Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Tumori ; 109(6): NP14-NP20, 2023 Dec.
Article En | MEDLINE | ID: mdl-37265183

Multiple myeloma is a hematological cancer characterized by relapse after treatment and poor prognosis. Ixazomib, a second-generation protease inhibitor, is one of the most recently available treatments for relapsed or refractory multiple myeloma, while it has also shown good potential as antitumoral agent in preclinical solid tumor models such as breast cancer cell lines. Here we report the case of a 68-year-old female with multiple myeloma and an incidental cT1b (9 mm) hormone receptor positive breast cancer lesion that showed a complete pathological response to a three-month combination therapy with Ixazomib, bendamustine and dexamethasone and no signs of disease relapse during the later follow-up. This is the first case report describing such clinical outcome in breast cancer following Ixazomib, bendamustine and dexamethasone combination therapy. To investigate the potential antitumoral activity of Ixazomib in breast cancer, we performed in vitro experiments using two hormone receptor positive breast cancer cell lines. We assessed the synergism between Ixazomib and bendamustine and the antiproliferative effect of Ixazomib. We found no synergistic interaction between the two drugs, while Ixazomib alone showed an antiproliferative effect against tumoral cells, suggesting that this drug has been responsible for tumor regression in our case.


Breast Neoplasms , Multiple Myeloma , Female , Humans , Aged , Multiple Myeloma/diagnosis , Bendamustine Hydrochloride/therapeutic use , Breast Neoplasms/drug therapy , Dexamethasone , Neoplasm Recurrence, Local/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Recurrence
2.
Crit Rev Oncol Hematol ; 185: 103963, 2023 May.
Article En | MEDLINE | ID: mdl-36931614

Liquid biopsy has the potential to drastically change clinical practice, paving the way to a novel non-invasive approach for cancer diagnosis and treatment. One of the limitations for the implementation of liquid biopsy in clinical practice is the lack of shared and reproducible standard operating procedures (SOPs) for sample collection, processing and storage. Here, we present a critical review of the literature focusing on the available SOPs to guide liquid biopsy management in research settings and describe SOPs that our laboratory developed and employed in the context of a prospective clinical-translational trial (RENOVATE, NCT04781062). The main aim of this manuscript is to address common issues, towards the implementation of interlaboratory shared protocols for optimized preanalytical handling of blood and urine samples. To our knowledge, this work is one of the few up-to-date, freely available comprehensive reports on trial-level procedures for the handling of liquid biopsy.


Specimen Handling , Humans , Prospective Studies , Specimen Handling/methods , Liquid Biopsy , Biomarkers
3.
Sci Adv ; 8(45): eabp9961, 2022 11 11.
Article En | MEDLINE | ID: mdl-36367935

Knowledge of the mechanisms underpinning the development of protective immunity conferred by mRNA vaccines is fragmentary. Here, we investigated responses to coronavirus disease 2019 (COVID-19) mRNA vaccination via high-temporal resolution blood transcriptome profiling. The first vaccine dose elicited modest interferon and adaptive immune responses, which peaked on days 2 and 5, respectively. The second vaccine dose, in contrast, elicited sharp day 1 interferon, inflammation, and erythroid cell responses, followed by a day 5 plasmablast response. Both post-first and post-second dose interferon signatures were associated with the subsequent development of antibody responses. Yet, we observed distinct interferon response patterns after each of the doses that may reflect quantitative or qualitative differences in interferon induction. Distinct interferon response phenotypes were also observed in patients with COVID-19 and were associated with severity and differences in duration of intensive care. Together, this study also highlights the benefits of adopting high-frequency sampling protocols in profiling vaccine-elicited immune responses.


COVID-19 Vaccines , COVID-19 , Humans , COVID-19/prevention & control , RNA, Messenger/genetics , Vaccines, Synthetic , Interferons , mRNA Vaccines
4.
Front Genet ; 12: 670727, 2021.
Article En | MEDLINE | ID: mdl-34211499

C3 Glomerulopathy (C3G) and Immune Complex-Mediated Membranoproliferative glomerulonephritis (IC-MPGN) are rare diseases characterized by glomerular deposition of C3 caused by dysregulation of the alternative pathway (AP) of complement. In approximately 20% of affected patients, dysregulation is driven by pathogenic variants in the two components of the AP C3 convertase, complement C3 (C3) and Factor B (CFB), or in complement Factor H (CFH) and Factor I (CFI), two genes that encode complement regulators. Copy number variations (CNVs) involving the CFH-related genes (CFHRs) that give rise to hybrid FHR proteins also have been described in a few C3G patients but not in IC-MPGN patients. In this study, we used multiplex ligation-dependent probe amplification (MLPA) to study the genomic architecture of the CFH-CFHR region and characterize CNVs in a large cohort of patients with C3G (n = 103) and IC-MPGN (n = 96) compared to healthy controls (n = 100). We identified new/rare CNVs resulting in structural variants (SVs) in 5 C3G and 2 IC-MPGN patients. Using long-read single molecule real-time sequencing (SMRT), we detected the breakpoints of three SVs. The identified SVs included: 1) a deletion of the entire CFH in one patient with IC-MPGN; 2) an increased number of CFHR4 copies in one IC-MPGN and three C3G patients; 3) a deletion from CFHR3-intron 3 to CFHR3-3'UTR (CFHR34 - 6 Δ) that results in a FHR3-FHR1 hybrid protein in a C3G patient; and 4) a CFHR31 - 5-CFHR410 hybrid gene in a C3G patient. This work highlights the contribution of CFH-CFHR CNVs to the pathogenesis of both C3G and IC-MPGN.

5.
Front Med (Lausanne) ; 7: 579418, 2020.
Article En | MEDLINE | ID: mdl-33224962

Atypical hemolytic uremic syndrome (aHUS) is an ultra-rare disease characterized by microangiopathic hemolysis, thrombocytopenia, and renal impairment and is associated with dysregulation of the alternative complement pathway on the microvascular endothelium. Outcomes have improved greatly with pharmacologic complement C5 blockade. Abnormalities in complement genes (CFH, CD46, CFI, CFB, C3, and THBD), CFH-CFHR genomic rearrangements, and anti-FH antibodies have been reported in 40-60% of cases. The penetrance of aHUS is incomplete in carriers of complement gene abnormalities; and multiple hits, including the CFH-H3 and CD46 GGAAC risk haplotypes and the CFHR1 * B risk allele, as well as environmental factors, contribute to disease development. Here, we investigated the determinants of penetrance of aHUS associated with CD46 genetic abnormalities. We studied 485 aHUS patients and found CD46 rare variants (RVs) in about 10%. The c.286+2T>G RV was the most prevalent (13/485) and was associated with <30% penetrance. We conducted an in-depth study of a large pedigree including a proband who is heterozygous for the c.286+2T>G RV who experienced a severe form of aHUS and developed end-stage renal failure. The father and paternal uncle with the same variant in homozygosity and six heterozygous relatives are unaffected. Flow cytometry analysis showed about 50% reduction of CD46 expression on blood mononuclear cells from the heterozygous proband and over 90% reduction in cells from the proband's unaffected homozygous father and aunt. Further genetic studies did not reveal RVs in known aHUS-associated genes or common genetic modifiers that segregated with the disease. Importantly, a specific ex vivo test showed excessive complement deposition on endothelial cells exposed to sera from the proband, and also from his mother and maternal uncle, who do not carry the c.286+2T>G RV, indicating that they share a circulating defect that results in complement dysregulation on the endothelium. These results highlight the complexity of the genetics of aHUS and indicate that CD46 deficiency may not be enough to induce aHUS. We hypothesize that the proband inherited from his mother a genetic abnormality in a complement circulating factor that has not been identified yet, which synergized with the CD46 RV in predisposing him to the aHUS phenotype.

6.
J Cell Physiol ; 235(4): 3508-3518, 2020 04.
Article En | MEDLINE | ID: mdl-31549411

Muscle loss is a major problem for many in lifetime. Muscle and bone degeneration has also been observed in individuals exposed to microgravity and in unloading conditions. C2C12 myoblst cells are able to form myotubes, and myofibers and these cells have been employed for muscle regeneration purposes and in myogenic regeneration and transplantation studies. We exposed C2C12 cells in an random position machine to simulate microgravity and study the energy and the biochemical challenges associated with this treatment. Simulated microgravity exposed C2C12 cells maintain positive proliferation indices and delay the differentiation process for several days. On the other hand this treatment significantly alters many of the biochemical and the metabolic characteristics of the cell cultures including calcium homeostasis. Recent data have shown that these perturbations are due to the inhibition of the ryanodine receptors on the membranes of intracellular calcium stores. We were able to reverse this perturbations treating cells with thapsigargin which prevents the segregation of intracellular calcium ions in the mitochondria and in the sarco/endoplasmic reticula. Calcium homeostasis appear a key target of microgravity exposure. In conclusion, in this study we reported some of the effects induced by the exposure of C2C12 cell cultures to simulated microgravity. The promising information obtained is of fundamental importance in the hope to employ this protocol in the field of regenerative medicine.


Cell Differentiation/physiology , Muscle Development/physiology , Regeneration/radiation effects , Weightlessness/adverse effects , Animals , Calcium Signaling/radiation effects , Cell Differentiation/genetics , Cell Line , Cell Proliferation/radiation effects , Humans , Mice , Muscle Development/radiation effects , Muscle Fibers, Skeletal/radiation effects , Myoblasts/metabolism , Myoblasts/radiation effects , Weightlessness Simulation/adverse effects
7.
Nephron ; 142(3): 264-270, 2019.
Article En | MEDLINE | ID: mdl-30889567

A 6-month-old boy presented with acute renal failure, thrombocytopenia, and severe non-immune hemolytic anemia. Infection by Shiga-like toxin-producing Escherichia coli and other causes of microangiopathic hemolysis were ruled out, leading to a diagnosis of atypical hemolytic uremic syndrome (aHUS). Neither pathogenic variants in HUS-associated genes nor anti-factor H antibodies were identified. Copy number variation analysis uncovered 4 copies of complement factor H related genes, CFHR1-CFHR4, conceivably leading to higher than normal levels of the corresponding proteins. However, this abnormality was also found in the healthy relatives, neither explaining the disease nor the excessive complement deposition on endothelial cells detected by an ex-vivo test. Whole-exome sequencing revealed a pathogenic homozygous variant in GRHPR encoding the glyoxylate and hydroxypyruvate reductase. Recessive GRHPR mutations cause primary hyperoxaluria type 2 (PH2). The presence of renal calculi in the patient and elevated oxalate levels in the urine were consistent with the genetic diagnosis of PH2. We hypothesize that, in this patient, hyperoxaluria caused by the GRHPR genetic defect triggered endothelial perturbation and complement activation, which was amplified by impaired factor H regulatory activity due to the increased -CFHR1-CFHR4 copy numbers, resulting in aHUS.


Atypical Hemolytic Uremic Syndrome/etiology , Hyperoxaluria, Primary/complications , Alcohol Oxidoreductases/genetics , Apolipoproteins/genetics , Complement Activation , Complement C3b Inactivator Proteins/genetics , Humans , Hyperoxaluria, Primary/genetics , Infant , Male , Exome Sequencing
8.
Am J Kidney Dis ; 74(1): 56-72, 2019 07.
Article En | MEDLINE | ID: mdl-30851964

RATIONALE & OBJECTIVE: Although primary atypical hemolytic uremic syndrome (aHUS) is associated with abnormalities in complement genes and antibodies to complement factor H, the role of complement in secondary aHUS remains debatable. We evaluated the usefulness of an ex vivo test to: (1) detect complement activation within the endothelium in primary and secondary aHUS, (2) differentiate active disease from remission, (3) monitor the effectiveness of eculizumab therapy, and (4) identify relapses during eculizumab dosage tapering and after discontinuation of treatment. STUDY DESIGN: Case series. SETTING & PARTICIPANTS: 121 patients with primary aHUS and 28 with secondary aHUS. Serum samples were collected during acute episodes, following remission, and during eculizumab treatment and were assessed using a serum-induced ex vivo C5b-9 endothelial deposition test. RESULTS: Serum-induced C5b-9 deposition on cultured microvascular endothelium was quantified by calculating the endothelial area covered by C5b-9 staining; values were expressed as percentage of C5b-9 deposits induced by a serum pool from healthy controls. Testing with adenosine diphosphate-activated endothelium demonstrated elevated C5b-9 deposits for all untreated patients with aHUS independent of disease activity, while testing with unstimulated endothelium demonstrated deposits only in active disease. Similar findings were observed in secondary aHUS. Serum-induced C5b-9 deposits on activated and unstimulated endothelium normalized during eculizumab treatment. 96% (22/23) of patients receiving eculizumab at extended 3- or 4-week dosing intervals demonstrated normal C5b-9 deposits on activated endothelium, despite most patients having CH50Eq (serum complement activity) > 20 UEq/mL, indicating that adequate complement control was achieved even with incomplete blockade of circulating C5. During eculizumab dosage tapering or after treatment discontinuation, all patients experiencing relapses versus only 6% (1/17) of those in stable remission had elevated C5b-9 deposits on unstimulated endothelium. LIMITATIONS: The C5b-9 endothelial deposition test can be performed in only specialized laboratories. Findings on eculizumab dosage tapering need to be confirmed with longitudinal monitoring of C5b-9 deposition. CONCLUSIONS: The C5b-9 endothelial deposition assay may represent an advance in our ability to monitor aHUS activity and individualize therapy.


Antibodies, Monoclonal, Humanized , Atypical Hemolytic Uremic Syndrome , Complement Activation/drug effects , Complement Membrane Attack Complex/analysis , Drug Monitoring/methods , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/drug therapy , Complement Factor H/analysis , Complement Factor H/genetics , Complement Inactivating Agents/administration & dosage , Complement Inactivating Agents/pharmacokinetics , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Female , Humans , In Vitro Techniques/methods , Male , Reproducibility of Results , Secondary Prevention/methods , Secondary Prevention/statistics & numerical data
9.
Metab Syndr Relat Disord ; 17(1): 53-59, 2019 02.
Article En | MEDLINE | ID: mdl-30376422

PURPOSE: Fanconi anemia (FA) is a complex tumor-prone disease defined by an entangled genotype and phenotype. Despite enormous efforts in the last 20 years, a comprehensive and integrated view of the disease is still missing. The aim of this pilot study was to establish whether a global microRNA (miRNA) analysis approach could be helpful in defining aspects in FA phenotype, which might deserve future attention with the perspective to develop miRNA-based therapies. METHODS: miRNA array were employed to characterize the global miRNA (miRNoma) profile of FA RNA samples with respect to normal samples. RESULTS: We report and compare miRNA profile from two FA established cell lines and three FA patients. This analysis reveals that 36 and 64 miRNAs, respectively, are found differentially expressed (>2-fold variation and P < 0.05) in the samples from FA cell lines and FA patients. Overlap of these data results in 24 miRNAs as shared in the two sample populations. Available bioinformatics methods were used to predict target genes for the differentially expressed miRNAs and to perform pathway enrichment analysis. CONCLUSIONS: Seven pathway results associated with the FA phenotype. It is interesting to note that some of these pathways were previously unrelated to FA phenotype. It might be important to focus on these pathways not previously emerged as dysfunctional in FA to better define the pathophysiological context of this disease. This is the first report of a global miRNA analysis in FA.


Fanconi Anemia/genetics , MicroRNAs/genetics , Transcriptome , Case-Control Studies , Cell Line , Child , Fanconi Anemia/epidemiology , Female , Gene Expression Profiling , Genotype , Humans , Male , Microarray Analysis , Phenotype , Pilot Projects
10.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1214-1221, 2017 06.
Article En | MEDLINE | ID: mdl-28315453

Energetic metabolism plays an essential role in the differentiation of haematopoietic stem cells (HSC). In Fanconi Anaemia (FA), DNA damage is accumulated during HSC differentiation, an event that is likely associated with bone marrow failure (BMF). One of the sources of the DNA damage is altered mitochondrial metabolism and an associated increment of oxidative stress. Recently, altered mitochondrial morphology and a deficit in the energetic activity in FA cells have been reported. Considering that mitochondria are the principal site of aerobic ATP production, we investigated FA metabolism in order to understand what pathways are able to compensate for this energy deficiency. In this work, we report that the impairment in mitochondrial oxidative phosphorylation (OXPHOS) in FA cells is countered by an increase in glycolytic flux. By contrast, glutaminolysis appears lower with respect to controls. Therefore, it is possible to conclude that in FA cells glycolysis represents the main pathway for producing energy, balancing the NADH/NAD+ ratio by the conversion of pyruvate to lactate. Finally, we show that a forced switch from glycolytic to OXPHOS metabolism increases FA cell oxidative stress. This could be the cause of the impoverishment in bone marrow HSC during exit from the homeostatic quiescent state. This is the first work that systematically explores FA energy metabolism, highlighting its flaws, and discusses the possible relationships between these defects and BMF.


Fanconi Anemia/metabolism , Glycolysis , Mitochondria/metabolism , Oxidative Phosphorylation , Oxidative Stress , Cell Line , Fanconi Anemia/pathology , Humans , Mitochondria/pathology
11.
Sci Rep ; 6: 25441, 2016 05 05.
Article En | MEDLINE | ID: mdl-27146429

Isomorphic mutation of the SBDS gene causes Shwachman-Diamond syndrome (SDS). SDS is a rare genetic bone marrow failure and cancer predisposition syndrome. SDS cells have ribosome biogenesis and their protein synthesis altered, which are two high-energy consuming cellular processes. The reported changes in reactive oxygen species production, endoplasmic reticulum stress response and reduced mitochondrial functionality suggest an energy production defect in SDS cells. In our work, we have demonstrated that SDS cells display a Complex IV activity impairment, which causes an oxidative phosphorylation metabolism defect, with a consequent decrease in ATP production. These data were confirmed by an increased glycolytic rate, which compensated for the energetic stress. Moreover, the signalling pathways involved in glycolysis activation also appeared more activated; i.e. we reported AMP-activated protein kinase hyper-phosphorylation. Notably, we also observed an increase in a mammalian target of rapamycin phosphorylation and high intracellular calcium concentration levels ([Ca(2+)]i), which probably represent new biochemical equilibrium modulation in SDS cells. Finally, the SDS cell response to leucine (Leu) was investigated, suggesting its possible use as a therapeutic adjuvant to be tested in clinical trials.


Bone Marrow Cells/metabolism , Bone Marrow Diseases/metabolism , Calcium/metabolism , Cytochrome-c Oxidase Deficiency/metabolism , Exocrine Pancreatic Insufficiency/metabolism , Lipomatosis/metabolism , Mitochondria/metabolism , Proteins/genetics , Ribosomes/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/deficiency , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Bone Marrow Diseases/genetics , Bone Marrow Diseases/pathology , Cytochrome-c Oxidase Deficiency/genetics , Cytochrome-c Oxidase Deficiency/pathology , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Exocrine Pancreatic Insufficiency/genetics , Exocrine Pancreatic Insufficiency/pathology , Gene Expression Regulation , Glycolysis/genetics , Humans , Leucine/pharmacology , Lipomatosis/genetics , Lipomatosis/pathology , Mitochondria/drug effects , Mitochondria/pathology , Mutation , Phosphorylation , Primary Cell Culture , Protein Biosynthesis , Proteins/metabolism , Reactive Oxygen Species/metabolism , Ribosomes/drug effects , Ribosomes/pathology , Shwachman-Diamond Syndrome , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
12.
Sci Rep ; 5: 8088, 2015 Jan 28.
Article En | MEDLINE | ID: mdl-25627108

Fanconi Anemia (FA) is a rare and complex inherited blood disorder associated with bone marrow failure and malignancies. Many alterations in FA physiology appear linked to red-ox unbalance including alterations in the morphology and structure of nuclei, intermediate filaments and mitochondria, defective respiration, reduced ATP production and altered ATP/AMP ratio. These defects are consistently associated with impaired oxygen metabolism indeed treatment with antioxidants N-acetylcysteine (NAC) and resveratrol (RV) does rescue FA physiology. Due to the importance of the intracellular calcium signaling and its key function in the control of intracellular functions we were interested to study calcium homeostasis in FA. We found that FANCA cells display a dramatically low intracellular calcium concentration ([Ca(2+)]i) in resting conditions. This condition affects cellular responses to stress. The flux of Ca(2+) mobilized by H2O2 from internal stores is significantly lower in FANCA cells in comparison to controls. The low basal [Ca(2+)]i in FANCA appears to be an actively maintained process controlled by a finely tuned interplay between different intracellular Ca(2+) stores. The defects associated with the altered Ca(2+) homeostasis appear consistently overlapping those related to the unbalanced oxidative metabolism in FA cells underlining a contiguity between oxidative stress and calcium homeostasis.


Calcium/metabolism , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fibroblasts/metabolism , Homeostasis , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Calcium-Transporting ATPases/metabolism , Carbocyanines/metabolism , Cell Line , Fanconi Anemia Complementation Group Proteins/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Heterocyclic Compounds, 3-Ring/metabolism , Homeostasis/drug effects , Humans , Hydrogen Peroxide/pharmacology , Kinetics , Microscopy, Confocal , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Resveratrol , Stilbenes/pharmacology , Thapsigargin/pharmacology
13.
PLoS One ; 9(7): e104857, 2014.
Article En | MEDLINE | ID: mdl-25126945

Fanconi anemia (FA) is a genetic disorder characterised by chromosome instability, cytokine ipersensibility, bone marrow failure and abnormal haematopoiesis associated with acute myelogenous leukemia. Recent reports are contributing to characterize the peculiar FA metabolism. Central to these considerations appears that cells from complementation group A (FANCA) display an altered red-ox metabolism. Consequently the possibility to improve FA phenotypical conditions with antioxidants is considered. We have characterized from the structural and biochemical point of view the response of FANCA lymphocytes to N-acetyl-cysteine (NAC) and resveratrol (RV). Surprisingly both NAC and RV failed to revert all the characteristic of FA phenotype and moreover their effects are not super imposable. Our data suggest that we must be aware of the biological effects coming from antioxidant treatment.


Acetylcysteine/pharmacology , Antioxidants/pharmacology , Fanconi Anemia/drug therapy , Stilbenes/pharmacology , Cells, Cultured , Drug Evaluation, Preclinical , Fanconi Anemia/pathology , Humans , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Resveratrol
14.
Trends Mol Med ; 19(9): 513-4, 2013 Sep.
Article En | MEDLINE | ID: mdl-23932594

Fanconi anemia (FA) is a rare, complex disorder that manifests in childhood. Children with FA suffer bone marrow failure, leukemias, or solid tumors. FA-associated mutations are found in 15 proteins that are involved in DNA repair. Some of these proteins have extranuclear activities involving redox balance, apoptosis, and energy metabolism; and recent data demonstrate respiratory impairment in FA cells, suggesting that altered mitochondrial function is a factor in this disease.


Electron Transport Complex I/genetics , Fanconi Anemia/genetics , Mitochondria/pathology , Neoplasms/genetics , Anemia, Aplastic , Apoptosis/genetics , Bone Marrow Diseases , Bone Marrow Failure Disorders , Carcinogenesis/genetics , Child , DNA Repair/genetics , Electron Transport Complex I/metabolism , Fanconi Anemia/complications , Fanconi Anemia/pathology , Hemoglobinuria, Paroxysmal/complications , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/pathology , Humans , Mitochondria/genetics , Neoplasms/complications , Neoplasms/pathology
15.
Biochimie ; 95(10): 1838-47, 2013 Oct.
Article En | MEDLINE | ID: mdl-23831462

Growing number of publication has proved an increasing of cellular function of the Fanconi anemia proteins. To chromosome stability and DNA repair new roles have been attributed to FA proteins in oxidative stress response and homeostasis, immune response and cytokines sensibility, gene expression. Our work shows a new role for FA-A protein: the organization of the cellular structure. By 2D-PAGE of FA-A and correct fibroblasts treated and untreated with H2O2 we identify different expression of protein involved in the structural organization of nucleus, intermediate filaments and mitochondria. Immunofluorescence and electronic microscopy analysis clearly show an already altered cellular structure in normal culture condition and this worsted after oxidative stress. FA-A cell appears structurally prone to physiologic stress and this could explain part of the phenotype of FA cells.


Cytoskeleton/metabolism , Fanconi Anemia/metabolism , Fibroblasts/metabolism , Lamin Type A/metabolism , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Vimentin/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression , Humans , Hydrogen Peroxide/pharmacology , Lamin Type A/genetics , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Muscle Proteins/genetics , Mutation , Oxidation-Reduction , Oxidative Stress , Vimentin/genetics
16.
Biochimie ; 95(10): 1828-37, 2013 Oct.
Article En | MEDLINE | ID: mdl-23791750

Fanconi anemia (FA) is a rare and complex inherited blood disorder of the child. At least 15 genes are associated with the disease. The highest frequency of mutations belongs to groups A, C and G. Genetic instability and cytokine hypersensitivity support the selection of leukemic over non-leukemic stem cells. FA cellular phenotype is characterized by alterations in red-ox state, mitochondrial functionality and energy metabolism as reported in the past however a clear picture of the altered biochemical phenotype in FA is still elusive and the final biochemical defect(s) still unknown. Here we report an analysis of the respiratory fluxes in FANCA primary fibroblasts, lymphocytes and lymphoblasts. FANCA mutants show defective respiration through Complex I, diminished ATP production and metabolic sufferance with an increased AMP/ATP ratio. Respiration in FANCC mutants is normal. Treatment with N-acetyl-cysteine (NAC) restores oxygen consumption to normal level. Defective respiration in FANCA mutants appear correlated with the FA pro-oxidative phenotype which is consistent with the altered morphology of FANCA mitochondria. Electron microscopy measures indeed show profound alterations in mitochondrial ultrastructure and shape.


Adenosine Triphosphate/metabolism , Electron Transport Complex I/metabolism , Fanconi Anemia/metabolism , Fibroblasts/metabolism , Lymphocytes/metabolism , Mitochondria/metabolism , Acetylcysteine/pharmacology , Adenosine Monophosphate/metabolism , Adenylate Kinase/metabolism , Adolescent , Case-Control Studies , Cell Respiration/drug effects , Cells, Cultured , Child , Child, Preschool , Electron Transport Complex I/genetics , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Lymphocytes/drug effects , Lymphocytes/pathology , Microscopy, Electron , Mitochondria/genetics , Mitochondria/ultrastructure , Mutation , Oxidation-Reduction , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism
17.
FEBS J ; 279(14): 2479-94, 2012 Jul.
Article En | MEDLINE | ID: mdl-22578062

Fanconi's anemia (FA) patients face severe pathological consequences. Bone marrow failure, the major cause of death in FA, accounting for as much as 80-90% of FA mortality, appears to be significantly linked to excessive apoptosis of hematopoietic cells induced by oxidative stress. However, 20-25% of FA patients develop malignancies of myeloid origin. A survival strategy for bone marrow and hematopoietic cells under selective pressure evidently exists. This study reports that lymphoblastoid cell lines derived from two FA patients displayed significant resistance to oxidative stress induced by treatments with H(2) O(2) and various glutathione (GSH) inhibitors that induce production of reactive oxygen species, GSH depletion and mitochondrial membrane depolarization. Among the various GSH inhibitors employed, FA cells appear particularly resistant to menadione (5 µm) and ethacrynic acid (ETA, 50 µm), two drugs that specifically target mitochondria. Even after pre-treatment with buthionine sulfoximine, a GSH synthesis inhibitor that induces enhanced induction of reactive oxygen species, FA cells maintain significant resistance to these drugs. These data suggest that the resistance to oxidative stress and the altered mitochondrial and metabolic functionality found in the FA mutant cells used in this study may indicate the survival strategy that is adopted in FA cells undergoing transformation. The study of redox and mitochondria regulation in FA may be of assistance in diagnosis of the disease and in the care of patients.


Glutathione/antagonists & inhibitors , Glutathione/metabolism , Hydrogen Peroxide/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Buthionine Sulfoximine/pharmacology , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Ethacrynic Acid/pharmacology , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Flow Cytometry , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Vitamin K 3/pharmacology , Vitamins/pharmacology
18.
J Biomed Sci ; 17: 63, 2010 Jul 28.
Article En | MEDLINE | ID: mdl-20667080

BACKGROUND: Whether microgravity might influence tumour growth and carcinogenesis is still an open issue. It is not clear also if and how normal and transformed cells are differently solicited by microgravity. The present study was designed to verify this issue. METHODS: Two normal, LB and HSC93, and two transformed, Jurkat and 1310, lymphoblast cell lines were used as representative for the two conditions. Two lymphoblast lines from Fanconi's anemia patients group A and C (FA-A and FA-C, respectively), along with their isogenic corrected counterparts (FA-A-cor and FA-C-cor) were also used. Cell lines were evaluated for their proliferative ability, vitality and apoptotic susceptibility upon microgravity exposure in comparison with unexposed cells. Different parameters correlated to energy metabolism, glucose consumption, mitochondrial membrane potential (MMP), intracellular ATP content, red-ox balance and ability of the cells to repair the DNA damage product 8-OHdG induced by the treatment of the cells with 20 mM KBrO3 were also evaluated. RESULTS: Transformed Jurkat and 1310 cells appear resistant to the microgravitational challenge. On the contrary normal LB and HSC93 cells display increased apoptotic susceptibility, shortage of energy storages and reduced ability to cope with oxidative stress. FA-A and FA-C cells appear resistant to microgravity exposure, analogously to transformed cells. FA corrected cells did shown intermediate sensitivity to microgravity exposure suggesting that genetic correction does not completely reverts cellular phenotype. CONCLUSIONS: In the light of the reported results microgravity should be regarded as an harmful condition either when considering normal as well as transformed cells. Modeled microgravity and space-based technology are interesting tools in the biomedicine laboratory and offer an original, useful and unique approach in the study of cellular biochemistry and in the regulation of metabolic pathways.


Fanconi Anemia/physiopathology , Lymphocytes/physiology , Weightlessness/adverse effects , 8-Hydroxy-2'-Deoxyguanosine , Adenosine Triphosphate/metabolism , Analysis of Variance , Apoptosis/physiology , Cell Line, Transformed , Cell Proliferation , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Energy Metabolism/physiology , Glucose/analysis , Humans , Jurkat Cells/physiology , Membrane Potential, Mitochondrial/physiology , Poly(ADP-ribose) Polymerases/metabolism , Thiobarbituric Acid Reactive Substances/analysis
...