1
|
Simon P, Behrens HM, Kristen A, Röcken C. Myocardial inflammatory cells in cardiac amyloidosis. Sci Rep 2024; 14:23313. [PMID: 39375494 PMCID: PMC11458899 DOI: 10.1038/s41598-024-74289-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Immunoglobulin derived AL amyloidosis and transthyretin derived ATTR amyloidosis are the most common forms of cardiac amyloidosis. Both may present with cardiac arrhythmias, heart failure, and extracardiac symptoms. Disease outcome is often fatal. Recently, it was proposed that amyloid may cause cardiac inflammation. Here we tested the hypothesis that immune cell infiltration in cardiac tissue correlates with clinicopathological patient characteristics. PATIENTS AND METHODS Myocardial biopsies from 157 patients with cardiac amyloidosis (46.5% AL, 53.3% ATTR) were immunohistochemically assessed for the presence and amount of T lymphocytes (CD3), macrophages (CD68) and neutrophils (MPO). Amyloid load, cardiomyocyte diameter, apoptosis (Caspase 3), necrosis (complement 9), and various clinical parameters were assessed and correlated with immune cell density. RESULTS Myocardial tissue was infiltrated with T lymphocytes (CD3), macrophages (CD68) and neutrophils (MPO) with variable amounts. Significant correlations were found between the number of macrophages and NYHA class. No correlations were found between the presence and amount of T lymphocytes, neutrophils and clinicopathological patient characteristics. CONCLUSION The significant correlation between cardiac macrophage density and heart failure points towards a significant role of macrophages in disease pathology.
Collapse
Affiliation(s)
- Philip Simon
- Department of Pathology, University Hospital Schleswig-Holstein, Christian-Albrechts-University, Arnold-Heller-Str. 3, Building U33, 24105, Kiel, Germany
| | - Hans-Michael Behrens
- Department of Pathology, University Hospital Schleswig-Holstein, Christian-Albrechts-University, Arnold-Heller-Str. 3, Building U33, 24105, Kiel, Germany
| | - Arnt Kristen
- Department of Cardiology, Angiology, Respiratory Medicine, Medical University of Heidelberg, Heidelberg, Germany
| | - Christoph Röcken
- Department of Pathology, University Hospital Schleswig-Holstein, Christian-Albrechts-University, Arnold-Heller-Str. 3, Building U33, 24105, Kiel, Germany.
| |
Collapse
|
2
|
Bellofatto IA, Nikolaou PE, Andreadou I, Canepa M, Carbone F, Ghigo A, Heusch G, Kleinbongard P, Maack C, Podesser BK, Stamatelopoulos K, Stellos K, Vilahur G, Montecucco F, Liberale L. Mechanisms of damage and therapies for cardiac amyloidosis: a role for inflammation? Clin Res Cardiol 2024:10.1007/s00392-024-02522-2. [PMID: 39167195 DOI: 10.1007/s00392-024-02522-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
The term cardiac amyloidosis (CA) refers to the accumulation of extracellular amyloid deposits in the heart because of different conditions often affecting multiple organs including brain, kidney and liver. Notably, cardiac involvement significantly impacts prognosis of amyloidosis, with cardiac biomarkers playing a pivotal role in prognostic stratification. Therapeutic management poses a challenge due to limited response to conventional heart failure therapies, necessitating targeted approaches aimed at preventing, halting or reversing amyloid deposition. Mechanisms underlying organ damage in CA are multifactorial, involving proteotoxicity, oxidative stress, and mechanical interference. While the role of inflammation in CA remains incompletely understood, emerging evidence suggests its potential contribution to disease progression as well as its utility as a therapeutic target. This review reports on the cardiac involvement in systemic amyloidosis, its prognostic role and how to assess it. Current and emerging therapies will be critically discussed underscoring the need for further efforts aiming at elucidating CA pathophysiology. The emerging evidence suggesting the contribution of inflammation to disease progression and its prognostic role will also be reviewed possibly offering insights into novel therapeutic avenues for CA.
Collapse
Affiliation(s)
- Ilaria Anna Bellofatto
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Panagiota Efstathia Nikolaou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece
| | - Marco Canepa
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- Cardiology Unit, Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, L.Go R. Benzi 10, 16132, Genoa, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, Turin, Italy
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), and Medical Clinic I, University Clinic Würzburg, Würzburg, Germany
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Stellos
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de La Santa Creu I Sant Pau, IIB-Sant Pau, C/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, L.Go R. Benzi 10, 16132, Genoa, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, L.Go R. Benzi 10, 16132, Genoa, Italy.
| |
Collapse
|
3
|
Wang J, Li J, Zhong L. Current status and prospect of anti-amyloid fibril therapy in AL amyloidosis. Blood Rev 2024; 66:101207. [PMID: 38692939 DOI: 10.1016/j.blre.2024.101207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/11/2024] [Accepted: 04/14/2024] [Indexed: 05/03/2024]
Abstract
Amyloid light-chain (AL) amyloidosis is a rare hematological disease that produces abnormal monoclonal immunoglobulin light chains to form amyloid fibrils that are deposited in tissues, resulting in organ damage and dysfunction. Advanced AL amyloidosis has a very poor prognosis with a high risk of early mortality. The combination of anti-plasma cell therapy and amyloid fibrils clearance is the optimal treatment strategy, which takes into account both symptoms and root causes. However, research on anti-amyloid fibrils lags far behind research on anti-plasma cells, and there is currently no approved treatment that could clear amyloid fibrils. Nevertheless, anti-amyloid fibril therapies are being actively investigated recently and have shown potential in clinical trials. In this review, we aim to outline the preclinical work and clinical efficacy of fibril-directed therapies for AL amyloidosis.
Collapse
Affiliation(s)
- Jinghua Wang
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jian Li
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Liye Zhong
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Zhu L, Xie Z, Yang G, Zhou G, Li L, Zhang S. Stanniocalcin-1 Promotes PARP1-Dependent Cell Death via JNK Activation in Colitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304123. [PMID: 38088577 PMCID: PMC10837357 DOI: 10.1002/advs.202304123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/28/2023] [Indexed: 02/04/2024]
Abstract
Stanniocalcin-1 (STC1) is upregulated by inflammation and modulates oxidative stress-induced cell death. Herein, the function of STC1 in colitis and stress-induced parthanatos, a newly identified type of programmed necrotic cell death dependent on the activation of poly-ADP ribose polymerase-1 (PARP1) is investigated. Results show that STC1 expression is markedly increased in the inflamed colonic mucosa of Crohn's disease (CD) patients and chemically-induced mice colitis models. Evaluation of parthanatos severity and pro-inflammatory cytokine expression shows that intestinal-specific Stc1 knockout (Stc1INT-KO ) mice are resistant to dextran sulfate sodium (DSS)-induced colitis and exhibit lower disease severity. STC1-overexpressing cells show an increased degree of parthanatos and proinflammatory cytokine expression, whereas STC1-knockout cells show a decreased degree of parthanatos. Co-immunoprecipitation, mass spectrometry, and proteomic analyses indicate that STC1 interacts with PARP1, which activates the JNK pathway via PARP1-JNK interactions. Moreover, inhibition of PARP1 and JNK alleviates parthanatos and inflammatory injuries triggered by STC1 overexpression. Finally, following restoration of Stc1 and Parp1 expression by adeno-associated viruses, and overexpression of Stc1 and Parp1 aggravated DSS-induced colitis in Stc1INT-KO mice. In conclusion, STC1 mediates oxidative stress-associated parthanatos and aggravates inflammation via the STC1-PARP1-JNK interactions and subsequent JNK pathway activation in CD pathogenesis.
Collapse
Affiliation(s)
- Liguo Zhu
- Department of GastroenterologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| | - Zhuo Xie
- Department of GastroenterologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| | - Guang Yang
- Department of Minimally Invasive InterventionState Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Gaoshi Zhou
- Department of GastroenterologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| | - Li Li
- Department of GastroenterologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| | - Shenghong Zhang
- Department of GastroenterologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| |
Collapse
|
5
|
Golatkar V, Bhatt LK. Emerging therapeutic avenues in cardiac amyloidosis. Eur J Pharmacol 2023; 960:176142. [PMID: 37866746 DOI: 10.1016/j.ejphar.2023.176142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/15/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Cardiac Amyloidosis (CA) is a toxic infiltrative cardiomyopathy occurred by the deposition of the amyloid fibres in the extracellular matrix of the myocardium. This results in severe clinical complications such as increased left ventricular wall thickness and interventricular stiffness, a decrease in left ventricular stroke volume and cardiac output, diastolic dysfunction, arrhythmia, etc. In a prolonged period, this condition progresses into heart failure. The amyloid fibres affecting the heart include immunoglobulin light chain (AL - amyloidosis) and transthyretin protein (ATTR - amyloidosis) misfolded amyloid fibres. ATTRwt has the highest prevalence of 155 to 191 cases per million while ATTRv has an estimated prevalence of 5.2 cases per million. The pathological findings and therapeutic approaches developed recently have aided in the treatment regimen of cardiac amyloidosis patients. In recent years, understanding the pathophysiology of amyloid fibres formation and mechanistic pathways triggered in both types of cardiac amyloidosis has led to the development of new therapeutic approaches and agents. This review focuses on the current status of emerging therapeutic agents in clinical trials. Earlier, melphalan and bortezomib in combination with alkylating agents and immunomodulatory drugs were used as a standard therapy for AL amyloidosis. Tafamidis, approved recently by FDA is used as a standard for ATTR amyloidosis. However, the emerging therapeutic agents under development for the treatment of AL and ATTR cardiac amyloidosis have shown a potent and rapid effect with a safety profile.
Collapse
Affiliation(s)
- Vaishnavi Golatkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
6
|
Del Pozo-Yauner L, Herrera GA, Perez Carreon JI, Turbat-Herrera EA, Rodriguez-Alvarez FJ, Ruiz Zamora RA. Role of the mechanisms for antibody repertoire diversification in monoclonal light chain deposition disorders: when a friend becomes foe. Front Immunol 2023; 14:1203425. [PMID: 37520549 PMCID: PMC10374031 DOI: 10.3389/fimmu.2023.1203425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/20/2023] [Indexed: 08/01/2023] Open
Abstract
The adaptive immune system of jawed vertebrates generates a highly diverse repertoire of antibodies to meet the antigenic challenges of a constantly evolving biological ecosystem. Most of the diversity is generated by two mechanisms: V(D)J gene recombination and somatic hypermutation (SHM). SHM introduces changes in the variable domain of antibodies, mostly in the regions that form the paratope, yielding antibodies with higher antigen binding affinity. However, antigen recognition is only possible if the antibody folds into a stable functional conformation. Therefore, a key force determining the survival of B cell clones undergoing somatic hypermutation is the ability of the mutated heavy and light chains to efficiently fold and assemble into a functional antibody. The antibody is the structural context where the selection of the somatic mutations occurs, and where both the heavy and light chains benefit from protective mechanisms that counteract the potentially deleterious impact of the changes. However, in patients with monoclonal gammopathies, the proliferating plasma cell clone may overproduce the light chain, which is then secreted into the bloodstream. This places the light chain out of the protective context provided by the quaternary structure of the antibody, increasing the risk of misfolding and aggregation due to destabilizing somatic mutations. Light chain-derived (AL) amyloidosis, light chain deposition disease (LCDD), Fanconi syndrome, and myeloma (cast) nephropathy are a diverse group of diseases derived from the pathologic aggregation of light chains, in which somatic mutations are recognized to play a role. In this review, we address the mechanisms by which somatic mutations promote the misfolding and pathological aggregation of the light chains, with an emphasis on AL amyloidosis. We also analyze the contribution of the variable domain (VL) gene segments and somatic mutations on light chain cytotoxicity, organ tropism, and structure of the AL fibrils. Finally, we analyze the most recent advances in the development of computational algorithms to predict the role of somatic mutations in the cardiotoxicity of amyloidogenic light chains and discuss the challenges and perspectives that this approach faces.
Collapse
Affiliation(s)
- Luis Del Pozo-Yauner
- Department of Pathology, University of South Alabama-College of Medicine, Mobile, AL, United States
| | - Guillermo A. Herrera
- Department of Pathology, University of South Alabama-College of Medicine, Mobile, AL, United States
| | | | - Elba A. Turbat-Herrera
- Department of Pathology, University of South Alabama-College of Medicine, Mobile, AL, United States
- Mitchell Cancer Institute, University of South Alabama-College of Medicine, Mobile, AL, United States
| | | | | |
Collapse
|
7
|
Aurich M, Bucur J, Vey JA, Greiner S, Aus dem Siepen F, Hegenbart U, Schönland S, Katus HA, Frey N, Mereles D. Prognosis of light chain amyloidosis: a multivariable analysis for survival prediction in patients with cardiac involvement proven by endomyocardial biopsy. Open Heart 2023; 10:e002310. [PMID: 37463823 PMCID: PMC10357742 DOI: 10.1136/openhrt-2023-002310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/16/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Cardiac involvement is a main determinant of mortality in light chain (AL) amyloidosis but data on survival of patients with cardiac AL amyloidosis proven by endomyocardial biopsy (EMB) are sparse. METHODS This study analysed clinical, laboratory, electrocardiography and echocardiographic parameters for their prognostic value in the assessment of patients with AL amyloidosis and cardiac involvement. Patients with AL amyloidosis who had their first visit to the amyloidosis centre at the University Hospital Heidelberg between 2006 and 2017 (n=1628) were filtered for cardiac involvement proven by EMB. In the final cohort, mortality-associated markers were analysed by univariate and multivariable Cox regression. Cut-off values for each parameter were calculated using the survival time. RESULTS One-hundred and seventy-four patients could be identified. Median overall survival time was 1.5 years and median follow-up time was 5.2 years. At the end of the investigation period, 115 patients had died. In multivariable analysis, New York Heart Association-functional class >II (HR 1.65; 95% CI 1.09 to 2.50; p=0.019), left ventricular global longitudinal strain (HR 1.12; 95% CI 1.03 to 1.22; p=0.007), left ventricular end-systolic volume (HR 1.02; 95% CI 1.01 to 1.03; p=0.001), systolic pulmonary artery pressure (HR 0.98; 95% CI 0.96 to 0.99; p=0.027), N-terminal pro-B-type natriuretic peptide (HR 1.57; 95% CI 1.17 to 2.11; p=0.003) and difference in free light chains (HR 1.30; 95% CI 1.05 to 1.62; p=0.017) were independently predictive. CONCLUSION Among all patients with AL amyloidosis those with cardiac involvement represent a high-risk population with limited therapy options. Therefore, accurate risk stratification is necessary to identify cardiac amyloidosis patients with favourable prognosis. Incorporation of modern imaging techniques into existing or newly developed scoring systems is a promising option that might enable the implementation of risk-adapted therapeutic strategies.
Collapse
Affiliation(s)
- Matthias Aurich
- Department of Internal Medicine III, Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Julian Bucur
- Department of Internal Medicine III, Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Johannes A Vey
- Institute of Medical Biometry, University of Heidelberg, Heidelberg, Germany
| | - Sebastian Greiner
- Department of Internal Medicine III, Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fabian Aus dem Siepen
- Department of Internal Medicine III, Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Schönland
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- Department of Internal Medicine III, Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Derliz Mereles
- Department of Internal Medicine III, Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
8
|
The Protein Network in Subcutaneous Fat Biopsies from Patients with AL Amyloidosis: More Than Diagnosis? Cells 2023; 12:cells12050699. [PMID: 36899835 PMCID: PMC10000381 DOI: 10.3390/cells12050699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/11/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
AL amyloidosis is caused by the misfolding of immunoglobulin light chains leading to an impaired function of tissues and organs in which they accumulate. Due to the paucity of -omics profiles from undissected samples, few studies have addressed amyloid-related damage system wide. To fill this gap, we evaluated proteome changes in the abdominal subcutaneous adipose tissue of patients affected by the AL isotypes κ and λ. Through our retrospective analysis based on graph theory, we have herein deduced new insights representing a step forward from the pioneering proteomic investigations previously published by our group. ECM/cytoskeleton, oxidative stress and proteostasis were confirmed as leading processes. In this scenario, some proteins, including glutathione peroxidase 1 (GPX1), tubulins and the TRiC complex, were classified as biologically and topologically relevant. These and other results overlap with those already reported for other amyloidoses, supporting the hypothesis that amyloidogenic proteins could induce similar mechanisms independently of the main fibril precursor and of the target tissues/organs. Of course, further studies based on larger patient cohorts and different tissues/organs will be essential, which would be a key point that would allow for a more robust selection of the main molecular players and a more accurate correlation with clinical aspects.
Collapse
|
9
|
Lu J, Zhao P, Qiao J, Yang Z, Tang D, Zhou X, Huang L, Xia L. The major factor of left ventricular systolic dysfunction in patients with cardiac amyloidosis: Amyloid overload or microcirculation impairment? Front Cardiovasc Med 2023; 10:1096130. [PMID: 36776256 PMCID: PMC9909486 DOI: 10.3389/fcvm.2023.1096130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Purpose Amyloid overload and microcirculation impairment are both detrimental to left ventricular (LV) systolic function, while it is not clear which factor dominates LV functional remodeling in patients with cardiac amyloidosis (CA). The purpose of this study was to investigate the major factor of LV systolic dysfunction using cardiac magnetic resonance imaging. Materials and methods Forty CA patients and 20 healthy controls were included in this study. The CA group was divided into two subgroups by the left ventricular ejection fraction (LVEF): patients with reduced LVEF (LVEF < 50%, rLVEF), and patients with preserved LVEF (LVEF ≥ 50%, pLVEF). The scanning sequences included cine, native and post-contrast T1 mapping, rest first-pass perfusion and late gadolinium enhancement. Perfusion and mapping parameters were compared among the three groups. Correlation analysis was performed to evaluate the relationship between LVEF and mapping parameters, as well as the relationship between LVEF and perfusion parameters. Results Remarkably higher native T1 value was observed in the rLVEF patients than the pLVEF patients (1442.2 ± 85.8 ms vs. 1407.0 ± 93.9 ms, adjusted p = 0.001). The pLVEF patients showed significantly lower slope dividing baseline signal intensity (slope%BL; rLVEF vs. pLVEF, 55.1 ± 31.0 vs. 46.2 ± 22.3, adjusted p = 0.001) and a lower maximal signal intensity subtracting baseline signal intensity (MaxSI-BL; rLVEF vs. pLVEF, 43.5 ± 23.9 vs. 37.0 ± 18.6, adjusted p = 0.003) compared to the rLVEF patients. CA patients required more time to reach the maximal signal intensity than the controls did (all adjusted p < 0.01). There was no significant correlation between LVEF and first-pass perfusion parameters, while significant negative correlation was observed between LVEF and native T1 (r = -0.434, p = 0.005) in CA patients. Conclusion Amyloid overload in the myocardial interstitium may be the major factor of LV systolic dysfunction in CA patients, other than microcirculation impairment.
Collapse
Affiliation(s)
- Jianyao Lu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peijun Zhao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,The Department of MRI, First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jinhan Qiao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoxia Yang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dazhong Tang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyue Zhou
- MR Collaboration, Siemens Healthineers Ltd., Shanghai, China
| | - Lu Huang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Lu Huang, ✉
| | - Liming Xia
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Liming Xia, ✉
| |
Collapse
|
10
|
Riefolo M, Conti M, Longhi S, Fabbrizio B, Leone O. Amyloidosis: What does pathology offer? The evolving field of tissue biopsy. Front Cardiovasc Med 2022; 9:1081098. [PMID: 36545023 PMCID: PMC9760761 DOI: 10.3389/fcvm.2022.1081098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Since the mid-nineteenth century pathology has followed the convoluted story of amyloidosis, recognized its morphology in tissues and made identification possible using specific staining. Since then, pathology studies have made a significant contribution and advanced knowledge of the disease, so providing valuable information on the pathophysiology of amyloid aggregation and opening the way to clinical studies and non-invasive diagnostic techniques. As amyloidosis is a heterogeneous disease with various organ and tissue deposition patterns, histology evaluation, far from offering a simple yes/no indication of amyloid presence, can provide a wide spectrum of qualitative and quantitative information related to and changing with the etiology of the disease, the comorbidities and the clinical characteristics of patients. With the exception of cardiac transthyretin related amyloidosis cases, which today can be diagnosed using non-biopsy algorithms when stringent clinical criteria are met, tissue biopsy is still an essential tool for a definitive diagnosis in doubtful cases and also to define etiology by typing amyloid fibrils. This review describes the histologic approach to amyloidosis today and the current role of tissue screening biopsy or targeted organ biopsy protocols in the light of present diagnostic algorithms and various clinical situations, with particular focus on endomyocardial and renal biopsies. Special attention is given to techniques for typing amyloid fibril proteins, necessary for the new therapies available today for cardiac transthyretin related amyloidosis and to avoid patients receiving inappropriate chemotherapy in presence of plasma cell dyscrasia unrelated to amyloidosis. As the disease is still burdened with high mortality, the role of tissue biopsy in early diagnosis to assure prompt treatment is also mentioned.
Collapse
Affiliation(s)
- Mattia Riefolo
- Cardiovascular and Cardiac Transplant Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Matteo Conti
- Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Public Health Department, AUSL Imola, Bologna, Italy
| | - Simone Longhi
- Department of Cardiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Benedetta Fabbrizio
- Department of Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Ornella Leone
- Cardiovascular and Cardiac Transplant Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
11
|
Martinez-Rivas G, Bender S, Sirac C. Understanding AL amyloidosis with a little help from in vivo models. Front Immunol 2022; 13:1008449. [PMID: 36458006 PMCID: PMC9707859 DOI: 10.3389/fimmu.2022.1008449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/27/2022] [Indexed: 08/01/2023] Open
Abstract
Monoclonal immunoglobulin (Ig) light chain amyloidosis (AL) is a rare but severe disease that may occur when a B or plasma cell clone secretes an excess of free Ig light chains (LCs). Some of these LCs tend to aggregate into organized fibrils with a β-sheet structure, the so-called amyloid fibrils, and deposit into the extracellular compartment of organs, such as the heart or kidneys, causing their dysfunction. Recent findings have confirmed that the core of the amyloid fibrils is constituted by the variable (V) domain of the LCs, but the mechanisms underlying the unfolding and aggregation of this fragment and its deposition are still unclear. Moreover, in addition to the mechanical constraints exerted by the massive accumulation of amyloid fibrils in organs, the direct toxicity of these variable domain LCs, full-length light chains, or primary amyloid precursors (oligomers) seems to play a role in the pathogenesis of the disease. Many in vitro studies have focused on these topics, but the variability of this disease, in which each LC presents unique properties, and the extent and complexity of affected organs make its study in vivo very difficult. Accordingly, several groups have focused on the development of animal models for years, with some encouraging but mostly disappointing results. In this review, we discuss the experimental models that have been used to better understand the unknowns of this pathology with an emphasis on in vivo approaches. We also focus on why reliable AL amyloidosis animal models remain so difficult to obtain and what this tells us about the pathophysiology of the disease.
Collapse
|
12
|
McNulty MT, Fermin D, Eichinger F, Jang D, Kretzler M, Burtt NP, Pollak MR, Flannick J, Weins A, Friedman DJ, Sampson MG. A glomerular transcriptomic landscape of apolipoprotein L1 in Black patients with focal segmental glomerulosclerosis. Kidney Int 2022; 102:136-148. [PMID: 34929253 PMCID: PMC9206042 DOI: 10.1016/j.kint.2021.10.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
Apolipoprotein L1 (APOL1)-associated focal segmental glomerulosclerosis (FSGS) is the dominant form of FSGS in Black individuals. There are no targeted therapies for this condition, in part because the molecular mechanisms underlying APOL1's pathogenic contribution to FSGS are incompletely understood. Studying the transcriptomic landscape of APOL1 FSGS in patient kidneys is an important way to discover genes and molecular behaviors that are unique or most relevant to the human disease. With the hypothesis that the pathology driven by the high-risk APOL1 genotype is reflected in alteration of gene expression across the glomerular transcriptome, we compared expression and co-expression profiles of 15,703 genes in 16 Black patients with FSGS at high-risk vs 14 Black patients with a low-risk APOL1 genotype. Expression data from APOL1-inducible HEK293 cells and normal human glomeruli were used to pursue genes and molecular pathways uncovered in these studies. We discovered increased expression of APOL1 and nine other significant differentially expressed genes in high-risk patients. This included stanniocalcin, which has a role in mitochondrial and calcium-related processes along with differential correlations between high- and low-risk APOL1 and metabolism pathway genes. There were similar correlations with extracellular matrix- and immune-related genes, but significant loss of co-expression of mitochondrial genes in high-risk FSGS, and an NF-κB-down regulating gene, NKIRAS1, as the most significant hub gene with strong differential correlations with NDUF family (mitochondrial respiratory genes) and immune-related (JAK-STAT) genes. Thus, differences in mitochondrial gene regulation appear to underlie many differences observed between high- and low-risk Black patients with FSGS.
Collapse
Affiliation(s)
- Michelle T McNulty
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, Massachusetts, USA; Kidney Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA
| | - Damian Fermin
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Felix Eichinger
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Dongkeun Jang
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Noël P Burtt
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA; Metabolism Program, Broad Institute, Cambridge, Massachusetts, USA
| | - Martin R Pollak
- Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA; Metabolism Program, Broad Institute, Cambridge, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Division of Genetics, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Astrid Weins
- Harvard Medical School, Boston, Massachusetts, USA
| | - David J Friedman
- Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew G Sampson
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, Massachusetts, USA; Kidney Disease Initiative, Broad Institute, Cambridge, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
13
|
Shen M, Pan H, Ke J, Zhao F. NF-κB-upregulated miR-155-5p promotes hepatocyte mitochondrial dysfunction to accelerate the development of nonalcoholic fatty liver disease through downregulation of STC1. J Biochem Mol Toxicol 2022; 36:e23025. [PMID: 35603999 DOI: 10.1002/jbt.23025] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 01/07/2022] [Indexed: 12/30/2022]
Abstract
Previous studies have highlighted the involvement of nuclear factor kappa B (NF-κB) in the development of nonalcoholic fatty liver disease (NAFLD). The purpose of our investigation is to explore the interaction among NF-κB, microRNA-155-5p (miR-155-5p), and Stanniocalcin 1 (STC1), and its effects on NAFLD by establishing a NAFLD model in Sprague Dawley rats. A highly-expressed miR-155-5p and NF-κB was revealed in the liver tissues of NAFLD rats, and a positive correlation was identified between miR-155-5p and NF-κB. Next, the expression of NF-κB and STC1 was altered in the modeled rats through lentivirus injection, followed by determination on the levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), total cholesterol, triglycerides, and low-density lipoprotein cholesterol. Furthermore, the hepatocyte mitochondria were separated to measure the activities of adenosine triphosphate (ATP), reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and mitochondrial respiratory chain complex, and to observe the number, length and ultrastructural length of mitochondrial cristae. The results demonstrated that NF-κB overexpression induced mitochondrial dysfunction, increased ROS level, decreased ATP and MMP contents, as well as inhibited the number and length of mitochondrial cristae in the hepatocyte mitochondria of NAFLD rats. Besides, miR-155-5p was found to negatively regulate STC1 expression based on dual luciferase reporter gene assay, which exert inhibition on mitochondrial activity of hepatocytes in NAFLD rats. These results uncover the possible involvement of NF-κB/miR-155-5p/STC1 axis in NAFLD progression, that NF-κB could increase miR-155-5p expression to inhibit STC1 expression, thus inducing hepatic mitochondrial dysfunction and promoting the occurrence and development of NAFLD.
Collapse
Affiliation(s)
- Miao Shen
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Hanghai Pan
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Jinjing Ke
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Fei Zhao
- Health Management Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| |
Collapse
|
14
|
Molecular Mechanisms of Cardiac Amyloidosis. Int J Mol Sci 2021; 23:ijms23010025. [PMID: 35008444 PMCID: PMC8744761 DOI: 10.3390/ijms23010025] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/18/2021] [Accepted: 12/18/2021] [Indexed: 12/22/2022] Open
Abstract
Cardiac involvement has a profound effect on the prognosis of patients with systemic amyloidosis. Therapeutic methods for suppressing the production of causative proteins have been developed for ATTR amyloidosis and AL amyloidosis, which show cardiac involvement, and the prognosis has been improved. However, a method for removing deposited amyloid has not been established. Methods for reducing cytotoxicity caused by amyloid deposition and amyloid precursor protein to protect cardiovascular cells are also needed. In this review, we outline the molecular mechanisms and treatments of cardiac amyloidosis.
Collapse
|
15
|
Wang Y, Yu Z, Fan Z, Fang Y, He L, Peng M, Chen Y, Hu Z, Zhao K, Zhang H, Liu C. Cardiac developmental toxicity and transcriptome analyses of zebrafish (Danio rerio) embryos exposed to Mancozeb. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 226:112798. [PMID: 34592528 DOI: 10.1016/j.ecoenv.2021.112798] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023]
Abstract
Mancozeb (MZ), an antibacterial pesticide, has been linked to reproductive toxicity, neurotoxicity, and endocrine disruption. However, whether MZ has cardiactoxicity is unclear. In this study, the cardiotoxic effects of exposure to environment-related MZ concentrations ranging from 1.88 μM to 7.52 μM were evaluated at the larval stage of zebrafish. Transcriptome sequencing predicted the mechanism of MZ-induced cardiac developmental toxicity in zebrafish by enrichment analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO). Consistent with morphological changes, the osm, pfkfb3, foxh1, stc1, and nrarpb genes may effect normal development of zebrafish heart by activating NOTCH signaling pathways, resulting in pericardial edema, myocardial fibrosis, and congestion in the heart area. Moreover, differential gene expression analysis indicated that cyp-related genes (cyp1c2 and cyp3c3) were significantly upregulated after MZ treatment, which may be related to apoptosis of myocardial cells. These results were verified by real-time quantitative RT-qPCR and acridine orange staining. Our findings suggest that MZ-mediated cardiotoxic development of zebrafish larvae may be related to the activation of Notch and apoptosis-related signaling pathways.
Collapse
Affiliation(s)
- Yongfeng Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Zhiquan Yu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Zunpan Fan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Yiwei Fang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Liting He
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Meili Peng
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Yuanyao Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Zhiyong Hu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| |
Collapse
|
16
|
Morgan GJ, Buxbaum JN, Kelly JW. Light Chain Stabilization: A Therapeutic Approach to Ameliorate AL Amyloidosis. HEMATO 2021; 2:645-659. [PMID: 35757512 PMCID: PMC9218996 DOI: 10.3390/hemato2040042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Non-native immunoglobulin light chain conformations, including aggregates, appear to cause light chain amyloidosis pathology. Despite significant progress in pharmacological eradication of the neoplastic plasma cells that secrete these light chains, in many patients impaired organ function remains. The impairment is apparently due to a subset of resistant plasma cells that continue to secrete misfolding-prone light chains. These light chains are susceptible to the proteolytic cleavage that may enable light chain aggregation. We propose that small molecules that preferentially bind to the natively folded state of full-length light chains could act as pharmacological kinetic stabilizers, protecting light chains against unfolding, proteolysis and aggregation. Although the sequence of the pathological light chain is unique to each patient, fortunately light chains have highly conserved residues that form binding sites for small molecule kinetic stabilizers. We envision that such stabilizers could complement existing and emerging therapies to benefit light chain amyloidosis patients.
Collapse
Affiliation(s)
- Gareth J. Morgan
- Section of Hematology and Medical Oncology, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
- The Amyloidosis Center, Boston University School of Medicine, Boston, MA 02118, USA
- Correspondence:
| | - Joel N. Buxbaum
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jeffery W. Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
17
|
Rognoni P, Mazzini G, Caminito S, Palladini G, Lavatelli F. Dissecting the Molecular Features of Systemic Light Chain (AL) Amyloidosis: Contributions from Proteomics. ACTA ACUST UNITED AC 2021; 57:medicina57090916. [PMID: 34577839 PMCID: PMC8471912 DOI: 10.3390/medicina57090916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/23/2021] [Accepted: 08/29/2021] [Indexed: 02/08/2023]
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.
Collapse
Affiliation(s)
- Paola Rognoni
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, V.le Golgi 19, 27100 Pavia, Italy; (G.M.); (S.C.); (G.P.)
- Correspondence: (P.R.); (F.L.); Tel.: +39-0382502984 (P.R.); +39-0382502994 (F.L.)
| | - Giulia Mazzini
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, V.le Golgi 19, 27100 Pavia, Italy; (G.M.); (S.C.); (G.P.)
| | - Serena Caminito
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, V.le Golgi 19, 27100 Pavia, Italy; (G.M.); (S.C.); (G.P.)
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | - Giovanni Palladini
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, V.le Golgi 19, 27100 Pavia, Italy; (G.M.); (S.C.); (G.P.)
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | - Francesca Lavatelli
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, V.le Golgi 19, 27100 Pavia, Italy; (G.M.); (S.C.); (G.P.)
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
- Correspondence: (P.R.); (F.L.); Tel.: +39-0382502984 (P.R.); +39-0382502994 (F.L.)
| |
Collapse
|
18
|
Rius B, Mesgarzadeh JS, Romine IC, Paxman RJ, Kelly JW, Wiseman RL. Pharmacologic targeting of plasma cell endoplasmic reticulum proteostasis to reduce amyloidogenic light chain secretion. Blood Adv 2021; 5:1037-1049. [PMID: 33599742 PMCID: PMC7903236 DOI: 10.1182/bloodadvances.2020002813] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023] Open
Abstract
Light chain (LC) amyloidosis (AL) involves the toxic aggregation of amyloidogenic immunoglobulin LCs secreted from a clonal expansion of diseased plasma cells. Current AL treatments use chemotherapeutics to ablate the AL plasma cell population. However, no treatments are available that directly reduce the toxic LC aggregation involved in AL pathogenesis. An attractive strategy to reduce toxic LC aggregation in AL involves enhancing endoplasmic reticulum (ER) proteostasis in plasma cells to reduce the secretion and subsequent aggregation of amyloidogenic LCs. Here, we show that the ER proteostasis regulator compound 147 reduces secretion of an amyloidogenic LC as aggregation-prone monomers and dimers in AL patient-derived plasma cells. Compound 147 was established to promote ER proteostasis remodeling by activating the ATF6 unfolded protein response signaling pathway through a mechanism involving covalent modification of ER protein disulfide isomerases (PDIs). However, we show that 147-dependent reductions in amyloidogenic LCs are independent of ATF6 activation. Instead, 147 reduces amyloidogenic LC secretion through the selective, on-target covalent modification of ER proteostasis factors, including PDIs, revealing an alternative mechanism by which this compound can influence ER proteostasis of amyloidogenic proteins. Importantly, compound 147 does not interfere with AL plasma cell toxicity induced by bortezomib, a standard chemotherapeutic used to ablate the underlying diseased plasma cells in AL. This shows that pharmacologic targeting of ER proteostasis through selective covalent modification of ER proteostasis factors is a strategy that can be used in combination with chemotherapeutics to reduce the LC toxicity associated with AL pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Jeffery W Kelly
- Department of Chemistry, and
- Skaggs Institute for Chemical Biology, Scripps Research Institute, La Jolla, CA
| | | |
Collapse
|
19
|
Roginić S, Vinter O, Trbušić M, Roginić M, Ćatić Ćuti E. Cardiac Amyloidosis Detected on Imaging of Patients with Heart Failure. AMERICAN JOURNAL OF CASE REPORTS 2020; 21:e926290. [PMID: 33175723 PMCID: PMC7669957 DOI: 10.12659/ajcr.926290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Case series Patients: Male, 63-year-old • Female, 72-year-old • Female, 55-year-old Final Diagnosis: Amyloid light-chain amyloidosis • cardiac amyloidosis • cardiomyopathy • heart failure • primary AL amyloidosis Symptoms: Aphasia • dyspnea • heart failure • thrombosis • tongue mass Medication: — Clinical Procedure: Biopsy • chemotherapy • echocardiography Specialty: Cardiology • Hematology
Collapse
Affiliation(s)
- Siniša Roginić
- Department of Cardiology, General Hospital Zabok, Zabok, Croatia.,Department of Cardiology, Hospital of Croatian Veterans, Zabok, Croatia
| | - Ozren Vinter
- Department of Cardiology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Matias Trbušić
- Department of Cardiology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Martina Roginić
- Department of Cardiology, General Hospital Zabok, Zabok, Croatia.,Department of Cardiology, Hospital of Croatian Veterans, Zabok, Croatia
| | - Edina Ćatić Ćuti
- Department of Nephrology, General Hospital Zabok and Hospital of Croatian Veterans, Zabok, Croatia
| |
Collapse
|
20
|
Xu F, Yu Y, Wang F, Sun W, Li P, Wu HF, Bian ZP, Chen XJ, Dong-Jie X. Analysis of gene expression profiling of amyloidogenic immunoglobulin light-chains on cultured rat cardiomyocytes. Exp Ther Med 2020; 19:3767-3777. [PMID: 32346441 PMCID: PMC7185198 DOI: 10.3892/etm.2020.8610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 02/25/2020] [Indexed: 11/20/2022] Open
Abstract
The present study aimed to investigate the toxic effects of different amyloidogenic light-chains (LCs) on cardiomyocytes, and demonstrate the differentially expressed genes (DEGs) and signaling pathways that participate in this process. Cultured cardiomyocytes were treated with recombinant κ LC peptide (AL-09) or with serum from a patient diagnosed with multiple myeloma (λ LC) with cardiac involvement. The 6xHis peptide or serum from healthy patients was used as peptide control or serum control, respectively. Cell viability was determined using CCK-8 assay and apoptosis was analyzed by flow cytometry. The DEGs were detected by RNA sequencing (RNA-Seq), followed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Changes in gene expression levels were confirmed by reverse transcription-quantitative PCR. The cell viability in the AL-09 peptide-treated (0.2 mg/ml) and patient serum-treated (1:10 dilution) cardiomyocytes decreased to 42 and -72% of the corresponding control groups. The extent of cell apoptosis increased in AL-09-treated cardiomyocytes compared with the control group. RNA-Seq showed 256 DEGs co-existed in the two paired groups, including 127 upregulated and 88 downregulated genes. The KEGG pathways for upregulated expressed genes included the ‘TGF-β signaling pathway’, the ‘Hedgehog signaling pathway’, the ‘ErbB signaling pathway’ and ‘lysine degradation’. The higher mRNA expression of bone morphogenetic protein (Bmp) 4, Bmp6, prostaglandin G/H synthase (Ptgs)1, Ptgs2, epiregulin, Tgfa and procollagen-lysine,2-oxoglutarate 5-dioxygenase 2 were confirmed. The KEGG pathways of downregulated expressed genes included genes involved with the ‘p53 signaling pathway’ and the ‘cell cycle’. The mRNA expression levels of E3 ubiquitin-protein ligase CCNB1IP1 showed significant downregulation in the AL-09 peptide group compared with those in the 6xHis peptide group. In conclusion, cardiomyocytes treated with amyloidogenic λ and κ LCs presented with decreased cell viability compared with controls. Cell apoptosis increased in κ LC-treated cells compared with controls. The gene expression profiles associated with transforming growth factor-β-bone morphogenetic protein, the receptor tyrosine-protein kinase erbB-2 signaling pathways, prostaglandins, collagen production, the p53 signaling pathway and the cell cycle were altered in light-chain-treated cardiomyocytes.
Collapse
Affiliation(s)
- Fei Xu
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yue Yu
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Fang Wang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Sun
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Peng Li
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Heng-Fang Wu
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhi-Ping Bian
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiang-Jian Chen
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xu Dong-Jie
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
21
|
|
22
|
Zhang Y, Shan P, Srivastava A, Li Z, Lee PJ. Endothelial Stanniocalcin 1 Maintains Mitochondrial Bioenergetics and Prevents Oxidant-Induced Lung Injury via Toll-Like Receptor 4. Antioxid Redox Signal 2019; 30:1775-1796. [PMID: 30187766 PMCID: PMC6479262 DOI: 10.1089/ars.2018.7514] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AIMS Oxidant-induced endothelial injury plays a critical role in the pathogenesis of acute lung injury (ALI) and subsequent respiratory failure. Our previous studies revealed an endogenous antioxidant and protective pathway in lung endothelium mediated by heat shock protein 70 (Hsp70)-toll-like receptor 4 (TLR4) signaling. However, the downstream effector mechanisms remained unclear. Stanniocalcin 1 (STC1) has been reported to mediate antioxidant responses in tissues such as the lungs. However, regulators of STC1 expression as well as its physiological function in the lungs were unknown. We sought to elucidate the relationship between TLR4 and STC1 in hyperoxia-induced lung injury in vitro and in vivo and to define the functional role of STC1 expression in lung endothelium. RESULTS We identified significantly decreased STC1 expression in TLR4 knockout mouse lungs and primary lung endothelium isolated from TLR4 knockout mice. Overexpression of STC1 was associated with endothelial cytoprotection, whereas decreased or insufficient expression was associated with increased oxidant-induced injury and death. An Hsp70-TLR4-nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signal mediates STC1 induction in the lungs and endothelial cells. We also demonstrated a previously unrecognized role for mitochondrial-associated STC1, via TLR4, in maintaining normal glycolysis, mitochondrial bioenergetics, and mitochondrial calcium levels. INNOVATION To date, a physiological role for STC1 in oxidant-induced ALI has not been identified. In addition, our studies show that STC1 is regulated by TLR4 and exerts lung and endothelial protection in response to sterile oxidant-induced lung injury. CONCLUSIONS Our studies reveal a novel TLR4-STC1-mediated mitochondrial pathway that has homeostatic as well as oxidant-induced cytoprotective functions in lung endothelium.
Collapse
Affiliation(s)
- Yi Zhang
- 1 Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Peiying Shan
- 1 Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Anup Srivastava
- 1 Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut.,2 Division of Endocrinology, Department of Medicine, College of Medicine, University of Arizona, Tucson, Arizona
| | - Zhenyu Li
- 1 Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut.,3 Intensive Care Unit, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Patty J Lee
- 1 Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
23
|
Mishra S, Joshi S, Ward JE, Buys EP, Mishra D, Mishra D, Morgado I, Fisch S, Lavatelli F, Merlini G, Dorbala S, MacRae CA, Liao R. Zebrafish model of amyloid light chain cardiotoxicity: regeneration versus degeneration. Am J Physiol Heart Circ Physiol 2019; 316:H1158-H1166. [PMID: 30875258 PMCID: PMC6580397 DOI: 10.1152/ajpheart.00788.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/27/2022]
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.
Collapse
Affiliation(s)
- Shikha Mishra
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Shaurya Joshi
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Jennifer E Ward
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Eva P Buys
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Deepak Mishra
- Department of Biological Engineering, Massachusetts Institute of Technology , Boston, Massachusetts
| | - Deepa Mishra
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Isabel Morgado
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Sudeshna Fisch
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Francesca Lavatelli
- Amyloidosis Research and Treatment Center, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, and Department of Molecular Medicine, University of Pavia , Pavia , Italy
| | - Giampaolo Merlini
- Amyloidosis Research and Treatment Center, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, and Department of Molecular Medicine, University of Pavia , Pavia , Italy
| | - Sharmila Dorbala
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Calum A MacRae
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Ronglih Liao
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
24
|
Richey T, Foster JS, Williams AD, Williams AB, Stroh A, Macy S, Wooliver C, Heidel RE, Varanasi SK, Ergen EN, Trent DJ, Kania SA, Kennel SJ, Martin EB, Wall JS. Macrophage-Mediated Phagocytosis and Dissolution of Amyloid-Like Fibrils in Mice, Monitored by Optical Imaging. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:989-998. [PMID: 30735627 DOI: 10.1016/j.ajpath.2019.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/27/2023]
Abstract
Light chain-associated amyloidosis is characterized by the extracellular deposition of amyloid fibrils in abdominothoracic organs, skin, soft tissue, and peripheral nerves. Phagocytic cells of the innate immune system appear to be ineffective at clearing the material; however, human light chain amyloid extract, injected subcutaneously into mice, is rapidly cleared in a process that requires neutrophil activity. To better elucidate the phagocytosis of light chain fibrils, a potential method of cell-mediated dissolution, amyloid-like fibrils were labeled with the pH-sensitive dye pHrodo red and a near infrared fluorophore. After injecting this material subcutaneously in mice, optical imaging was used to quantitatively monitor phagocytosis and dissolution of fibrils concurrently. Histologic evaluation of the residual fibril masses revealed the presence of CD68+, F4/80+, ionized calcium binding adaptor molecule 1- macrophages containing Congo red-stained fibrils as well as neutrophil-associated proteins with no evidence of intact neutrophils. These data suggest an early infiltration of neutrophils, followed by extensive phagocytosis of the light chain fibrils by macrophages, leading to dissolution of the mass. Optical imaging of this novel murine model, coupled with histologic evaluation, can be used to study the cellular mechanisms underlying dissolution of synthetic amyloid-like fibrils and human amyloid extracts. In addition, it may serve as a test bed to evaluate investigational opsonizing agents that might serve as therapeutic agents for light chain-associated amyloidosis.
Collapse
Affiliation(s)
- Tina Richey
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - James S Foster
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Angela D Williams
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | | | - Alexa Stroh
- Department of Biochemistry, Cellular and Molecular Biology, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Sallie Macy
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Craig Wooliver
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - R Eric Heidel
- Department of Surgery, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Siva K Varanasi
- Department of Biochemistry, Cellular and Molecular Biology, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Elizabeth N Ergen
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Dianne J Trent
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Stephen A Kania
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Stephen J Kennel
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Emily B Martin
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Jonathan S Wall
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee.
| |
Collapse
|
25
|
Development and validation of a survival staging system incorporating BNP in patients with light chain amyloidosis. Blood 2018; 133:215-223. [PMID: 30333122 DOI: 10.1182/blood-2018-06-858951] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/12/2018] [Indexed: 01/08/2023] Open
Abstract
Immunoglobulin light chain amyloidosis (AL amyloidosis) is caused by misfolded light chains that form soluble toxic aggregates that deposit in tissues and organs, leading to organ dysfunction. The leading determinant of survival is cardiac involvement. Current staging systems use N-terminal pro-brain natriuretic peptide (NT-proBNP) and cardiac troponins T and I (TnT and TnI) for prognostication, but many centers do not offer NT-proBNP. We sought to derive a new staging system using brain natriuretic peptide (BNP) that would correlate with the Mayo 2004 staging system and be predictive for survival in AL amyloidosis. Two cohorts of patients were created: a derivation cohort of 249 consecutive patients who had BNP, NT-proBNP, and TnI drawn simultaneously to create the staging system and a complementary cohort of 592 patients with 10 years of follow-up to determine survival. In the derivation cohort, we found that a BNP threshold of more than 81 pg/mL best associated with Mayo 2004 stage and also best identified cardiac involvement. Three stages were developed based on a BNP higher than 81 pg/mL and a TnI higher than 0.1 ng/mL and compared with Mayo 2004 with high concordance (κ = 0.854). In the complementary cohort, 25% of patients had stage I, 44% had stage II, 15% had stage III, and 16% had stage IIIb disease with a median survival not reached in stage I, 9.4 years in stage II, 4.3 years in stage III, and 1 year in stage IIIb. This new Boston University biomarker scoring system will allow centers without access to NT-proBNP the ability to appropriately stage patients with AL amyloidosis. This trial was registered at www.clinicaltrials.gov as #NCT00898235.
Collapse
|
26
|
Blancas-Mejia LM, Misra P, Dick CJ, Cooper SA, Redhage KR, Bergman MR, Jordan TL, Maar K, Ramirez-Alvarado M. Immunoglobulin light chain amyloid aggregation. Chem Commun (Camb) 2018; 54:10664-10674. [PMID: 30087961 DOI: 10.1039/c8cc04396e] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Light chain (AL) amyloidosis is a devastating, complex, and incurable protein misfolding disease. It is characterized by an abnormal proliferation of plasma cells (fully differentiated B cells) producing an excess of monoclonal immunoglobulin light chains that are secreted into circulation, where the light chains misfold, aggregate as amyloid fibrils in target organs, and cause organ dysfunction, organ failure, and death. In this article, we will review the factors that contribute to AL amyloidosis complexity, the findings by our laboratory from the last 16 years and the work from other laboratories on understanding the structural, kinetics, and thermodynamic contributions that drive immunoglobulin light chain-associated amyloidosis. We will discuss the role of cofactors and the mechanism of cellular damage. Last, we will review our recent findings on the high resolution structure of AL amyloid fibrils. AL amyloidosis is the best example of protein sequence diversity in misfolding diseases, as each patient has a unique combination of germline donor sequences and multiple amino acid mutations in the protein that forms the amyloid fibril.
Collapse
Affiliation(s)
- Luis M Blancas-Mejia
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lee WH, Tsai MJ, Chang WA, Wu LY, Wang HY, Chang KF, Su HM, Kuo PL. Deduction of novel genes potentially involved in hypoxic AC16 human cardiomyocytes using next-generation sequencing and bioinformatics approaches. Int J Mol Med 2018; 42:2489-2502. [PMID: 30226555 PMCID: PMC6192719 DOI: 10.3892/ijmm.2018.3851] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 08/28/2018] [Indexed: 12/21/2022] Open
Abstract
Atherosclerotic cardiovascular disease and acute myocardial infarction are the leading causes of mortality worldwide, and apoptosis is the major pathway of cardiomyocyte death under hypoxic conditions. Although studies have reported changes in the expression of certain pro‑apoptotic and anti‑apoptotic genes in hypoxic cardiomyocytes, genetic regulations are complex in human cardiomyocytes and there is much that remains to be fully elucidated. The present study aimed to identify differentially expressed genes in hypoxic human AC16 cardiomyocytes using next‑generation sequencing and bioinformatics. A total of 24 genes (15 upregulated and 9 downregulated) with potential micro (mi)RNA‑mRNA interactions were identified in the miRmap database. Utilising the Gene Expression Omnibus database of cardiac microvascular endothelial cells, tensin 1, B‑cell lymphoma 2‑interacting protein 3 like, and stanniocalcin 1 were found to be upregulated, and transferrin receptor and methyltransferase like 7A were found to be downregulated in response to hypoxia. Considering the results from miRmap, TargetScan and miRDB together, two potential miRNA‑mRNA interactions were identified: hsa‑miRNA (miR)‑129‑5p/CDC42EP3 and hsa‑miR‑330‑3p/HELZ. These findings contribute important insights into possible novel diagnostic or therapeutic strategies for targeting cardiomyocytes under acute hypoxic stress in conditions, including acute myocardial infarction. The results of the present study also introduce an important novel approach in investigating acute hypoxic pathophysiology.
Collapse
Affiliation(s)
- Wen-Hsien Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ming-Ju Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ling-Yu Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Han-Ying Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | | | - Ho-Ming Su
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
28
|
Pagourelias ED, Mirea O, Vovas G, Duchenne J, Michalski B, Van Cleemput J, Bogaert J, Vassilikos VP, Voigt JU. Relation of regional myocardial structure and function in hypertrophic cardiomyopathy and amyloidois: a combined two-dimensional speckle tracking and cardiovascular magnetic resonance analysis. Eur Heart J Cardiovasc Imaging 2018; 20:426-437. [DOI: 10.1093/ehjci/jey107] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/12/2018] [Accepted: 07/10/2018] [Indexed: 11/13/2022] Open
Affiliation(s)
- Efstathios D Pagourelias
- Department of Cardiovascular Diseases, University Hospital Leuven, Catholic University Leuven, Herestraat 49, Leuven, Belgium
- Third Cardiology Department, Hippokrateion University Hospital, Aristotle University of Thessaloniki, 49 Konstantinoupoleos Str, Thessaloniki, Greece
| | - Oana Mirea
- Department of Cardiovascular Diseases, University Hospital Leuven, Catholic University Leuven, Herestraat 49, Leuven, Belgium
- Department of Cardiology, University of Medicine and Pharmacy of Craiova, University County Hospital of Craiova, 1 Tabaci Str, Craiova, Romania
| | - Georgios Vovas
- Department of Radiology, University Hospital Leuven, Catholic University Leuven, Herestraat 49, Leuven, Belgium
| | - Jürgen Duchenne
- Department of Cardiovascular Diseases, University Hospital Leuven, Catholic University Leuven, Herestraat 49, Leuven, Belgium
| | - Blazej Michalski
- Department of Cardiovascular Diseases, University Hospital Leuven, Catholic University Leuven, Herestraat 49, Leuven, Belgium
| | - Johan Van Cleemput
- Department of Cardiovascular Diseases, University Hospital Leuven, Catholic University Leuven, Herestraat 49, Leuven, Belgium
| | - Jan Bogaert
- Department of Radiology, University Hospital Leuven, Catholic University Leuven, Herestraat 49, Leuven, Belgium
| | - Vasilios P Vassilikos
- Third Cardiology Department, Hippokrateion University Hospital, Aristotle University of Thessaloniki, 49 Konstantinoupoleos Str, Thessaloniki, Greece
| | - Jens-Uwe Voigt
- Department of Cardiovascular Diseases, University Hospital Leuven, Catholic University Leuven, Herestraat 49, Leuven, Belgium
| |
Collapse
|
29
|
Growth differentiation factor-15 is a new biomarker for survival and renal outcomes in light chain amyloidosis. Blood 2018; 131:1568-1575. [DOI: 10.1182/blood-2017-12-819904] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 01/18/2018] [Indexed: 11/20/2022] Open
Abstract
Key Points
GDF-15 level is a new prognostic factor for survival in AL amyloidosis, and its reduction after therapy is associated with better outcome. GDF-15 level is probably the strongest predictor for renal outcomes in patients with AL amyloidosis.
Collapse
|
30
|
Abstract
"Cardiac amyloidosis" is the term commonly used to reflect the deposition of abnormal protein amyloid in the heart. This process can result from several different forms, most commonly from light-chain (AL) amyloidosis and transthyretin (ATTR) amyloidosis, which in turn can represent wild-type (ATTRwt) or genetic form. Regardless of the origin, cardiac involvement is usually associated with poor prognosis, especially in AL amyloidosis. Although several treatment options, including chemotherapy, exist for different forms of the disease, cardiac transplantation is increasingly considered. However, high mortality on the transplantation list, typical for patients with amyloidosis, and suboptimal post-transplant outcomes are major issues. We are reviewing the literature and summarizing pros and cons of listing patients with amyloidosis for cardiac or combine organ transplant, appropriate work-up, and intermediate and long-term outcomes. Both AL and ATTR amyloidosis are included in this review.
Collapse
|
31
|
Abstract
The heart, like any organ in the body, is susceptible to amyloid deposition. Although more than 30 types of protein can cause amyloidosis, only two types commonly deposit in the ventricular myocardium: amyloid light chain and amyloid transthyretin. Amyloid cardiomyopathy is usually a major determinant of patient outcomes, and the diagnosis of heart involvement can be often relatively under-diagnosed, owing to nonspecific presenting symptoms and signs at a subclinical stage. The diagnosis of cardiac amyloidosis is usually performed by endomyocardial biopsy; however, the invasive nature and related high-risk complications restrict its wide use in clinical settings. Recently, with the advent of innovative techniques used for evaluating cardiac amyloidosis, noninvasive methods become increasingly important, especially in earlier diagnosis, distinguishing typing, risk prediction and response to treatment. Here, we will review recent developments in the noninvasive methods used in the assessment of cardiac amyloidosis, focused on the laboratory biomarkers and imaging modalities.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Dongcheng district, Beijing, 100730, China
| | - Quan Fang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Wangfujing, Dongcheng district, Beijing, 100730, China.
| |
Collapse
|
32
|
Imperlini E, Gnecchi M, Rognoni P, Sabidò E, Ciuffreda MC, Palladini G, Espadas G, Mancuso FM, Bozzola M, Malpasso G, Valentini V, Palladini G, Orrù S, Ferraro G, Milani P, Perlini S, Salvatore F, Merlini G, Lavatelli F. Proteotoxicity in cardiac amyloidosis: amyloidogenic light chains affect the levels of intracellular proteins in human heart cells. Sci Rep 2017; 7:15661. [PMID: 29142197 PMCID: PMC5688098 DOI: 10.1038/s41598-017-15424-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 10/27/2017] [Indexed: 12/31/2022] Open
Abstract
AL amyloidosis is characterized by widespread deposition of immunoglobulin light chains (LCs) as amyloid fibrils. Cardiac involvement is frequent and leads to life-threatening cardiomyopathy. Besides the tissue alteration caused by fibrils, clinical and experimental evidence indicates that cardiac damage is also caused by proteotoxicity of prefibrillar amyloidogenic species. As in other amyloidoses, the damage mechanisms at cellular level are complex and largely undefined. We have characterized the molecular changes in primary human cardiac fibroblasts (hCFs) exposed in vitro to soluble amyloidogenic cardiotoxic LCs from AL cardiomyopathy patients. To evaluate proteome alterations caused by a representative cardiotropic LC, we combined gel-based with label-free shotgun analysis and performed bioinformatics and data validation studies. To assess the generalizability of our results we explored the effects of multiple LCs on hCF viability and on levels of a subset of cellular proteins. Our results indicate that exposure of hCFs to cardiotropic LCs translates into proteome remodeling, associated with apoptosis activation and oxidative stress. The proteome alterations affect proteins involved in cytoskeletal organization, protein synthesis and quality control, mitochondrial activity and metabolism, signal transduction and molecular trafficking. These results support and expand the concept that soluble amyloidogenic cardiotropic LCs exert toxic effects on cardiac cells.
Collapse
Affiliation(s)
- Esther Imperlini
- IRCCS SDN, Naples, Italy.,CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Massimiliano Gnecchi
- Coronary Care Unit and Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy.,Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Paola Rognoni
- Amyloidosis Research and Treatment Center, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Eduard Sabidò
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Maria Chiara Ciuffreda
- Coronary Care Unit and Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giovanni Palladini
- Amyloidosis Research and Treatment Center, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Guadalupe Espadas
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Francesco Mattia Mancuso
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Margherita Bozzola
- Amyloidosis Research and Treatment Center, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Giuseppe Malpasso
- Coronary Care Unit and Laboratory of Experimental Cardiology for Cell and Molecular Therapy, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Veronica Valentini
- Amyloidosis Research and Treatment Center, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Giuseppina Palladini
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Stefania Orrù
- IRCCS SDN, Naples, Italy.,CEINGE-Biotecnologie Avanzate, Naples, Italy.,Department of Movement Sciences, "Parthenope" University, Naples, Italy
| | - Giovanni Ferraro
- Amyloidosis Research and Treatment Center, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Paolo Milani
- Amyloidosis Research and Treatment Center, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Stefano Perlini
- Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate, Naples, Italy. .,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Pavia, Italy.
| | - Giampaolo Merlini
- Amyloidosis Research and Treatment Center, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy.
| | - Francesca Lavatelli
- Amyloidosis Research and Treatment Center, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| |
Collapse
|
33
|
Sapp V, Jain M, Liao R. Viewing Extrinsic Proteotoxic Stress Through the Lens of Amyloid Cardiomyopathy. Physiology (Bethesda) 2017; 31:294-9. [PMID: 27252164 DOI: 10.1152/physiol.00047.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Proteotoxicity refers to toxic stress caused by misfolded proteins of extrinsic or intrinsic origin and plays an integral role in the pathogenesis of cardiovascular diseases. Herein, we provide an overview of the current understanding of mechanisms underlying proteotoxicity and its contribution in the pathogenesis of amyloid cardiomyopathy.
Collapse
Affiliation(s)
- Valerie Sapp
- Departments of Medicine & Pharmacology, University of California San Diego, San Diego, California; and
| | - Mohit Jain
- Departments of Medicine & Pharmacology, University of California San Diego, San Diego, California; and
| | - Ronglih Liao
- Division of Genetics and Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
34
|
Zhang C, Huang X, Li J. Light chain amyloidosis: Where are the light chains from and how they play their pathogenic role? Blood Rev 2017; 31:261-270. [PMID: 28336182 DOI: 10.1016/j.blre.2017.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 03/03/2017] [Indexed: 12/17/2022]
Abstract
Amyloid light-chain (AL) amyloidosis is a plasma-cell dyscrasia, as well as the most common type of systematic amyloidosis. Pathogenic plasma cells that have distinct cytogenetic and molecular properties secrete an excess amount of amyloidogenic light chains. Assisted by post-translational modifications, matrix components, and other environmental factors, these light chains undergo a conformational change that triggers the formation of amyloid fibrils that overrides the extracellular protein quality control system. Moreover, the amyloidogenic light-chain itself is cytotoxic. As a consequence, organ dysfunction is caused by both organ architecture disruption and the direct cytotoxic effect of amyloidogenic light chains. Here, we reviewed the molecular mechanisms underlying this sequence of events that ultimately leads to AL amyloidosis and also discuss current in vitro and in vivo models, as well as relevant novel therapeutic approaches.
Collapse
Affiliation(s)
- Chunlan Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xufei Huang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jian Li
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
35
|
Perlini S, Mussinelli R, Salinaro F. New and Evolving Concepts Regarding the Prognosis and Treatment of Cardiac Amyloidosis. Curr Heart Fail Rep 2016; 13:267-272. [PMID: 27900617 DOI: 10.1007/s11897-016-0311-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Systemic amyloidoses are rare and proteiform diseases, caused by extracellular accumulation of insoluble misfolded fibrillar proteins. Prognosis is dictated by cardiac involvement, which is especially frequent in light chain (AL) and in transthyretin variants (ATTR, both mutated, (ATTRm), and wild-type, (ATTRwt)). Recently, ATTRwt has emerged as a potentially relevant cause of a heart failure with preserved ejection fraction (HFpEF). Cardiac amyloidosis is an archetypal example of restrictive cardiomyopathy, with signs and symptoms of global heart failure and diastolic dysfunction. Independent of the aetiology, cardiac amyloidosis is associated with left ventricular concentric "hypertrophy" (i.e. increased wall thickness), preserved (or mildly depressed) ejection fraction, reduced midwall fractional shortening and global longitudinal function, as well as evident diastolic dysfunction, up to an overly restrictive pattern of the left ventricular filling. Cardiac biomarkers such as troponins and natriuretic peptides are very robust and widely accepted diagnostic as well as prognostic tools. Owing to its dismal prognosis, accurate and early diagnosis is mandatory and potentially life-saving. Although pathogenesis is still not completely understood, direct cardiomyocyte toxicity of the amyloidogenic precursor proteins and/or oligomer aggregates adds on tissue architecture disruption caused by amyloid deposition. The clarification of mechanisms of cardiac damage is offering new potential therapeutic targets, and several treatment options with a relevant impact on prognosis are now available.
Collapse
Affiliation(s)
- Stefano Perlini
- Clinica Medica II, Department of Internal Medicine, University of Pavia, P. le Golgi 19, 27100, Pavia, Italy.
- Amyloidosis Research and Treatment Center, Fondazione Policlinico IRCCS San Matteo, University of Pavia, P. le Golgi 19, 27100, Pavia, Italy.
| | - Roberta Mussinelli
- Clinica Medica II, Department of Internal Medicine, University of Pavia, P. le Golgi 19, 27100, Pavia, Italy
| | - Francesco Salinaro
- Clinica Medica II, Department of Internal Medicine, University of Pavia, P. le Golgi 19, 27100, Pavia, Italy
- Experimental Medicine PhD Program, University of Pavia, Pavia, Italy
| |
Collapse
|
36
|
Meshitsuka S, Shingaki S, Hotta M, Goto M, Kobayashi M, Ukawa Y, Sagesaka YM, Wada Y, Nojima M, Suzuki K. Phase 2 trial of daily, oral epigallocatechin gallate in patients with light-chain amyloidosis. Int J Hematol 2016; 105:295-308. [PMID: 27815860 DOI: 10.1007/s12185-016-2112-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/12/2016] [Accepted: 10/18/2016] [Indexed: 12/26/2022]
Abstract
Previous studies have suggested that an increase in mitochondrial reactive oxygen species may cause organ damage in patients with light-chain (AL) amyloidosis; however, this damage can be decreased by antioxidant-agent treatment. Epigallocatechin gallate (EGCG), the major natural catechin in green tea, has potent antioxidant activity. Because EGCG has recently been reported to have a favorable toxicity profile for treating amyloidosis, we sought to examine the clinical efficacy and toxicity of EGCG in patients with AL amyloidosis. Fifty-seven patients were randomly assigned to the EGCG and observation groups and observed for six months. There were no increases in grade 3-5 adverse events and EGCG therapy was well tolerated. Although a decrease in the urinary albumin level was found in the EGCG group in patients with obvious albuminuria after treatment initiation, its antioxidant activity may not be sufficient to clarify the potential effect of EGCG in patients with AL amyloidosis. Because some of the biological markers responsible for organ damage were well correlated to the level of antioxidant potential in patients' plasma, the status of oxidative stress in the blood may indicate the extent of organ damage in clinical situations.
Collapse
Affiliation(s)
- Sohsuke Meshitsuka
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan. .,Center for Translational Research, The Institute of Medical Science Hospital, The University of Tokyo, 4-6-1, Shiroganedai, Minato, Tokyo, Japan.
| | - Sumito Shingaki
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Masatoshi Hotta
- Department of Radiology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Miku Goto
- Laboratory for Clinical Nutrition, Department of Food and Nutrition, Faculty of Human Life, Jumonji University, Niiza, Japan
| | | | - Yuuichi Ukawa
- Central Research Institute, ITO EN, Ltd, Makinohara, Japan
| | | | - Yasuyo Wada
- Laboratory for Clinical Nutrition, Department of Food and Nutrition, Faculty of Human Life, Jumonji University, Niiza, Japan
| | - Masanori Nojima
- Center for Translational Research, The Institute of Medical Science Hospital, The University of Tokyo, 4-6-1, Shiroganedai, Minato, Tokyo, Japan
| | - Kenshi Suzuki
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| |
Collapse
|
37
|
Renz M, Torres R, Dolan PJ, Tam SJ, Tapia JR, Li L, Salmans JR, Barbour RM, Shughrue PJ, Nijjar T, Schenk D, Kinney GG, Zago W. 2A4 binds soluble and insoluble light chain aggregates from AL amyloidosis patients and promotes clearance of amyloid deposits by phagocytosis †. Amyloid 2016; 23:168-177. [PMID: 27494229 PMCID: PMC5152553 DOI: 10.1080/13506129.2016.1205974] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Amyloid light chain (AL) amyloidosis is characterized by misfolded light chain (LC) (amyloid) deposition in various peripheral organs, leading to progressive dysfunction and death. There are no regulatory agency-approved treatments for AL amyloidosis, and none of the available standard of care approaches directly targets the LC protein that constitutes the amyloid. NEOD001, currently in late-stage clinical trials, is a conformation-specific, anti-LC antibody designed to specifically target misfolded LC aggregates and promote phagocytic clearance of AL amyloid deposits. The present study demonstrated that the monoclonal antibody 2A4, the murine form of NEOD001, binds to patient-derived soluble and insoluble LC aggregates and induces phagocytic clearance of AL amyloid in vitro. 2A4 specifically labeled all 21 fresh-frozen organ samples studied, which were derived from 10 patients representing both κ and λ LC amyloidosis subtypes. 2A4 immunoreactivity largely overlapped with thioflavin T-positive labeling, and 2A4 bound both soluble and insoluble LC aggregates extracted from patient tissue. Finally, 2A4 induced macrophage engagement and phagocytic clearance of AL amyloid deposits in vitro. These findings provide further evidence that 2A4/NEOD001 can effectively clear and remove human AL-amyloid from tissue and further support the rationale for the evaluation of NEOD001 in patients with AL amyloidosis.
Collapse
Affiliation(s)
- Mark Renz
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| | - Ronald Torres
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| | - Philip J Dolan
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| | - Stephen J Tam
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| | - Jose R Tapia
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| | - Lauri Li
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| | | | | | | | | | - Dale Schenk
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| | - Gene G Kinney
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| | - Wagner Zago
- a Prothena Biosciences Inc , South San Francisco , CA , USA
| |
Collapse
|
38
|
Rationale, application and clinical qualification for NT-proBNP as a surrogate end point in pivotal clinical trials in patients with AL amyloidosis. Leukemia 2016; 30:1979-1986. [PMID: 27416985 PMCID: PMC5056962 DOI: 10.1038/leu.2016.191] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/17/2016] [Accepted: 07/04/2016] [Indexed: 12/16/2022]
Abstract
Amyloid light-chain (LC) amyloidosis (AL amyloidosis) is a rare and fatal disease for which there are no approved therapies. In patients with AL amyloidosis, LC aggregates progressively accumulate in organs, resulting in organ failure that is particularly lethal when the heart is involved. A significant obstacle in the development of treatments for patients with AL amyloidosis, as well as for those with any disease that is rare, severe and heterogeneous, has been satisfying traditional clinical trial end points (for example, overall survival or progression-free survival). It is for this reason that many organizations, including the United States Food and Drug Administration through its Safety and Innovation Act Accelerated Approval pathway, have recognized the need for biomarkers as surrogate end points. The international AL amyloidosis expert community is in agreement that the N-terminal fragment of the pro-brain natriuretic peptide (NT-proBNP) is analytically validated and clinically qualified as a biomarker for use as a surrogate end point for survival in patients with AL amyloidosis. Underlying this consensus is the demonstration that NT-proBNP is an indicator of cardiac response in all interventional studies in which it has been assessed, despite differences in patient population, treatment type and treatment schedule. Furthermore, NT-proBNP expression is directly modulated by amyloidogenic LC-elicited signal transduction pathways in cardiomyocytes. The use of NT-proBNP will greatly facilitate the development of targeted therapies for AL amyloidosis. Here, we review the data supporting the use of NT-proBNP, a biomarker that is analytically validated, clinically qualified, directly modulated by LC and universally accepted by AL amyloidosis specialists, as a surrogate end point for survival.
Collapse
|
39
|
What is new in diagnosis and management of light chain amyloidosis? Blood 2016; 128:159-68. [PMID: 27053535 DOI: 10.1182/blood-2016-01-629790] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 04/05/2016] [Indexed: 12/21/2022] Open
Abstract
Light chain (AL) amyloidosis is caused by a usually small plasma cell clone producing a misfolded light chain that deposits in tissues. Survival is mostly determined by the severity of heart involvement. Recent studies are clarifying the mechanisms of cardiac damage, pointing to a toxic effect of amyloidogenic light chains and offering new potential therapeutic targets. The diagnosis requires adequate technology, available at referral centers, for amyloid typing. Late diagnosis results in approximately 30% of patients presenting with advanced, irreversible organ involvement and dying in a few months despite modern treatments. The availability of accurate biomarkers of clonal and organ disease is reshaping the approach to patients with AL amyloidosis. Screening of early organ damage based on biomarkers can help identify patients with monoclonal gammopathy of undetermined significance who are developing AL amyloidosis before they become symptomatic. Staging systems and response assessment based on biomarkers facilitate the design and conduction of clinical trials, guide the therapeutic strategy, and allow the timely identification of refractory patients to be switched to rescue therapy. Treatment should be risk-adapted. Recent studies are linking specific characteristics of the plasma cell clone to response to different types of treatment, moving toward patient-tailored therapy. In addition, novel anti-amyloid treatments are being developed that might be combined with anti-plasma cell chemotherapy.
Collapse
|
40
|
Abstract
Immunoglobulin light chain amyloidosis (AL) is a rare, complex disease caused by misfolded free light chains produced by a usually small, indolent plasma cell clone. Effective treatments exist that can alter the natural history, provided that they are started before irreversible organ damage has occurred. The cornerstones of the management of AL amyloidosis are early diagnosis, accurate typing, appropriate risk-adapted therapy, tight follow-up, and effective supportive treatment. The suppression of the amyloidogenic light chains using the cardiac biomarkers as guide to choose chemotherapy is still the mainstay of therapy. There are exciting possibilities ahead, including the study of oral proteasome inhibitors, antibodies directed at plasma cell clone, and finally antibodies attacking the amyloid deposits are entering the clinic, offering unprecedented opportunities for radically improving the care of this disease.
Collapse
Affiliation(s)
- Angela Dispenzieri
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Division of Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - Giampaolo Merlini
- Amyloidosis Research and Treatment Center, Foundation IRCCS Policlinico San Matteo and Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
41
|
McWilliams-Koeppen HP, Foster JS, Hackenbrack N, Ramirez-Alvarado M, Donohoe D, Williams A, Macy S, Wooliver C, Wortham D, Morrell-Falvey J, Foster CM, Kennel SJ, Wall JS. Light Chain Amyloid Fibrils Cause Metabolic Dysfunction in Human Cardiomyocytes. PLoS One 2015; 10:e0137716. [PMID: 26393799 PMCID: PMC4579077 DOI: 10.1371/journal.pone.0137716] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 08/20/2015] [Indexed: 12/23/2022] Open
Abstract
Light chain (AL) amyloidosis is the most common form of systemic amyloid disease, and cardiomyopathy is a dire consequence, resulting in an extremely poor prognosis. AL is characterized by the production of monoclonal free light chains that deposit as amyloid fibrils principally in the heart, liver, and kidneys causing organ dysfunction. We have studied the effects of amyloid fibrils, produced from recombinant λ6 light chain variable domains, on metabolic activity of human cardiomyocytes. The data indicate that fibrils at 0.1 μM, but not monomer, significantly decrease the enzymatic activity of cellular NAD(P)H-dependent oxidoreductase, without causing significant cell death. The presence of amyloid fibrils did not affect ATP levels; however, oxygen consumption was increased and reactive oxygen species were detected. Confocal fluorescence microscopy showed that fibrils bound to and remained at the cell surface with little fibril internalization. These data indicate that AL amyloid fibrils severely impair cardiomyocyte metabolism in a dose dependent manner. These data suggest that effective therapeutic intervention for these patients should include methods for removing potentially toxic amyloid fibrils.
Collapse
Affiliation(s)
- Helen P. McWilliams-Koeppen
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
| | - James S. Foster
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
| | - Nicole Hackenbrack
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
| | - Marina Ramirez-Alvarado
- Department of Biochemistry/Mol. Biol. and Immunology, Mayo Clinic, Rochester, MN, United States of America
| | - Dallas Donohoe
- Department of Nutrition, University of Tennessee Knoxville, TN, United States of America
| | - Angela Williams
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
| | - Sallie Macy
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
| | - Craig Wooliver
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
| | - Dale Wortham
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
| | - Jennifer Morrell-Falvey
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States of America
| | - Carmen M. Foster
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States of America
| | - Stephen J. Kennel
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
- Department of Radiology, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
| | - Jonathan S. Wall
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
- Department of Radiology, University of Tennessee Graduate School of Medicine, Knoxville, TN United States of America
- * E-mail:
| |
Collapse
|
42
|
Lavatelli F, Imperlini E, Orrù S, Rognoni P, Sarnataro D, Palladini G, Malpasso G, Soriano ME, Di Fonzo A, Valentini V, Gnecchi M, Perlini S, Salvatore F, Merlini G. Novel mitochondrial protein interactors of immunoglobulin light chains causing heart amyloidosis. FASEB J 2015. [PMID: 26220173 DOI: 10.1096/fj.15-272179] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
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.
Collapse
Affiliation(s)
- Francesca Lavatelli
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Esther Imperlini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Stefania Orrù
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Paola Rognoni
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Daniela Sarnataro
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Giuseppina Palladini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Giuseppe Malpasso
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Maria Eugenia Soriano
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Andrea Di Fonzo
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Veronica Valentini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Massimiliano Gnecchi
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Stefano Perlini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Francesco Salvatore
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Giampaolo Merlini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
43
|
Guan J, Mishra S, Qiu Y, Shi J, Trudeau K, Las G, Liesa M, Shirihai OS, Connors LH, Seldin DC, Falk RH, MacRae CA, Liao R. Lysosomal dysfunction and impaired autophagy underlie the pathogenesis of amyloidogenic light chain-mediated cardiotoxicity. EMBO Mol Med 2015; 6:1493-507. [PMID: 25319546 PMCID: PMC4237473 DOI: 10.15252/emmm.201404190] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AL amyloidosis is the consequence of clonal production of amyloidogenic immunoglobulin light chain (LC) proteins, often resulting in a rapidly progressive and fatal amyloid cardiomyopathy. Recent work has found that amyloidogenic LC directly initiate a cardio-toxic response underlying the pathogenesis of the cardiomyopathy; however, the mechanisms that contribute to this proteotoxicity remain unknown. Using human amyloidogenic LC isolated from patients with amyloid cardiomyopathy, we reveal that dysregulation of autophagic flux is critical for mediating amyloidogenic LC proteotoxicity. Restoration of autophagic flux by pharmacological intervention using rapamycin protected against amyloidogenic light chain protein-induced pathologies including contractile dysfunction and cell death at the cellular and organ level and also prolonged survival in an in vivo zebrafish model of amyloid cardiotoxicity. Mechanistically, we identify impaired lysosomal function to be the major cause of defective autophagy and amyloidogenic LC-induced proteotoxicity. Collectively, these findings detail the downstream molecular mechanisms underlying AL amyloid cardiomyopathy and highlight potential targeting of autophagy and lysosomal dysfunction in patients with amyloid cardiomyopathy.
Collapse
Affiliation(s)
- Jian Guan
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shikha Mishra
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yiling Qiu
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jianru Shi
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kyle Trudeau
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Guy Las
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Marc Liesa
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Orian S Shirihai
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Lawreen H Connors
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | - David C Seldin
- Amyloidosis Center, Boston University School of Medicine, Boston, MA, USA
| | - Rodney H Falk
- Cardiac Amyloidosis Program, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Calum A MacRae
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ronglih Liao
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA Cardiac Amyloidosis Program, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Dispenzieri A, Gertz MA, Saenger A, Kumar SK, Lacy MQ, Buadi FK, Dingli D, Leung N, Zeldenrust S, Hayman SR, Kapoor P, Grogan M, Hwa L, Russell SJ, Go RS, Rajkumar SV, Kyle RA, Jaffe A. Soluble suppression of tumorigenicity 2 (sST2), but not galactin-3, adds to prognostication in patients with systemic AL amyloidosis independent of NT-proBNP and troponin T. Am J Hematol 2015; 90:524-8. [PMID: 25753178 DOI: 10.1002/ajh.24001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 02/26/2015] [Accepted: 03/03/2015] [Indexed: 12/13/2022]
Abstract
The use of soluble cardiac biomarkers such as N-terminal pro-brain natriuretic peptide (NT-proBNP) and troponin has revolutionized prognostication for patients with AL amyloidosis. Soluble ST2 (sST2) and galectin-3 have also been reported to have prognostic value in other cardiac patient populations. We identified 502 patients with AL amyloidosis, who provided a research sample and consent to review their medical records between 1/1/2006-12/31/2010 within 90 days of their diagnosis. Samples were assayed for sST2 and galectin-3. Within this AL amyloidosis population, overall survival (OS) was 25.5 months (95% CI 18, 35.7 months). Receiver operating curve analyses were done to detect the best cut-points for sST2 and galectin-3 to predict both 1- and 5-year OS. The respective cut points for sST2 were 30 and 29.7 ng/mL, while the median sST2 for the entire population was 31 ng/mL (IQR 19.8, 53.6). The respective cut points for galectin-3 were 11 and 10.4 ng/mL while the median for the entire population was 16.6 ng/mL (IQR 11.5, 24.0). Although on univariate analysis, both sST2 and galectin-3 were prognostic, upon multivariate analysis, only sST2 was independent of troponin, NT-proBNP, serum immunoglobulin free light chain, and blood pressure. Not only did sST2 add to previously reported prognostication systems, but a novel prognostication 5-point system including sST2 was possible. The addition of sST2 - but not galectin-3 - to existing prognostication systems for patients with AL amyloidosis strengthens the ability to predict for death.
Collapse
Affiliation(s)
- Angela Dispenzieri
- Division of Hematology; Mayo Clinic; Rochester Minnesota
- Division of Laboratory Medicine; Mayo Clinic; Rochester Minnesota
| | - Morie A. Gertz
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | - Amy Saenger
- Division of Laboratory Medicine; Mayo Clinic; Rochester Minnesota
| | - Shaji K. Kumar
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | - Martha Q. Lacy
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | | | - David Dingli
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | - Nelson Leung
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | | | | | | | - Martha Grogan
- Division of Cardiology; Mayo Clinic; Rochester Minnesota
| | - Lisa Hwa
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | | | - Ronald S. Go
- Division of Hematology; Mayo Clinic; Rochester Minnesota
| | | | - Robert A. Kyle
- Division of Hematology; Mayo Clinic; Rochester Minnesota
- Division of Laboratory Medicine; Mayo Clinic; Rochester Minnesota
| | - Allan Jaffe
- Division of Laboratory Medicine; Mayo Clinic; Rochester Minnesota
- Division of Cardiology; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
45
|
Prognostic value of depressed midwall systolic function in cardiac light-chain amyloidosis. J Hypertens 2014; 32:1121-31; discussion 1131. [PMID: 24509117 DOI: 10.1097/hjh.0000000000000120] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Cardiac amyloidosis represents an archetypal form of restrictive heart disease, characterized by profound diastolic dysfunction. As ejection fraction is preserved until the late stage of the disease, the majority of patients do fulfill the definition of diastolic heart failure, that is, heart failure with preserved ejection fraction (HFpEF). In another clinical model of HFpEF, that is, pressure-overload hypertrophy, depressed midwall fractional shortening (mFS) has been shown to be a powerful prognostic factor. OBJECTIVE AND METHODS To assess the potential prognostic role of mFS in cardiac light-chain amyloidosis with preserved ejection fraction, we enrolled 221 consecutive untreated patients, in whom a first diagnosis of cardiac light-chain amyloidosis was concluded between 2008 and 2010. HFpEF was present in 181 patients. Patients in whom cardiac involvement was excluded served as controls (n = 121). Prognosis was assessed after a median follow-up of 561 days. RESULTS When compared with light-chain amyloidosis patients without myocardial involvement, cardiac light-chain amyloidosis was characterized by increased wall thickness (P <0.001), reduced end-diastolic left ventricular volumes (P <0.001), and diastolic dysfunction (P <0.001). In patients with preserved ejection fraction, mFS was markedly depressed [10.6% (8.7-13.5) vs. 17.8% (15.9-19.5) P <0.001]. At multivariable analysis, mFS, troponin I, and NT-pro-brain natriuretic peptide were the only significant prognostic determinants (P <0.001), whereas other indices of diastolic (E/E' ratio, transmitral and pulmonary vein flow velocities) and systolic function (tissue Doppler systolic indices, ejection fraction), or the presence/absence of congestive heart failure did not enter the model. CONCLUSION In cardiac light-chain amyloidosis with normal ejection fraction, depressed circumferential mFS, a marker of myocardial contractile dysfunction, is a powerful predictor of survival.
Collapse
|
46
|
Diomede L, Rognoni P, Lavatelli F, Romeo M, di Fonzo A, Foray C, Fiordaliso F, Palladini G, Valentini V, Perfetti V, Salmona M, Merlini G. Investigating heart-specific toxicity of amyloidogenic immunoglobulin light chains: A lesson from C. elegans. WORM 2014; 3:e965590. [PMID: 26430549 DOI: 10.4161/21624046.2014.965590] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/01/2014] [Accepted: 09/09/2014] [Indexed: 12/27/2022]
Abstract
Abnormalities in protein folding are involved in many localized and systemic diseases, all of which are characterized by insoluble amyloid formation and deposition. In immunoglobulin light chain (LC) amyloidosis, the most frequent systemic form of amyloidosis, the amyloid involvement of the heart dictates the prognosis and the elucidation of the mechanism of heart targeting and toxicity is essential for designing and testing new effective treatments. To this end, the availability of an appropriate animal model is crucial. We recently described the use of C. elegans as an innovative experimental system to investigate in vivo the pathogenic effects of monoclonal LC. This idea stems from the knowledge that the worm's pharynx is an "ancestral heart" with the additional ability to recognize stressor compounds. The feeding of worms with LC purified from patients suffering from cardiomyopathy, selectively and permanently impaired the pharyngeal function. This irreversible damage resulted in time, in a significant reduction in the lifespan of worms. We also reported that the ability of LC to generate reactive oxygen species was associated with their toxic effects and was counteracted by anti-oxidant compounds. This new nematode-based assay represents a promising model for elucidating the heart-specific toxicity of LC and for a rapid screening of new therapeutic strategies.
Collapse
Affiliation(s)
- Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology; IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" ; Milan, Italy
| | - Paola Rognoni
- Amyloid Research and Treatment Center; Foundation IRCCS Policlinico San Matteo ; Pavia, Italy
| | - Francesca Lavatelli
- Amyloid Research and Treatment Center; Foundation IRCCS Policlinico San Matteo ; Pavia, Italy
| | - Margherita Romeo
- Department of Molecular Biochemistry and Pharmacology; IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" ; Milan, Italy
| | - Andrea di Fonzo
- Amyloid Research and Treatment Center; Foundation IRCCS Policlinico San Matteo ; Pavia, Italy
| | - Claudia Foray
- Bio-imaging Unit, Department of Cardiovascular Research; IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" ; Milano, Italy
| | - Fabio Fiordaliso
- Bio-imaging Unit, Department of Cardiovascular Research; IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" ; Milano, Italy
| | - Giovanni Palladini
- Amyloid Research and Treatment Center; Foundation IRCCS Policlinico San Matteo ; Pavia, Italy ; Department of Molecular Medicine; University of Pavia ; Pavia, Italy
| | - Veronica Valentini
- Amyloid Research and Treatment Center; Foundation IRCCS Policlinico San Matteo ; Pavia, Italy
| | - Vittorio Perfetti
- Medical Oncology Unit; Foundation IRCCS Policlinico San Matteo ; Pavia, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology; IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri" ; Milan, Italy
| | - Giampaolo Merlini
- Amyloid Research and Treatment Center; Foundation IRCCS Policlinico San Matteo ; Pavia, Italy ; Department of Molecular Medicine; University of Pavia ; Pavia, Italy
| |
Collapse
|
47
|
Abstract
Amyloid cardiomyopathy should be suspected in any patient who presents with heart failure and preserved ejection fraction. In patients with echocardiographic evidence of ventricular thickening and without a clear history of hypertension, infiltrative cardiomyopathy should be considered. If imaging suggests the presence of amyloid deposits, confirmation by biopsy is required, although endomyocardial biopsy is generally not necessary. Assessment of aspirated subcutaneous fat and bone-marrow biopsy samples verifies the diagnosis in 40-80% of patients, dependent on the type of amyloidosis. Mass spectroscopy can be used to determine the protein subunit and classify the disease as immunoglobulin light-chain amyloidosis or transthyretin-related amyloidosis associated with mutant or wild-type TTR (formerly known as familial amyloid cardiomyopathy and senile cardiac amyloidosis, respectively). In this Review, we discuss the characteristics of cardiac amyloidosis, and present a structured approach to both the assessment of patients and treatment with emerging therapies and organ transplantation.
Collapse
Affiliation(s)
- Morie A Gertz
- Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Angela Dispenzieri
- Division of Clinical Biochemistry and Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Taimur Sher
- Division of Hematology/Oncology, Cancer Center, and Breast Clinic, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
48
|
Palladini G, Milani P, Foli A, Vidus Rosin M, Basset M, Lavatelli F, Nuvolone M, Obici L, Perlini S, Merlini G. Melphalan and dexamethasone with or without bortezomib in newly diagnosed AL amyloidosis: a matched case–control study on 174 patients. Leukemia 2014; 28:2311-6. [DOI: 10.1038/leu.2014.227] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/04/2014] [Accepted: 07/15/2014] [Indexed: 01/01/2023]
|
49
|
|
50
|
A Caenorhabditis elegans-based assay recognizes immunoglobulin light chains causing heart amyloidosis. Blood 2014; 123:3543-52. [PMID: 24665135 DOI: 10.1182/blood-2013-10-525634] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
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.
Collapse
|