Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Type of study
Publication year range
1.
EMBO J ; 43(8): 1445-1483, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499786

ABSTRACT

Regulatory T (TREG) cells develop via a program orchestrated by the transcription factor forkhead box protein P3 (FOXP3). Maintenance of the TREG cell lineage relies on sustained FOXP3 transcription via a mechanism involving demethylation of cytosine-phosphate-guanine (CpG)-rich elements at conserved non-coding sequences (CNS) in the FOXP3 locus. This cytosine demethylation is catalyzed by the ten-eleven translocation (TET) family of dioxygenases, and it involves a redox reaction that uses iron (Fe) as an essential cofactor. Here, we establish that human and mouse TREG cells express Fe-regulatory genes, including that encoding ferritin heavy chain (FTH), at relatively high levels compared to conventional T helper cells. We show that FTH expression in TREG cells is essential for immune homeostasis. Mechanistically, FTH supports TET-catalyzed demethylation of CpG-rich sequences CNS1 and 2 in the FOXP3 locus, thereby promoting FOXP3 transcription and TREG cell stability. This process, which is essential for TREG lineage stability and function, limits the severity of autoimmune neuroinflammation and infectious diseases, and favors tumor progression. These findings suggest that the regulation of intracellular iron by FTH is a stable property of TREG cells that supports immune homeostasis and limits the pathological outcomes of immune-mediated inflammation.


Subject(s)
Apoferritins , T-Lymphocytes, Regulatory , Animals , Humans , Mice , Apoferritins/genetics , Apoferritins/metabolism , Cell Lineage/genetics , Cytosine/metabolism , Forkhead Transcription Factors , Iron/metabolism
2.
Life Sci Alliance ; 7(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38307624

ABSTRACT

Severe presentations of malaria emerge as Plasmodium (P.) spp. parasites invade and lyse red blood cells (RBC), producing extracellular hemoglobin (HB), from which labile heme is released. Here, we tested whether scavenging of extracellular HB and/or labile heme, by haptoglobin (HP) and/or hemopexin (HPX), respectively, counter the pathogenesis of severe presentations of malaria. We found that circulating labile heme is an independent risk factor for cerebral and non-cerebral presentations of severe P. falciparum malaria in children. Labile heme was negatively correlated with circulating HP and HPX, which were, however, not risk factors for severe P. falciparum malaria. Genetic Hp and/or Hpx deletion in mice led to labile heme accumulation in plasma and kidneys, upon Plasmodium infection This was associated with higher incidence of mortality and acute kidney injury (AKI) in ageing but not adult Plasmodium-infected mice, and was corroborated by an inverse correlation between heme and HPX with serological markers of AKI in P. falciparum malaria. In conclusion, HP and HPX act in an age-dependent manner to prevent the pathogenesis of severe presentation of malaria in mice and presumably in humans.


Subject(s)
Acute Kidney Injury , Malaria , Child , Humans , Mice , Animals , Heme , Hemoglobins , Haptoglobins
3.
Int J Mol Sci ; 24(5)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36901722

ABSTRACT

NFIX, a member of the nuclear factor I (NFI) family of transcription factors, is known to be involved in muscle and central nervous system embryonic development. However, its expression in adults is limited. Similar to other developmental transcription factors, NFIX has been found to be altered in tumors, often promoting pro-tumorigenic functions, such as leading to proliferation, differentiation, and migration. However, some studies suggest that NFIX can also have a tumor suppressor role, indicating a complex and cancer-type dependent role of NFIX. This complexity may be linked to the multiple processes at play in regulating NFIX, which include transcriptional, post-transcriptional, and post-translational processes. Moreover, other features of NFIX, including its ability to interact with different NFI members to form homodimers or heterodimers, therefore allowing the transcription of different target genes, and its ability to sense oxidative stress, can also modulate its function. In this review, we examine different aspects of NFIX regulation, first in development and then in cancer, highlighting the important role of NFIX in oxidative stress and cell fate regulation in tumors. Moreover, we propose different mechanisms through which oxidative stress regulates NFIX transcription and function, underlining NFIX as a key factor for tumorigenesis.


Subject(s)
NFI Transcription Factors , Neoplasms , Humans , Cell Differentiation/physiology , NFI Transcription Factors/metabolism , Oxidative Stress
4.
Front Genet ; 13: 1107969, 2022.
Article in English | MEDLINE | ID: mdl-36568380
5.
Cell Metab ; 34(8): 1183-1200.e12, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35841892

ABSTRACT

Hypoglycemia is a clinical hallmark of severe malaria, the often-lethal outcome of Plasmodium falciparum infection. Here, we report that malaria-associated hypoglycemia emerges from a non-canonical resistance mechanism, whereby the infected host reduces glycemia to starve Plasmodium. This hypometabolic response is elicited by labile heme, a byproduct of hemolysis that induces illness-induced anorexia and represses hepatic glucose production. While transient repression of hepatic glucose production prevents unfettered immune-mediated inflammation, organ damage, and anemia, when sustained over time it leads to hypoglycemia, compromising host energy expenditure and adaptive thermoregulation. The latter arrests the development of asexual stages of Plasmodium via a mechanism associated with parasite mitochondrial dysfunction. In response, Plasmodium activates a transcriptional program associated with the reduction of virulence and sexual differentiation toward the generation of transmissible gametocytes. In conclusion, malaria-associated hypoglycemia represents a trade-off of a hypometabolic-based defense strategy that balances parasite virulence versus transmission.


Subject(s)
Hypoglycemia , Malaria, Falciparum , Malaria , Glucose , Humans , Malaria, Falciparum/parasitology , Plasmodium falciparum
6.
Front Genet ; 12: 673002, 2021.
Article in English | MEDLINE | ID: mdl-34394183

ABSTRACT

Cells are subjected to endogenous [e.g., reactive oxygen species (ROS), replication stress] and exogenous insults (e.g., UV light, ionizing radiation, and certain chemicals), which can affect the synthesis and/or stability of different macromolecules required for cell and tissue function. Oxidative stress, caused by excess ROS, and DNA damage, triggered in response to different sources, are countered and resolved by specific mechanisms, allowing the normal physiological equilibrium of cells and tissues to be restored. One process that is affected by oxidative stress and DNA damage is extracellular matrix (ECM) remodeling, which is a continuous and highly controlled mechanism that allows tissues to readjust in reaction to different challenges. The crosstalk between oxidative stress/DNA damage and ECM remodeling is not unidirectional. Quite on the contrary, mutations in ECM genes have a strong impact on tissue homeostasis and are characterized by increased oxidative stress and potentially also accumulation of DNA damage. In this review, we will discuss how oxidative stress and DNA damage affect the expression and deposition of ECM molecules and conversely how mutations in genes encoding ECM components trigger accumulation of oxidative stress and DNA damage. Both situations hamper the reestablishment of cell and tissue homeostasis, with negative impacts on tissue and organ function, which can be a driver for severe pathological conditions.

7.
Cell Host Microbe ; 29(3): 347-361.e12, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33497603

ABSTRACT

Most mammals express a functional GGTA1 gene encoding the N-acetyllactosaminide α-1,3-galactosyltransferase enzyme, which synthesizes Gal-α1-3Gal-ß1-4GlcNAc (α-gal) and are thus tolerant to this self-expressed glycan. Old World primates including humans, however, carry loss-of-function mutations in GGTA1 and lack α-gal. Presumably, fixation of such mutations was propelled by natural selection, favoring the emergence of α-gal-specific immunity, conferring resistance to α-gal-expressing pathogens. Here, we show that loss of Ggta1 function in mice enhances resistance to bacterial sepsis, irrespectively of α-Gal-specific immunity. Rather, the absence of α-gal from IgG-associated glycans increases IgG effector function via a mechanism associated with enhanced IgG-Fc gamma receptor (FcγR) binding. The ensuing survival advantage against sepsis comes alongside a cost of accelerated reproductive senescence in Ggta1-deleted mice. Mathematical modeling of this trade-off suggests that high exposure to virulent pathogens exerts sufficient selective pressure to fix GGTA1 loss-of-function mutations, as likely occurred during the evolution of primates toward humans.


Subject(s)
Biological Evolution , Disaccharides , Sepsis/microbiology , Animals , Bacteria , Carrier Proteins , DNA-Binding Proteins , Female , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Glycoproteins , Hominidae , Humans , Immunoglobulin G/immunology , Male , Mammals/immunology , Mice , Mice, Knockout , Polysaccharides , Primates
8.
Mol Metab ; 24: 64-79, 2019 06.
Article in English | MEDLINE | ID: mdl-30954544

ABSTRACT

OBJECTIVE: The ferritin heavy/heart chain (FTH) gene encodes the ferroxidase component of the iron (Fe) sequestering ferritin complex, which plays a central role in the regulation of cellular Fe metabolism. Here we tested the hypothesis that ferritin regulates organismal Fe metabolism in a manner that impacts energy balance and thermal homeostasis. METHODS: We developed a mouse strain, referred herein as FthR26 fl/fl, expressing a tamoxifen-inducible Cre recombinase under the control of the Rosa26 (R26) promoter and carrying two LoxP (fl) sites: one at the 5'end of the Fth promoter and another the 3' end of the first Fth exon. Tamoxifen administration induces global deletion of Fth in adult FthR26Δ/Δ mice, testing whether FTH is required for maintenance of organismal homeostasis. RESULTS: Under standard nutritional Fe supply, Fth deletion in adult FthR26Δ/Δ mice led to a profound deregulation of organismal Fe metabolism, oxidative stress, inflammation, and multi-organ damage, culminating in death. Unexpectedly, Fth deletion was also associated with a profound atrophy of white and brown adipose tissue as well as with collapse of energy expenditure and thermogenesis. This was attributed mechanistically to mitochondrial dysfunction, as assessed in the liver and in adipose tissue. CONCLUSION: The FTH component of ferritin acts as a master regulator of organismal Fe homeostasis, coupling nutritional Fe supply to organismal redox homeostasis, energy expenditure and thermoregulation.


Subject(s)
Energy Metabolism , Ferritins/metabolism , Thermogenesis , Adipose Tissue/metabolism , Animals , Cells, Cultured , Ferritins/genetics , Gene Deletion , Hepatocytes/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidative Stress
9.
Proc Natl Acad Sci U S A ; 116(12): 5681-5686, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30833408

ABSTRACT

Malaria, the disease caused by Plasmodium spp. infection, remains a major global cause of morbidity and mortality. Host protection from malaria relies on immune-driven resistance mechanisms that kill Plasmodium However, these mechanisms are not sufficient per se to avoid the development of severe forms of disease. This is accomplished instead via the establishment of disease tolerance to malaria, a defense strategy that does not target Plasmodium directly. Here we demonstrate that the establishment of disease tolerance to malaria relies on a tissue damage-control mechanism that operates specifically in renal proximal tubule epithelial cells (RPTEC). This protective response relies on the induction of heme oxygenase-1 (HMOX1; HO-1) and ferritin H chain (FTH) via a mechanism that involves the transcription-factor nuclear-factor E2-related factor-2 (NRF2). As it accumulates in plasma and urine during the blood stage of Plasmodium infection, labile heme is detoxified in RPTEC by HO-1 and FTH, preventing the development of acute kidney injury, a clinical hallmark of severe malaria.


Subject(s)
Heme/metabolism , Kidney/metabolism , Malaria/physiopathology , Animals , Apoferritins/metabolism , Cell Line , Disease Progression , Epithelial Cells/metabolism , Ferritins/metabolism , Ferritins/physiology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/physiology , Humans , Immune Tolerance/physiology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/physiology , Oxidoreductases , Plasmodium berghei/metabolism , Plasmodium berghei/parasitology , Up-Regulation
10.
Annu Rev Immunol ; 37: 405-437, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30673535

ABSTRACT

Pathogenic organisms exert a negative impact on host health, revealed by the clinical signs of infectious diseases. Immunity limits the severity of infectious diseases through resistance mechanisms that sense and target pathogens for containment, killing, or expulsion. These resistance mechanisms are viewed as the prevailing function of immunity. Under pathophysiologic conditions, however, immunity arises in response to infections that carry health and fitness costs to the host. Therefore, additional defense mechanisms are required to limit these costs, before immunity becomes operational as well as thereafter to avoid immunopathology. These are tissue damage control mechanisms that adjust the metabolic output of host tissues to different forms of stress and damage associated with infection. Disease tolerance is the term used to define this defense strategy, which does not exert a direct impact on pathogens but is essential to limit the health and fitness costs of infection. Under this argument, we propose that disease tolerance is an inherent component of immunity.


Subject(s)
Disease Resistance/immunology , Immunity, Innate , Infections/immunology , Microbiota/immunology , Animals , Host-Pathogen Interactions , Humans , Immune Tolerance , Immunomodulation
11.
Front Immunol ; 9: 2498, 2018.
Article in English | MEDLINE | ID: mdl-30425714

ABSTRACT

Infectious diseases are associated with disruption of host homeostasis. This can be triggered directly by pathogens or indirectly by host immune-driven resistance mechanisms. Disease tolerance is a defense strategy against infection that sustains host homeostasis, without exerting a direct negative impact on pathogens. The mechanisms governing disease tolerance encompass host metabolic responses that maintain vital homeostatic parameters within a range compatible with survival. Central to this defense strategy is the host's ability to sense and adapt to variations in nutrients, such as iron and glucose. Here we address how host responses regulating iron and glucose metabolism interact to establish disease tolerance and possibly modulate resistance to infection.


Subject(s)
Communicable Diseases/immunology , Glucose/metabolism , Host-Pathogen Interactions , Iron/metabolism , Nutritional Status , Adaptation, Physiological , Animals , Disease Resistance , Homeostasis , Humans , Immunity
13.
Cell ; 169(7): 1263-1275.e14, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28622511

ABSTRACT

Sepsis is an often lethal syndrome resulting from maladaptive immune and metabolic responses to infection, compromising host homeostasis. Disease tolerance is a defense strategy against infection that preserves host homeostasis without exerting a direct negative impact on pathogens. Here, we demonstrate that induction of the iron-sequestering ferritin H chain (FTH) in response to polymicrobial infections is critical to establish disease tolerance to sepsis. The protective effect of FTH is exerted via a mechanism that counters iron-driven oxidative inhibition of the liver glucose-6-phosphatase (G6Pase), and in doing so, sustains endogenous glucose production via liver gluconeogenesis. This is required to prevent the development of hypoglycemia that otherwise compromises disease tolerance to sepsis. FTH overexpression or ferritin administration establish disease tolerance therapeutically. In conclusion, disease tolerance to sepsis relies on a crosstalk between adaptive responses controlling iron and glucose metabolism, required to maintain blood glucose within a physiologic range compatible with host survival.


Subject(s)
Glucose/metabolism , Iron/metabolism , Sepsis/metabolism , Animals , Apoferritins/genetics , Apoferritins/metabolism , Ceruloplasmin/metabolism , Gluconeogenesis , Glucose-6-Phosphatase/metabolism , Mice , Mice, Inbred C57BL
15.
EMBO J ; 34(3): 410-24, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25582120

ABSTRACT

Loss of telomere protection occurs during physiological cell senescence and ageing, due to attrition of telomeric repeats and insufficient retention of the telomere-binding factor TRF2. Subsequently formed telomere fusions trigger rampant genomic instability leading to cell death or tumorigenesis. Mechanistically, telomere fusions require either the classical non-homologous end-joining (C-NHEJ) pathway dependent on Ku70/80 and LIG4, or the alternative non-homologous end-joining (A-NHEJ), which relies on PARP1 and LIG3. Here, we show that the tumour suppressor BRCA1, together with its interacting partner CtIP, both acting in end resection, also promotes end-joining of uncapped telomeres. BRCA1 and CtIP do not function in the ATM-dependent telomere damage signalling, nor in telomere overhang removal, which are critical for telomere fusions by C-NHEJ. Instead, BRCA1 and CtIP act in the same pathway as LIG3 to promote joining of de-protected telomeres by A-NHEJ. Our work therefore ascribes novel roles for BRCA1 and CtIP in end-processing and fusion reactions at uncapped telomeres, underlining the complexity of DNA repair pathways that act at chromosome ends lacking protective structures. Moreover, A-NHEJ provides a mechanism of previously unanticipated significance in telomere dysfunction-induced genome instability.


Subject(s)
BRCA1 Protein/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA End-Joining Repair/physiology , Telomere/metabolism , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , DNA Damage , DNA Ligase ATP , DNA Ligases/genetics , DNA Ligases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Ku Autoantigen , Mice , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Poly-ADP-Ribose Binding Proteins , Telomere/genetics , Xenopus Proteins
16.
Nat Commun ; 4: 2697, 2013.
Article in English | MEDLINE | ID: mdl-24162189

ABSTRACT

ARF is a tumour suppressor activated by oncogenic stress, which stabilizes p53. Although p53 is a key component of the response to DNA damage, a similar function for ARF has not been ascribed. Here we show that primary mouse and human cells lacking the tumour suppressor BRCA2 accumulate DNA damage, which triggers checkpoint signalling and ARF activation. Furthermore, senescence induced by Brca2 deletion in primary mouse and human cells is reversed by the loss of ARF, a phenotype recapitulated in cells lacking RAD51. Surprisingly, ARF is not necessary for p53 accumulation per se but for altering the spectrum of genes activated by this transcription factor. Specifically, ARF enables p53 transcription of Dusp4 and Dusp7, which encode a pair of phosphatases known to inactivate the MAP kinases ERK1/2. Our results ascribe a previously unanticipated function to the ARF tumour suppressor in genome integrity, controlled by replicative stress and ATM/ATR-dependent checkpoint responses.


Subject(s)
BRCA2 Protein/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Cricetinae , DNA Damage , Epigenesis, Genetic , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , Gene Deletion , HeLa Cells , Humans , Mice , Phenotype , Phosphoric Monoester Hydrolases/metabolism , RNA, Small Interfering/metabolism , Rad51 Recombinase/metabolism , Signal Transduction , Transcription Factors
17.
Nat Struct Mol Biol ; 17(12): 1461-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21076401

ABSTRACT

The tumor suppressor protein BRCA2 is a key component of the homologous recombination pathway of DNA repair, acting as the loader of RAD51 recombinase at sites of double-strand breaks. Here we show that BRCA2 associates with telomeres during the S and G2 phases of the cell cycle and facilitates the loading of RAD51 onto telomeres. Conditional deletion of Brca2 and inhibition of Rad51 in mouse embryonic fibroblasts (MEFs), but not inactivation of Brca1, led to shortening of telomeres and accumulation of fragmented telomeric signals--a hallmark of telomere fragility that is associated with replication defects. These findings suggest that BRCA2-mediated homologous recombination reactions contribute to the maintenance of telomere length by facilitating telomere replication and imply that BRCA2 has an essential role in maintaining telomere integrity during unchallenged cell proliferation. Mouse mammary tumors that lacked Brca2 accumulated telomere dysfunction-induced foci. Human breast tumors in which BRCA2 was mutated had shorter telomeres than those in which BRCA1 was mutated, suggesting that the genomic instability in BRCA2-deficient tumors was due in part to telomere dysfunction.


Subject(s)
BRCA2 Protein/physiology , Rad51 Recombinase/metabolism , Telomere/metabolism , Animals , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , DNA Repair , G2 Phase , Gene Deletion , Genomic Instability , Mice , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/genetics , S Phase , Telomere/chemistry
18.
Int J Food Microbiol ; 129(2): 194-9, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19108923

ABSTRACT

Enterococci are ubiquitous organisms able to promote both health (fermented food/probiotics) and illness (human/animal infections). Disturbingly, several enterococcal species commonly found in artisanal cheeses, such as Enterococcus faecalis and E. faecium, are being increasingly established as causes of infection, posing a problem for food safety. In this study enterococci from ewe's milk and cheese were compared to clinical and reference strains by growth in media simulating environmental colonization and infection sites: 2YT, BHI, skim milk, urine and rabbit serum at different pHs, NaCl concentrations and temperatures. Growth curves were obtained with Microbiology Workstation Bioscreen C and used to calculate relative indexes--RIs--(based on absorbance, lag phase and specific growth rate) for each strain and environmental condition. Similar or higher RIs were obtained for food strains growing in infection-related environments when compared to clinical ones, revealing their ability to adapt and grow in these conditions. A dendrogram built using Pearson's correlation coefficient and a PCA analysis clustered the strains regardless of their origin or species allocation, suggesting a strain-specific mode of growth and a high environmental adaptability of enterococcal strains. These evidences turn essential the evaluation of strains to be used as starters or probiotics.


Subject(s)
Cheese/microbiology , Consumer Product Safety , Enterococcus/growth & development , Milk/microbiology , Animals , Colony Count, Microbial , Dairy Products/microbiology , Enterococcus faecalis/growth & development , Enterococcus faecalis/pathogenicity , Enterococcus faecium/growth & development , Enterococcus faecium/pathogenicity , Female , Food Contamination/analysis , Food Contamination/prevention & control , Food Microbiology , Humans , Kinetics , Probiotics , Sheep , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...