolysis may have in the fibrillogenesis pathway. Here, we investigate the role of major truncated species of the disease-associated AL55 light chain that were previously identified in natural deposits. Specifically, we study structure, molecular dynamics, thermal stability, and capacity to form fibrils of a fragment containing both the VL and part of the CL (133-AL55), in comparison with the full-length protein and its variable domain alone, under shear stress and physiological conditions. Whereas the full-length light chain forms exclusively amorphous aggregates, both fragments generate fibrils, although, with different kinetics, aggregate structure, and interplay with the unfragmented protein. More specifically, the VL-CL 133-AL55 fragment entirely converts into amyloid fibrils microscopically and spectroscopically similar to their ex vivo counterpart and increases the amorphous aggregation of full-length AL55. Overall, our data support the idea that light chain structure and proteolysis are both relevant for amyloidogenesis in vivo and provide a novel biocompatible model of light chain fibrillogenesis suitable for future mechanistic studies.


Subject(s)
Amyloid , Immunoglobulin Light Chains , Amyloid/metabolism , Amyloid/chemistry , Humans , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , Molecular Dynamics Simulation , Immunoglobulin Constant Regions/metabolism , Immunoglobulin Constant Regions/genetics , Immunoglobulin Constant Regions/chemistry , Immunoglobulin Light-chain Amyloidosis/metabolism , Immunoglobulin Light-chain Amyloidosis/pathology , Kinetics , Protein Domains
2.
Vet Pathol ; : 3009858241257903, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864284

ABSTRACT

AA-amyloidosis is frequent in shelter cats, and chronic kidney disease is the foremost cause of death. The aims were to describe kidney laboratory and microscopic findings in shelter cats with AA-amyloidosis. Cats were included if kidney specimens were collected post-mortem and laboratory data were available within 6 months before death. Renal lesions were evaluated with optical and electron microscopy. Mass spectrometry was used to characterize amyloid. Nine domestic short-hair cats were included; 4 females and 5 males with a median age of 8 years (range = 2-13). All cats had blood analyses and urinalyses available. Serum creatinine concentrations were increased in 6 cats and symmetric dimethylarginine was increased in all of the cats. All of the cats had proteinuria. Eight of 9 cats had amyloid in the medulla, and 9 had amyloid in the cortex (glomeruli). All cats had amyloid in the interstitium. Six cats had concurrent interstitial nephritis and 1 had membranoproliferative glomerulonephritis. All cats had extrarenal amyloid deposits. Amyloid was AA in each case. In conclusion, renal deposition of amyloid occurs in both cortex and medulla in shelter cats and is associated with azotemia and proteinuria. Renal involvement of systemic AA-amyloidosis should be considered in shelter cats with chronic kidney disease. The cat represents a natural model of renal AA-amyloidosis.

3.
J Biol Chem ; 295(49): 16572-16584, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32952127

ABSTRACT

Amyloid fibrils are polymeric structures originating from aggregation of misfolded proteins. In vivo, proteolysis may modulate amyloidogenesis and fibril stability. In light chain (AL) amyloidosis, fragmented light chains (LCs) are abundant components of amyloid deposits; however, site and timing of proteolysis are debated. Identification of the N and C termini of LC fragments is instrumental to understanding involved processes and enzymes. We investigated the N and C terminome of the LC proteoforms in fibrils extracted from the hearts of two AL cardiomyopathy patients, using a proteomic approach based on derivatization of N- and C-terminal residues, followed by mapping of fragmentation sites on the structures of native and fibrillar relevant LCs. We provide the first high-specificity map of proteolytic cleavages in natural AL amyloid. Proteolysis occurs both on the LC variable and constant domains, generating a complex fragmentation pattern. The structural analysis indicates extensive remodeling by multiple proteases, largely taking place on poorly folded regions of the fibril surfaces. This study adds novel important knowledge on amyloid LC processing: although our data do not exclude that proteolysis of native LC dimers may destabilize their structure and favor fibril formation, the data show that LC deposition largely precedes the proteolytic events documentable in mature AL fibrils.


Subject(s)
Amyloid/chemistry , Immunoglobulin Light-chain Amyloidosis/pathology , Myocardium/metabolism , Amino Acid Sequence , Amyloid/metabolism , Chromatography, High Pressure Liquid , Electrophoresis, Gel, Two-Dimensional , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light-chain Amyloidosis/metabolism , Peptides/analysis , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis , Tandem Mass Spectrometry
4.
Molecules ; 26(7)2021 Mar 29.
Article in English | MEDLINE | ID: mdl-33805439

ABSTRACT

Amyloidosis is a relatively rare human disease caused by the deposition of abnormal protein fibres in the extracellular space of various tissues, impairing their normal function. Proteomic analysis of patients' biopsies, developed by Dogan and colleagues at the Mayo Clinic, has become crucial for clinical diagnosis and for identifying the amyloid type. Currently, the proteomic approach is routinely used at National Amyloidosis Centre (NAC, London, UK) and Istituto di Tecnologie Biomediche-Consiglio Nazionale delle Ricerche (ITB-CNR, Milan, Italy). Both centres are members of the European Proteomics Amyloid Network (EPAN), which was established with the aim of sharing and discussing best practice in the application of amyloid proteomics. One of the EPAN's activities was to evaluate the quality and the confidence of the results achieved using different software and algorithms for protein identification. In this paper, we report the comparison of proteomics results obtained by sharing NAC proteomics data with the ITB-CNR centre. Mass spectrometric raw data were analysed using different software platforms including Mascot, Scaffold, Proteome Discoverer, Sequest and bespoke algorithms developed for an accurate and immediate amyloid protein identification. Our study showed a high concordance of the obtained results, suggesting a good accuracy of the different bioinformatics tools used in the respective centres. In conclusion, inter-centre data exchange is a worthwhile approach for testing and validating the performance of software platforms and the accuracy of results, and is particularly important where the proteomics data contribute to a clinical diagnosis.


Subject(s)
Amyloidosis/diagnosis , Computational Biology , Information Dissemination , Proteomics/methods , Software , Algorithms , Amyloidogenic Proteins/metabolism , Amyloidosis/metabolism , Humans , Italy , United Kingdom
5.
Medicina (Kaunas) ; 57(9)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34577839

ABSTRACT

Amyloidoses are characterized by aggregation of proteins into highly ordered amyloid fibrils, which deposit in the extracellular space of tissues, leading to organ dysfunction. In AL (amyloid light chain) amyloidosis, the most common form in Western countries, the amyloidogenic precursor is a misfolding-prone immunoglobulin light chain (LC), which, in the systemic form, is produced in excess by a plasma cell clone and transported to target organs though blood. Due to the primary role that proteins play in the pathogenesis of amyloidoses, mass spectrometry (MS)-based proteomic studies have gained an established position in the clinical management and research of these diseases. In AL amyloidosis, in particular, proteomics has provided important contributions for characterizing the precursor light chain, the composition of the amyloid deposits and the mechanisms of proteotoxicity in target organ cells and experimental models of disease. This review will provide an overview of the major achievements of proteomic studies in AL amyloidosis, with a presentation of the most recent acquisitions and a critical discussion of open issues and ongoing trends.


Subject(s)
Amyloidosis , Immunoglobulin Light-chain Amyloidosis , Amyloid , Amyloidosis/genetics , Humans , Immunoglobulin Light Chains , Proteomics
6.
Anal Chem ; 91(4): 2894-2900, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30676723

ABSTRACT

Deposition of misfolded proteins as extracellular amyloid aggregates is the pathological hallmark of systemic amyloidoses. Subcutaneous fat acquired by fine needle aspiration is the preferred screening tissue in suspected patients. In this study we employed Fourier transform infrared (FTIR) spectroscopy in attenuated total reflection (ATR) to investigate human abdominal fat aspirates with the aim of detecting disease-related changes in the molecular structure and composition of the tissue and exploiting the potentiality of the method to discriminate between amyloid-positive and -negative samples. The absorption and second-derivative spectra of Congo Red (CR) positive and CR-negative specimens were analyzed by three multivariate methods in four spectral regions. The proposed ATR-FTIR method is label-free, rapid, and relatively inexpensive and requires minimal sample preparation. We found that the ATR-FTIR approach can differentiate fat aspirates containing amyloid deposits from control specimens with high sensitivity and specificity, both at 100 [89-100]%. It is worth noting that the wavenumbers most important for discrimination indicate that changes both in the protein conformation and in resident lipids are intrinsic features of affected subcutaneous fat in comparison with the CR-negative controls. In this proof of concept study, we show that this approach could be useful for assessing tissue amyloid aggregates and for acquiring novel knowledge of the molecular bases of the disease.


Subject(s)
Adipose Tissue/pathology , Amyloid/analysis , Amyloidosis/diagnosis , Spectroscopy, Fourier Transform Infrared/methods , Abdominal Fat/chemistry , Abdominal Fat/pathology , Adipose Tissue/chemistry , Humans , Multivariate Analysis
7.
Am J Physiol Heart Circ Physiol ; 316(5): H1158-H1166, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30875258

ABSTRACT

Cardiac dysfunction is the most frequent cause of morbidity and mortality in amyloid light chain (AL) amyloidosis caused by a clonal immunoglobulin light chain (LC). Previously published transgenic animal models of AL amyloidosis have not recapitulated the key phenotype of cardiac dysfunction seen in AL amyloidosis, which has limited our understanding of the disease mechanisms in vivo, as well as the development of targeted AL therapeutics. We have developed a transgenic zebrafish model in which a λ LC derived from a patient with AL amyloidosis is conditionally expressed in the liver under the control of the Gal4 upstream activation sequence enhancer system. Circulating LC levels of 125 µg/ml in these transgenic zebrafish are comparable to median pathological serum LC levels. Functional analysis links abnormal contractile function with evidence of cellular and molecular proteotoxicity in the heart, including increased cell death and autophagy. However, despite pathological and functional phenotypes analogous to human AL, the lifespan of the transgenic fish is comparable to control fish without the expressed AL-LC transgene. Nuclear labeling experiments suggest increased cardiac proliferation in the transgenic fish, which can be counteracted by treatment with a small molecule proliferation inhibitor leading to increased zebrafish mortality because of cardiac apoptosis and functional deterioration. This transgenic zebrafish model provides a platform to study underlying AL disease mechanisms in vivo further. NEW & NOTEWORTHY Heart failure is a major cause of mortality in amyloid light (AL) amyloidosis, yet it has been difficult to model in animals. We report the generation of a transgenic zebrafish model for AL amyloidosis with pathological concentration of circulating human light chain protein that results in cardiac dysfunction. The light chain toxicity triggers regeneration in the zebrafish heart resulting in functional compensation early in life, but with age develops into cardiac dysfunction.


Subject(s)
Amyloidosis/metabolism , Apoptosis , Cardiomyopathies/metabolism , Cell Proliferation , Immunoglobulin lambda-Chains/metabolism , Myocardium/metabolism , Regeneration , Amyloidosis/embryology , Amyloidosis/genetics , Amyloidosis/physiopathology , Animals , Animals, Genetically Modified , Cardiomyopathies/embryology , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Cardiotoxicity , Disease Models, Animal , Humans , Immunoglobulin lambda-Chains/genetics , Myocardium/pathology , Zebrafish
8.
Proteomics ; 18(7): e1700353, 2018 04.
Article in English | MEDLINE | ID: mdl-29389069

ABSTRACT

Detection and typing of amyloid deposits in tissues are two crucial steps in the management of systemic amyloidoses. The presence of amyloid deposits is routinely evaluated through Congo red staining, whereas proteomics is now a mainstay in the identification of the deposited proteins. In article number 1700236, Winter et al. [Proteomics 2017, 17, Issue 22] describe a novel method based on MALDI-MS imaging coupled to ion mobility separation and peptide filtering, to detect the presence of amyloid in histology samples and to identify its composition, while preserving the spatial distribution of proteins in tissues.


Subject(s)
Amyloidosis , Amyloid , Congo Red , Humans , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Blood ; 125(14): 2239-44, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25636337

ABSTRACT

Accurate diagnosis of systemic amyloidosis is necessary both for assessing the prognosis and for delineating the appropriate treatment. It is based on histologic evidence of amyloid deposits and characterization of the amyloidogenic protein. We prospectively evaluated the diagnostic performance of immunoelectron microscopy (IEM) of abdominal fat aspirates from 745 consecutive patients with suspected systemic amyloidoses. All cases were extensively investigated with clinical and laboratory data, with a follow-up of at least 18 months. The 423 (56.8%) cases with confirmed systemic forms were used to estimate the diagnostic performance of IEM. Compared with Congo-red-based light microscopy, IEM was equally sensitive (75% to 80%) but significantly more specific (100% vs 80%; P < .001). In amyloid light-chain (AL) amyloidosis, κ cases were more difficult to diagnose (sensitivity 71%), whereas the analysis of abdominal aspirate was informative in only 40% of patients with transthyretin amyloidosis. We found a high prevalence (20%) of a monoclonal component in patients with non-AL amyloidosis, highlighting the risk of misdiagnosis and the need for unequivocal amyloid typing. Notably, IEM identified correctly the specific form of amyloidosis in >99% of the cases. IEM of abdominal fat aspirates is an effective tool in the routine diagnosis of systemic amyloidoses.


Subject(s)
Abdominal Fat/chemistry , Amyloid/analysis , Amyloidosis/diagnosis , Amyloidosis/metabolism , Microscopy, Immunoelectron/methods , Adult , Aged , Aged, 80 and over , Amyloidosis/classification , Biopsy, Fine-Needle , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Young Adult
11.
Clin Chem Lab Med ; 55(11): 1734-1743, 2017 Oct 26.
Article in English | MEDLINE | ID: mdl-28343171

ABSTRACT

BACKGROUND: The measurement of circulating free light chain (FLC) is essential in the diagnosis, prognostic stratification and evaluation of response to therapy in light chain (AL) amyloidosis. For more than 10 years, this has been done with an immunonephelometric assay based on polyclonal antibodies (Freelite), and cutoffs for staging and response assessment have been validated with this method. Recently, a new assay based on monoclonal antibodies (N latex FLC) has been marketed in Europe. METHODS: We evaluated and compared the clinical performance of the two assays in 426 patients with newly diagnosed AL amyloidosis. RESULTS: We found suboptimal agreement between the two methods, with differences between values obtained with the Freelite and N latex FLC assays increasing with the concentration of clonal FLC. The diagnostic sensitivity of the Freelite (82%) and N latex FLC (84%) assays was similar, and both improved to 98% in combination with serum and urine immunofixation. The concentration of FLC measured with both methods had prognostic significance. Less pronounced decreases in FLC best predicted improved survival with the N latex FLC assay (33% vs. 50%), and there was poor concordance (84%) in discrimination of responders. CONCLUSIONS: The two assays have similar diagnostic and prognostic performance. However, they are not interchangeable, and follow-up should be done with either one. New response criteria are needed for the N latex FLC assay.


Subject(s)
Amyloidosis/diagnosis , Immunoassay/standards , Immunoglobulin Light Chains/blood , Aged , Antibodies, Monoclonal/immunology , Female , Humans , Immunoglobulin Light Chains/immunology , Immunoglobulin Light-chain Amyloidosis , Latex/chemistry , Male , Middle Aged , Nephelometry and Turbidimetry/standards , Prognosis
12.
Expert Rev Proteomics ; 13(11): 1017-1027, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27678147

ABSTRACT

INTRODUCTION: More than ten distinct forms of amyloidoses that can involve the heart have been described, classified according to which protein originates the deposits. Cardiac amyloid infiltration translates into progressive and often life-threatening cardiomyopathy, but disease severity, prognosis and treatment drastically differ according to the amyloidosis type. The notion that protein misfolding and aggregation play a more general role in human cardiomyopathies has further raised attention towards the definition of the proteotoxicity mechanisms. Areas covered: Mass spectrometry-based proteomics plays an important role as a diagnostic tool and for understanding the molecular bases of amyloid cardiomyopathies. The landscape of applications of proteomics to the study of cardiac amyloidoses and amyloid-related cardiotoxicity is summarized, with a critical synthesis of the major achievements. Expert commentary: Current strengths and limitations of proteomics in the clinical setting and in translational research on amyloid cardiomyopathy are discussed, with the foreseen potential future directions in the field.

13.
Blood ; 123(23): 3543-52, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24665135

ABSTRACT

Poor prognosis and limited therapeutic options characterize immunoglobulin light-chain (AL) amyloidosis with major heart involvement. Reliable experimental models are needed to study light-chain (LC)/heart interactions and to explore strategies for prevention of cardiac damage. We have exploited the nematode Caenorhabditis elegans as a novel tool, because its pharynx is evolutionarily related to the vertebrate heart. Our data demonstrate that the pharyngeal pumping of C elegans is significantly and selectively reduced by LCs from AL patients suffering from cardiomyopathy, but not by amyloid LCs with different organ tropism or nonamyloidogenic LCs from multiple myeloma. This functional alteration is dependent on the LC concentration and results in persistent pharyngeal dysfunction and in a significant reduction of the worms' lifespan. These manifestations are paralleled by an increase of mitochondrial reactive oxygen species and can be prevented by treatment with antioxidant agents. In conclusion, these data indicate that this nematode-based assay is a promising surrogate model for investigating the heart-specific toxicity of amyloidogenic LCs and for a rapid screening of new therapeutic strategies.


Subject(s)
Amyloidosis/diagnosis , Caenorhabditis elegans , Heart Diseases/diagnosis , Immunoglobulin Light Chains/immunology , Adult , Aged , Amyloidosis/immunology , Animals , Biological Assay , Cardiotoxins/isolation & purification , Cardiotoxins/pharmacology , Cell Survival/drug effects , Female , Heart Diseases/immunology , Humans , Male , Middle Aged , Multiple Myeloma/immunology , Pharynx/cytology , Pharynx/drug effects , Pharynx/physiology
15.
FASEB J ; 29(11): 4614-28, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26220173

ABSTRACT

In immunoglobulin (Ig) light-chain (LC) (AL) amyloidosis, AL deposition translates into life-threatening cardiomyopathy. Clinical and experimental evidence indicates that soluble cardiotoxic LCs are themselves harmful for cells, by which they are internalized. Hypothesizing that interaction of soluble cardiotoxic LCs with cellular proteins contributes to damage, we characterized their interactome in cardiac cells. LCs were purified from patients with AL amyloidosis cardiomyopathy or multiple myeloma without amyloidosis (the nonamyloidogenic/noncardiotoxic LCs served as controls) and employed at concentrations in the range observed in AL patients' sera. A functional proteomic approach, based on direct and inverse coimmunoprecipitation and mass spectrometry, allowed identifying LC-protein complexes. Findings were validated by colocalization, fluorescence lifetime imaging microscopy (FLIM)-fluorescence resonance energy transfer (FRET), and ultrastructural studies, using human primary cardiac fibroblasts (hCFs) and stem cell-derived cardiomyocytes. Amyloidogenic cardiotoxic LCs interact in vitro with specific intracellular proteins involved in viability and metabolism. Imaging confirmed that, especially in hCFs, cardiotoxic LCs (not controls) colocalize with mitochondria and spatially associate with selected interactors: mitochondrial optic atrophy 1-like protein and peroxisomal acyl-coenzyme A oxidase 1 (FLIM-FRET efficiencies 11 and 6%, respectively). Cardiotoxic LC-treated hCFs display mitochondrial ultrastructural changes, supporting mitochondrial involvement. We show that cardiotoxic LCs establish nonphysiologic protein-protein contacts in human cardiac cells, offering new clues on the pathogenesis of AL cardiomyopathy.


Subject(s)
Amyloidosis/metabolism , Fibroblasts/metabolism , Heart Diseases/metabolism , Immunoglobulin Light Chains/metabolism , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Adult , Amyloidosis/pathology , Animals , Female , Fibroblasts/pathology , Heart Diseases/pathology , Humans , Male , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley
17.
Clin Chem Lab Med ; 54(6): 939-45, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26943606

ABSTRACT

BACKGROUND: The measurement of circulating free light chains (FLC) is of utmost importance in immunoglobulin light chain (AL) amyloidosis, being a fundamental part of the diagnostic workup, prognostic stratification and assessment of response to therapy. Renal failure is a common feature of AL amyloidosis and can considerably affect the concentration of FLC. METHODS: We assessed the impact of renal failure on the clinical performance of the Freelite assay in 982 consecutive, newly diagnosed patients with AL amyloidosis, 822 with estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2, and 160 with eGFR <30 mL/min/1.73 m2. RESULTS: The diagnostic sensitivity of the κ/λ FLC ratio was lower for λ amyloidogenic FLC in patients with renal failure (81% vs. 60%, p<0.001) and the FLC concentration had no independent prognostic significance in patients with severe renal dysfunction. However, FLC response to chemotherapy could still discriminate patients with better outcome. CONCLUSIONS: Renal failure is a relevant interference factor when using the Freelite assay for the identification of the amyloidogenic light chain and for prognostic assessment in patients with AL amyloidosis and renal failure.


Subject(s)
Amyloidosis/diagnosis , Immunoglobulin kappa-Chains/blood , Immunoglobulin lambda-Chains/blood , Renal Insufficiency/diagnosis , Aged , Amyloidosis/blood , Amyloidosis/physiopathology , Amyloidosis/therapy , Female , Humans , Male , Middle Aged , Prognosis , Renal Insufficiency/blood , Renal Insufficiency/physiopathology , Renal Insufficiency/therapy
18.
Blood ; 119(8): 1844-7, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-21917755

ABSTRACT

Considering the important advances in treating specific types of systemic amyloidoses, unequivocal typing of amyloid deposits is now essential. Subcutaneous abdominal fat aspiration is the easiest, most common diagnostic procedure. We developed a novel, automated approach, based on Multidimensional Protein Identification Technology, for typing amyloidosis. Fat aspirates were obtained from patients with the most common systemic amyloidoses (ALλ, ALκ, transthyretin, and reactive amyloidosis), with Congo red score more than or equal to 3+, and nonaffected controls. Peptides from extracted and digested proteins were analyzed by Multidimensional Protein Identification Technology. On semiquantitative differential analysis (patients vs controls) of mass spectrometry data, specific proteins up-represented in patients were identified and used as deposit biomarkers. An algorithm was developed to classify patients according to type and abundance of amyloidogenic proteins in samples; in all cases, proteomic characterization was concordant with fibril identification by immunoelectron microscopy and consistent with clinical presentation. Our approach allows reliable amyloid classification using readily available fat aspirates.


Subject(s)
Amyloidosis/metabolism , Proteome/analysis , Proteomics/methods , Subcutaneous Fat/metabolism , Adult , Aged , Aged, 80 and over , Amyloid/analysis , Amyloid/chemistry , Amyloidosis/classification , Amyloidosis/pathology , Biopsy, Fine-Needle , Congo Red/chemistry , Female , Humans , Male , Mass Spectrometry , Middle Aged , Reproducibility of Results , Subcutaneous Fat/pathology
19.
Haematologica ; 99(4): 743-50, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24213149

ABSTRACT

The combination of oral melphalan and dexamethasone is considered standard therapy for patients with light-chain amyloidosis ineligible for autologous stem cell transplantation. However, previous trials reported different rates of response and survival, mainly because of the different proportions of high-risk patients. In the present study, including a total of 259 subjects, we treated 119 patients with full-dose melphalan and dexamethasone (dexamethasone 40 mg days 1-4), and 140 patients with advanced cardiac disease with an attenuated dexamethasone schedule (20 mg). Hematologic response rates were 76% in the full-dose group and 51% in the patients receiving the attenuated schedule; the corresponding complete response rates were 31% and 12%, respectively. The median survival was 7.4 years in the full-dose group and 20 months in the attenuated-dose group. Use of high-dose dexamethasone, amino-terminal pro-natriuretic peptide type-B >1800 ng/L, a difference between involved and uninvolved free light chains of >180 mg/L, troponin I >0.07 ng/mL, and response to therapy were independent prognostic determinants. In relapsed/refractory subjects bortezomib combinations granted high hematologic response rates (79% and 63%, respectively), proving the most effective rescue treatment after melphalan and dexamethasone. In summary, melphalan plus dexamethasone was highly effective with minimal toxicity, confirming its central role in the treatment of AL amyloidosis. Future randomized trials will clarify whether bortezomib is best used in frontline combination with melphalan and dexamethasone or as rescue treatment.


Subject(s)
Amyloidosis/drug therapy , Dexamethasone/administration & dosage , Immunoglobulin Light Chains , Melphalan/administration & dosage , Administration, Oral , Adult , Aged , Aged, 80 and over , Amyloidosis/complications , Amyloidosis/diagnosis , Amyloidosis/mortality , Dexamethasone/adverse effects , Drug Therapy, Combination , Female , Humans , Immunoglobulin Light-chain Amyloidosis , Male , Melphalan/adverse effects , Middle Aged , Retreatment , Treatment Failure , Treatment Outcome
20.
Clin Chem Lab Med ; 52(11): 1517-31, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24870609

ABSTRACT

Systemic amyloid diseases are characterized by widespread protein deposition as amyloid fibrils. Precise diagnostic framing is the prerequisite for a correct management of patients. This complex process is achieved through a series of steps, which include detection of the tissue amyloid deposits, identification of the amyloid type, demonstration of the amyloidogenic precursor, and evaluation of organ dysfunction/damage. Laboratory medicine plays a central role in the diagnosis and management of systemic amyloidoses, through the quantification of the amyloidogenic precursor and evaluation of end-organ damage using biomarkers.


Subject(s)
Amyloidosis/diagnosis , Biomarkers/metabolism , Amyloidosis/pathology , Amyloidosis/prevention & control , Apolipoproteins A/metabolism , Biomarkers/blood , Early Diagnosis , Humans , Immunoglobulin Light Chains/metabolism , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Prealbumin/metabolism , Proteomics , Troponin/blood
SELECTION OF CITATIONS
SEARCH DETAIL