Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters








Publication year range
1.
Vaccines (Basel) ; 12(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39066428

ABSTRACT

Post-acute COVID-19 vaccination syndrome (PACVS) is a chronic disease triggered by SARS-CoV-2 vaccination (estimated prevalence 0.02%). PACVS is discriminated from the normal post-vaccination state by altered receptor antibodies, most notably angiotensin II type 1 and alpha-2B adrenergic receptor antibodies. Here, we investigate the clinical phenotype using a study registry encompassing 191 PACVS-affected persons (159 females/32 males; median ages: 39/42 years). Unbiased clustering (modified Jaccard index) of reported symptoms revealed a prevalent cross-cohort symptomatology of malaise and chronic fatigue (>80% of cases). Overlapping clusters of (i) peripheral nerve dysfunction, dysesthesia, motor weakness, pain, and vasomotor dysfunction; (ii) cardiovascular impairment; and (iii) cognitive impairment, headache, and visual and acoustic dysfunctions were also frequently represented. Notable abnormalities of standard serum markers encompassing increased interleukins 6 and 8 (>80%), low free tri-iodine thyroxine (>80%), IgG subclass imbalances (>50%), impaired iron storage (>50%), and increased soluble neurofilament light chains (>30%) were not associated with specific symptoms. Based on these data, 131/191 participants fit myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and simultaneously also several other established dysautonomia syndromes. Furthermore, 31/191 participants fit none of these syndromes. In conclusion, PACVS could either be an outlier of ME/CFS or a dysautonomia syndrome sui generis.

2.
Geriatrics (Basel) ; 8(6)2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38132491

ABSTRACT

BACKGROUND: The recommended chronic kidney disease (CKD) first-line diagnostic test is based on the creatinine-derived (estimated) glomerular filtration rate (eGFR). Cystatin C use may provide a better assessment. METHODS: We compared creatinine- and cystatin C-derived eGFR determination as the first-line diagnostic test for 112 hospital patients aged > 60 years (median = 76 years). The patients were judged to not have CKD (no-CKD group) according to the first-line diagnostic recommendations (n = 61, eGFR (CKD Epidemiology Collaboration (CKD-EPI)) ≥ 60 mL/min/1.73 m2, total urine protein < 150 mg/g creatinine, urinary red/white blood cells not increased) or classified to be at risk for kidney insufficiency due to aortic valve dysfunction (at-risk group; n = 51). The accuracy of the eGFR values was evaluated retrospectively with the final case diagnoses. RESULTS: The eGFR (Caucasian, Asian, pediatric, and adult formula (CAPA)) was found to be linearly correlated to the eGFR (CKD-EPI) (R2 = 0.5, slope = 0.69, p < 0.0001). In 93/112 (>80%) cases, the eGFR (CAPA) yielded lower values (on average ≈-20%). In 55/112 (49%) cases, the cystatin C-derived CKD stage was lower. CKD reclassification from no-CKD to a kidney-insufficient state (i.e., CKD1/2 to CKD3a/b or 4) or reclassification to a more severe kidney insufficiency (i.e., CKD3a → 3b/4 or 3b → 4) was found in 41/112 (37%) cases. A worse CKD classification (no-CKD → kidney-insufficient) based on the eGFR (CAPA) was plausible in 30% of cases in light of the final case diagnoses. CONCLUSION: In elderly patients (>60 years), renal function appears to be systematically overestimated by the creatinine-based eGFR (CKD-EPI), indicating that, for this group, the cystatin C-based eGFR (CAPA) should be used as the first-line diagnostic test.

3.
Vaccines (Basel) ; 11(11)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-38005974

ABSTRACT

SARS-CoV-2 mRNA vaccination can entail chronic fatigue/dysautonomia tentatively termed post-acute COVID-19 vaccination syndrome (PACVS). We explored receptor autoantibodies and interleukin-6 (IL-6) as somatic correlates of PACVS. Blood markers determined before and six months after first-time SARS-CoV-2 vaccination of healthy controls (N = 89; 71 females; mean/median age: 39/49 years) were compared with corresponding values of PACVS-affected persons (N = 191; 159 females; mean/median age: 40/39 years) exhibiting chronic fatigue/dysautonomia (≥three symptoms for ≥five months after the last SARS-CoV-2 mRNA vaccination) not due to SARS-CoV-2 infection and/or confounding diseases/medications. Normal vaccination response encompassed decreases in 11 receptor antibodies (by 25-50%, p < 0.0001), increases in two receptor antibodies (by 15-25%, p < 0.0001) and normal IL-6. In PACVS, serological vaccination-response appeared significantly (p < 0.0001) altered, allowing discrimination from normal post-vaccination state (sensitivity = 90%, p < 0.0001) by increased Angiotensin II type 1 receptor antibodies (cut-off ≤ 10.7 U/mL, ROC-AUC = 0.824 ± 0.027), decreased alpha-2B adrenergic receptor antibodies (cut-off ≥ 25.2 U/mL, ROC-AUC = 0.828 ± 0.025) and increased IL-6 (cut-off ≤ 2.3 pg/mL, ROC-AUC = 0.850 ± 0.022). PACVS is thus indicated as a somatic syndrome delineated/detectable by diagnostic blood markers.

4.
Vaccines (Basel) ; 11(3)2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36992182

ABSTRACT

(1) Background: The high incidence of SARS-CoV-2 infection in vaccinated persons underscores the importance of individualized re-vaccination. PanIg antibodies that act against the S1/-receptor binding domain quantified in serum by a routine diagnostic test (ECLIA, Roche) can be used to gauge the individual ex vivo capacity of SARS-CoV-2 neutralization. However, that test is not adapted to mutations in the S1/-receptor binding domain, having accumulated in SARS-CoV-2 variants. Therefore, it might be unsuited to determine immune-reactivity against SARS-CoV-2 BA.5.1. (2) Method: To address this concern, we re-investigated sera obtained six months after second vaccinations with un-adapted mRNA vaccine Spikevax (Moderna). We related serum levels of panIg against the S1/-receptor binding domain quantified by the un-adapted ECLIA with full virus neutralization capacity against SARS-CoV-2 B.1 or SARS-CoV-2 BA5.1. (3) Results: 92% of the sera exhibited sufficient neutralization capacity against the B.1 strain. Only 20% of the sera sufficiently inhibited the BA5.1 strain. Sera inhibiting BA5.1 could not be distinguished from non-inhibiting sera by serum levels of panIg against the S1/-receptor binding domain quantified by the un-adapted ECLIA. (4) Conclusion: Quantitative serological tests for an antibody against the S1/-receptor binding domain are unsuited as vaccination companion diagnostics, unless they are regularly adapted to mutations that have accumulated in that domain.

5.
ESC Heart Fail ; 10(2): 1258-1269, 2023 04.
Article in English | MEDLINE | ID: mdl-36717981

ABSTRACT

AIMS: A causal link between non-ischaemic heart failure (HF) and humoral autoimmunity against G-protein-coupled receptors (GPCR) remains unclear except for Chagas' cardiomyopathy. Uncertainty arises from ambiguous reports on incidences of GPCR autoantibodies, spurious correlations of autoantibody levels with disease activity, and lack of standardization and validation of measuring procedures for putatively cardio-pathogenic GPCR autoantibodies. Here, we use validated and certified immune assays presenting native receptors as binding targets. We compared candidate GPCR autoantibody species between HF patients and healthy controls and tested associations of serum autoantibody levels with serological, haemodynamic, metabolic, and functional parameters in HF. METHODS: Ninety-five non-ischaemic HF patients undergoing transcatheter endomyocardial biopsy and 60 healthy controls were included. GPCR autoantibodies were determined in serum by IgG binding to native receptors or a cyclic peptide (for ß1AR autoantibodies). In patients, cardiac function, volumes, and myocardial structural properties were assessed by cardiac magnetic resonance imaging; right heart catheterization served for determination of cardiac haemodynamics; endomyocardial biopsies were used for histological assessment of cardiomyopathy and determination of cardiac mitochondrial oxidative function by high-resolution respirometry. RESULTS: Autoantibodies against ß1 adrenergic (ß1 AR) , M5-muscarinic (M5AR), and angiotensin II type 2 receptors (AT2R) were increased in HF (all P < 0.001). Autoantibodies against α1 -adrenergic (α1 AR) and angiotensin II type 1 receptors (AT1R) were decreased in HF (all P < 0.001). Correlation of alterations of GPCR autoantibodies with markers of cardiac or systemic inflammation or cardiac damage, haemodynamics, myocardial histology, or left ventricular inflammation (judged by T2 mapping) were weak, even when corrected for total IgG. ß1 AR autoantibodies were related inversely to markers of left ventricular fibrosis indicated by T1 mapping (r = -0.362, P < 0.05) and global longitudinal strain (r = -0.323, P < 0.05). AT2R autoantibodies were associated with improved myocardial mitochondrial coupling as measured by high-resolution respirometry in myocardial biopsies (r = -0.352, P < 0.05). In insulin-resistant HF patients, AT2R autoantibodies were decreased (r = -.240, P < 0.05), and AT1R autoantibodies were increased (r = 0.212, P < 0.05). CONCLUSIONS: GPCR autoantibodies are markedly altered in HF. However, they are correlated poorly or even inversely to haemodynamic, metabolic, and functional markers of disease severity, myocardial histology, and myocardial mitochondrial efficiency. These observations do not hint towards a specific cardio-pathogenic role of GPCR autoantibodies and suggest that further investigations are required before specific therapies directed at GPCR autoantibodies can be clinically tested in non-ischaemic HF.


Subject(s)
Autoantibodies , Heart Failure , Humans , Angiotensin II , Heart Failure/pathology , Receptors, G-Protein-Coupled/metabolism , Inflammation , Immunoglobulin G
6.
Vaccines (Basel) ; 10(7)2022 Jun 29.
Article in English | MEDLINE | ID: mdl-35891208

ABSTRACT

Purpose: We describe a diagnostic procedure suitable for scheduling (re-)vaccination against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) according to individual state of humoral immunization. Methods: To clarify the relation between quantitative antibody measurements and humoral ex vivo immune responsiveness, we monitored 124 individuals before, during and six months after vaccination with Spikevax (Moderna, Cambridge, MA, USA). Antibodies against SARS-CoV-2 spike (S1) protein receptor-binding domain (S1-AB) and against nucleocapsid antigens were measured by chemiluminescent immunoassay (Roche). Virus-neutralizing activities were determined by surrogate assays (NeutraLISA, Euroimmune; cPass, GenScript). Neutralization of SARS-CoV-2 in cell culture (full virus NT) served as an ex vivo correlate for humoral immune responsiveness. Results: Vaccination responses varied considerably. Six months after the second vaccination, participants still positive for the full virus NT were safely determined by S1-AB levels ≥1000 U/mL. The full virus NT-positive fraction of participants with S1-AB levels <1000 U/mL was identified by virus-neutralizing activities >70% as determined by surrogate assays (NeutraLISA or cPas). Participants that were full virus NT-negative and presumably insufficiently protected could thus be identified by a sensitivity of >83% and a specificity of >95%. Conclusion: The described diagnostic strategy possibly supports individualized (re-)vaccination schedules based on simple and rapid measurement of serum-based SARS-CoV-2 antibody levels. Our data apply only to WUHAN-type SARS-CoV-2 virus and the current version of the mRNA vaccine from Moderna (Cambridge, MA, USA). Adaptation to other vaccines and more recent SARS-CoV-2 strains will require modification of cut-offs and re-evaluation of sensitivity/specificity.

7.
Aging (Albany NY) ; 12(16): 16609-16620, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32857064

ABSTRACT

Autoantibodies against muscarinic and beta1-adrenergic receptors are considered a potential cause and/or risk factor for chronic heart failure. Association of periodontitis with such autoantibodies and with impaired heart function has been observed in patients exposed to endemic Chagas' disease, which triggers by itself cardiomyopathy and receptor immunization.Here we studied the association between periodontitis, markers of cardiac injury and receptor autoimmunization in periodontitis patients (n = 147) not exposed to Chagas' disease. The autoantibodies were determined by IgG binding to native intact muscarinic and beta1-adrenergic receptors or to a cyclic peptide mimicking the disease-relevant conformational autoepitope presented by the active beta1-adrenergic receptor. Possible cardiac injury and inflammatory status were judged by serum levels of proBNP/Troponin I and CRP/IL-6, respectively. These parameters were analysed in healthy and periodontally diseased individuals as well as before and after periodontal therapy.Patients with periodontitis had significantly (p < 0.001) higher levels of autoantibodies against M5-muscarinic and beta1-adrenergic receptors, which further increased following periodontal therapy. Receptor autoantibodies were associated with increased inflammatory status but not with increased markers of cardiac injury. Thus, our data indicate that periodontitis triggers systemic inflammation, which is associated with receptor autoimmunization, and, independently thereof, with cardiac injury.


Subject(s)
Autoantibodies/blood , Periodontitis/immunology , Receptor, Muscarinic M5/immunology , Receptors, Adrenergic, beta-1/immunology , Adolescent , Adult , C-Reactive Protein/analysis , Case-Control Studies , Female , Humans , Inflammation Mediators/blood , Interleukin-6/blood , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Periodontitis/blood , Periodontitis/diagnosis , Periodontitis/therapy , Prospective Studies , Treatment Outcome , Troponin I/blood , Young Adult
8.
ESC Heart Fail ; 7(4): 1830-1841, 2020 08.
Article in English | MEDLINE | ID: mdl-32436653

ABSTRACT

AIMS: Chronic heart failure (CHF) can be caused by autoantibodies stimulating the heart via binding to first and/or second extracellular loops of cardiac ß1 -adrenoceptors. Allosteric receptor activation depends on conformational features of the autoantibody binding site. Elucidating these features will pave the way for the development of specific diagnostics and therapeutics. Our aim was (i) to fine-map the conformational epitope within the second extracellular loop of the human ß1 -adrenoceptor (ß1 ECII ) that is targeted by stimulating ß1 -receptor (auto)antibodies and (ii) to generate competitive cyclopeptide inhibitors of allosteric receptor activation, which faithfully conserve the conformational auto-epitope. METHODS AND RESULTS: Non-conserved amino acids within the ß1 ECII loop (compared with the amino acids constituting the ECII loop of the ß2 -adrenoceptor) were one by one replaced with alanine; potential intra-loop disulfide bridges were probed by cysteine-serine exchanges. Effects on antibody binding and allosteric receptor activation were assessed (i) by (auto)antibody neutralization using cyclopeptides mimicking ß1 ECII  ± the above replacements, and (ii) by (auto)antibody stimulation of human ß1 -adrenoceptors bearing corresponding point mutations. With the use of stimulating ß1 -receptor (auto)antibodies raised in mice, rats, or rabbits and isolated from exemplary dilated cardiomyopathy patients, our series of experiments unmasked two features of the ß1 ECII loop essential for (auto)antibody binding and allosteric receptor activation: (i) the NDPK211-214 motif and (ii) the intra-loop disulfide bond C209 ↔C215 . Of note, aberrant intra-loop disulfide bond C209 ↔C216 almost fully disrupted the functional auto-epitope in cyclopeptides. CONCLUSIONS: The conformational auto-epitope targeted by cardio-pathogenic ß1 -receptor autoantibodies is faithfully conserved in cyclopeptide homologues of the ß1 ECII loop bearing the NDPK211-214 motif and the C209 ↔C215 bridge while lacking cysteine C216 . Such molecules provide promising tools for novel diagnostic and therapeutic approaches in ß1 -autoantibody-positive CHF.


Subject(s)
Cardiomyopathy, Dilated , Heart Failure , Animals , Autoantibodies , Cardiomyopathy, Dilated/diagnosis , Epitopes , Heart Failure/diagnosis , Humans , Mice , Rabbits , Rats , Receptors, Adrenergic, beta-1/genetics
9.
J Appl Lab Med ; 4(3): 391-403, 2019 11.
Article in English | MEDLINE | ID: mdl-31659076

ABSTRACT

BACKGROUND: Dilated cardiomyopathy (DCM) is a common cause of heart failure with high morbidity and mortality rates. The association of anti-ß1 adrenergic receptor (ß1AR) autoantibodies with disease progression was shown by various studies and in vivo animal experiments. The prevalence of these disease-driving autoantibodies was estimated as 25% to 75% in DCM. The removal of autoantibodies or the interruption of their action leads to a prolonged improvement of heart function. However, presence and impact of the autoimmune aspect in DCM patients must be examined for targeted treatment. METHODS: We developed a heterogeneous immunoassay to support the diagnosis of anti-ß1AR autoantibody-induced DCM. The presentation of the native conformational epitope was enabled by reconstitution of human ß1AR into lipid bilayer nanodiscs, which stabilize the incorporated receptor in aqueous solution for measurements with standard immunological techniques. RESULTS: The incorporation of ß1AR into nanodiscs was verified by chromatographic, spectroscopic, and immunological methods. The functionality was shown by interaction assays with appropriate binding partners. Furthermore, ß1AR nanodiscs were applied to immunoassays for the detection of anti-ß1AR in human sera. Surface plasmon resonance spectroscopy and ELISA were developed, optimized, and validated. The optimized ß1AR nanodisc ELISA enabled a simultaneous measurement of 40 samples in duplicate. An interassay variance of 24% and an intraassay variance of 5% were determined. The limit of detection and the limit of quantification were determined as 0.64 ng/mL and 1.26 ng/mL, respectively (related to a monoclonal anti-ß1AR). CONCLUSIONS: Nanodisc technology is suitable as a novel biomimetic membrane system to stabilize and present ß1AR for detection of autoantibodies with immunological methods in DCM patients.


Subject(s)
Autoantibodies/blood , Autoantigens , Cardiomyopathy, Dilated/blood , Cardiomyopathy, Dilated/diagnosis , Immunoassay/methods , Receptors, Adrenergic, beta-1 , Adult , Autoantibodies/immunology , Autoantigens/immunology , Cardiomyopathy, Dilated/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Fluorescent Antibody Technique , Genes, Reporter , Humans , Male , Nanotechnology , Receptors, Adrenergic, beta-1/immunology , Sensitivity and Specificity
10.
Front Biosci (Landmark Ed) ; 23(11): 2146-2156, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29772551

ABSTRACT

Patients suffering from chronic heart failure (CHF) caused or promoted by autoantibodies against cardiac beta1-adrenergic receptors (beta1AR) could benefit from specific therapies aimed at tolerance induction, removal or neutralisation of beta1AR autoantibodies, provided the patients can be selected for these therapies by reliable detection and quantitation of beta1AR autoantibodies in their circulation and by a valid assessment of the autoantibodies's putative cardio-pathogenic potential. Here, we discuss the current state of knowledge regarding the effects of CHF-associated (auto)antibodies on beta1AR function and beta1AR-mediated signal tranduction and discuss the presumed role of these effects in the development and progression of CHF. Identification of disease-relevant functional autoantibody effects and their specific assessment in medical diagnostic will be a prerequesite for the implementation of novel specific therapies not only for CHF caused or promoted by beta1AR autoantibodies but alos for most other diseases involving autoantibodies that target G-protein coupled receptors.


Subject(s)
Autoantibodies/immunology , Heart Failure/immunology , Receptors, Adrenergic, beta-1/immunology , Signal Transduction/immunology , Animals , Autoantibodies/blood , Chronic Disease , Disease Progression , Heart Failure/blood , Heart Failure/diagnosis , Humans , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Prognosis
13.
Aging (Albany NY) ; 8(9): 1876-1895, 2016 08 28.
Article in English | MEDLINE | ID: mdl-27574892

ABSTRACT

Autophagy and the circadian clock counteract tissue degeneration and support longevity in many organisms. Accumulating evidence indicates that aging compromises both the circadian clock and autophagy but the mechanisms involved are unknown. Here we show that the expression levels of transcriptional repressor components of the circadian oscillator, most prominently the human Period homologue PER2, are strongly reduced in primary dermal fibroblasts from aged humans, while raising the expression of PER2 in the same cells partially restores diminished autophagy levels. The link between clock gene expression and autophagy is corroborated by the finding that the circadian clock drives cell-autonomous, rhythmic autophagy levels in immortalized murine fibroblasts, and that siRNA-mediated downregulation of PER2 decreases autophagy levels while leaving core clock oscillations intact. Moreover, the Period homologue lin-42 regulates autophagy and life span in the nematode Caenorhabditis elegans, suggesting an evolutionarily conserved role for Period proteins in autophagy control and aging. Taken together, this study identifies circadian clock proteins as set-point regulators of autophagy and puts forward a model, in which age-related changes of clock gene expression promote declining autophagy levels.


Subject(s)
Aging/genetics , Autophagy/genetics , Circadian Clocks/genetics , Gene Expression , Period Circadian Proteins/genetics , Adult , Aged , Aging/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Female , Fibroblasts/metabolism , Humans , Middle Aged , Period Circadian Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Young Adult
16.
Clin Chem Lab Med ; 54(4): 683-91, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26408610

ABSTRACT

BACKGROUND: Autoantibodies against ß1-adrenergic receptors (ß1AR) that stimulate cardiac cAMP-production play a causal role in the pathogenesis of human heart failure. Patients can be subjected to specific therapies, if the presence of potentially cardio-noxious ß1AR-autoantibodies is reliably diagnosed. This requires assessment of IgG-interactions with the native ß1AR because ß1AR-autoantibodies target a conformational epitope inadequately presented by denatured receptors or linear peptides. Here, we report on a standardised diagnostic procedure for the assessment of ß1AR-autoantibodies in heart failure patients, which is based on IgG-binding to native human ß1AR. METHODS: Good laboratory practice (GLP)-conform measurement of ß1AR-autoantibodies was based on flow-cytometric quantification of differential IgG-binding to native HT1080 cells overexpressing biofluorescent human ß1AR or not. Receptor-specific IgG-binding was derived from IgG-related median fluorescence of ß1AR-positive cells corrected for background staining of ß1AR-negative cells admixed to each measurement. The slope of IgG binding at two different concentrations was used as measure for the titre/avidity of ß1AR-autoantibodies. RESULTS: Sensitivity and specificity of the novel procedure for high ß1AR-autoantibody levels in dilated cardiomyopathy patients (n=40, NYHA class III-IV) relative to n=40 matched healthy subjects was >90%. It was similar to functional assays considered the gold standard and vastly superior to existing screening-procedures employing fixed cells or linear receptor-peptides as auto-antigenic targets. Inter-assay scatter was 7%-15% and linear dilution recovery was within ±10% of expected values throughout. CONCLUSIONS: The novel assay possibly provides a tool to determine true prevalence and clinical impact of ß1AR-autoantibodies. Furthermore, it may serve as companion diagnostic for therapies specifically directed at ß1AR-autoantibodies.


Subject(s)
Autoantibodies/blood , Autoantibodies/immunology , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/immunology , Flow Cytometry/standards , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/immunology , Adult , Aged , Cardiomyopathy, Dilated/blood , Female , Humans , Male , Middle Aged , Sensitivity and Specificity
18.
Gerontology ; 62(4): 409-16, 2016.
Article in English | MEDLINE | ID: mdl-26675034

ABSTRACT

Research into ageing and its underlying molecular basis enables us to develop and implement targeted interventions to ameliorate or cure its consequences. However, the efficacy of interventions often differs widely between individuals, suggesting that populations should be stratified or even individualized. Large-scale cohort studies in humans, similar systematic studies in model organisms as well as detailed investigations into the biology of ageing can provide individual validated biomarkers and mechanisms, leading to recommendations for targeted interventions. Human cohort studies are already ongoing, and they can be supplemented by in silico simulations. Systematic studies in animal models are made possible by the use of inbred strains or genetic reference populations of mice. Combining the two, a comprehensive picture of the various determinants of ageing and 'health span' can be studied in detail, and an appreciation of the relevance of results from model organisms to humans is emerging. The interactions between genotype and environment, particularly the psychosocial environment, are poorly studied in both humans and model organisms, presenting serious challenges to any approach to a personalized medicine of ageing. To increase the success of preventive interventions, we argue that there is a pressing need for an individualized evaluation of interventions such as physical exercise, nutrition, nutraceuticals and calorie restriction mimetics as well as psychosocial and environmental factors, separately and in combination. The expected extension of the health span enables us to refocus health care spending on individual prevention, starting in late adulthood, and on the brief period of morbidity at very old age.


Subject(s)
Aging , Healthy Aging , Precision Medicine/trends , Animals , Computational Biology , Humans , Longevity , Mice , Models, Animal
20.
Cell Cycle ; 14(11): 1704-15, 2015.
Article in English | MEDLINE | ID: mdl-25875233

ABSTRACT

To maintain genome stability, the thousands of replication origins of mammalian genomes must only initiate replication once per cell cycle. This is achieved by a strict temporal separation of ongoing replication in S phase, and the formation of pre-replicative complexes in the preceding G1 phase, which "licenses" each origin competent for replication. The contribution of the loading factor Cdc6 to the timing of the licensing process remained however elusive due to seemingly contradictory findings concerning stabilization, degradation and nuclear export of Cdc6. Using fluorescently tagged Cdc6 (Cdc6-YFP) expressed in living cycling cells, we demonstrate here that Cdc6-YFP is stable and chromatin-associated during mitosis and G1 phase. It undergoes rapid proteasomal degradation during S phase initiation followed by active export to the cytosol during S and G2 phases. Biochemical fractionation abolishes this nuclear exclusion, causing aberrant chromatin association of Cdc6-YFP and, likely, endogenous Cdc6, too. In addition, we demonstrate association of Cdc6 with centrosomes in late G2 and during mitosis. These results show that multiple Cdc6-regulatory mechanisms coexist but are tightly controlled in a cell cycle-specific manner.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication/physiology , Genomic Instability/physiology , Mitosis/physiology , Nuclear Proteins/metabolism , Bacterial Proteins/metabolism , Blotting, Western , Centrosome/metabolism , Chromatin/metabolism , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Proliferating Cell Nuclear Antigen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL