Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Int J Mol Sci ; 24(9)2023 Apr 27.
Article En | MEDLINE | ID: mdl-37175654

Parkinson's disease (PD) is a multifactorial neurodegenerative pathology characterized by the progressive loss of dopaminergic neurons in the substantia nigra of the brain. Aging is considered the main risk factor for the development of idiopathic PD. However, immunity and inflammation play a crucial role in the pathogenesis of this disorder. In mice, we showed that pro-inflammatory priming of the brain sensitizes to severe PD development, regardless of animal age. Age-related sub-acute inflammation, as well as the activation of the immune response upon exposure to harmful stimuli, enhances PD manifestations. The severity of PD is influenced by the engagement of host resistance mechanisms against infection based on the removal of iron (Fe) from the circulation. The sequestration of Fe by immune cells prevents pathogens from proliferating. However, it leads to the formation of a Fe-loaded circulating compartment. When entering the brain through a compromised blood-brain barrier, Fe-loaded immune cells contribute to enhancing neuroinflammation and brain Fe overload. Thus, pro-inflammatory priming of the brain exacerbates neuronal damage and represents a risk factor for the development of severe PD symptoms. Further investigations are now required to better understand whether therapeutic interventions inhibiting this phenomenon might protect against PD.


Parkinson Disease , Mice , Animals , Parkinson Disease/drug therapy , Brain/pathology , Substantia Nigra/pathology , Inflammation/pathology , Dopaminergic Neurons/pathology
2.
Microorganisms ; 11(4)2023 Mar 29.
Article En | MEDLINE | ID: mdl-37110302

Sub-chronic inflammation, caused by age-related dysbiosis, primes the brain to neuroinflammation and neurodegenerative diseases. Evidence revealed that Parkinson's disease (PD) might originate in the gut, demonstrating gastro-intestinal disturbances, as reported by PD patients long before developing motor symptoms. In this study, we conducted comparative analyses in relatively young and old mice maintained in conventional or gnotobiotic conditions. We aimed to confirm that the effects induced by age-related dysbiosis, rather than aging itself, sensitize to PD onset. This hypothesis was confirmed in germ-free (GF) mice, which proved resistant to the pharmacological induction of PD, regardless of their age. Contrary to conventional animals, old GF mice did not develop an inflammatory phenotype or an accumulation of iron in the brain, two catalysts sensitizing to disease onset. The resistance of GF mice to PD is reverted when colonized with stool collected from conventional old animals, but not if receiving bacterial content from young mice. Hence, changes in gut microbiota composition are a risk factor for PD development and can be targeted preventively by iron chelators, shown to protect the brain from pro-inflammatory intestinal priming that sensitizes to neuroinflammation and the development of severe PD.

3.
Elife ; 112022 12 07.
Article En | MEDLINE | ID: mdl-36476511

Anthracyclines are among the most used and effective anticancer drugs. Their activity has been attributed to DNA double-strand breaks resulting from topoisomerase II poisoning and to eviction of histones from select sites in the genome. Here, we show that the extensively used anthracyclines Doxorubicin, Daunorubicin, and Epirubicin decrease the transcription of nuclear factor kappa B (NF-κB)-dependent gene targets, but not interferon-responsive genes in primary mouse (Mus musculus) macrophages. Using an NMR-based structural approach, we demonstrate that anthracyclines disturb the complexes formed between the NF-κB subunit RelA and its DNA-binding sites. The anthracycline variants Aclarubicin, Doxorubicinone, and the newly developed Dimethyl-doxorubicin, which share anticancer properties with the other anthracyclines but do not induce DNA damage, also suppressed inflammation, thus uncoupling DNA damage from the effects on inflammation. These findings have implications for anticancer therapy and for the development of novel anti-inflammatory drugs with limited side effects for life-threatening conditions such as sepsis.


Anthracyclines , NF-kappa B , Animals , Mice , Anthracyclines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , DNA Damage , DNA
4.
Antioxid Redox Signal ; 35(6): 453-473, 2021 08 20.
Article En | MEDLINE | ID: mdl-33233941

Significance: There is still no cure for neurodegenerative diseases, such as Parkinson's disease (PD). Current treatments are based on the attempt to reduce dopaminergic neuronal loss, and multidisciplinary approaches have been used to provide only a temporary symptoms' relief. In addition to the difficulties of drugs developed against PD to access the brain, the specificity of those inhibitory compounds could be a concern. This because neurons might degenerate by activating distinct signaling pathways, which are often initiated by the same stimulus. Recent Advances: Apoptosis, necroptosis, and ferroptosis were shown to significantly contribute to PD progression and, so far, are the main death programs described as capable to alter brain homeostasis. Their activation is characterized by different biochemical and morphological features, some of which might even share the same molecular players. Critical Issues: If there is a pathological need to engage, in PD, multiple death programs, sequentially or simultaneously, is not clear yet. Possibly the activation of apoptosis, necroptosis, and/or ferroptosis correlates to different PD stages and symptom severities. This would imply that the efficacy of therapeutic approaches against neuronal death might depend on the death program they target and the relevance of this death pathway on a specific PD phase. Future Directions: In this review, we describe the molecular mechanisms underlying the activation of apoptosis, necroptosis, and ferroptosis in PD. Understanding the interrelationship between different death pathways' activation in PD is of utmost importance for the development of therapeutic approaches against disease progression. Antioxid. Redox Signal. 35, 453-473.


Dopaminergic Neurons/metabolism , Iron/metabolism , Parkinson Disease/metabolism , Animals , Cell Death , Humans
5.
Pharmaceuticals (Basel) ; 12(4)2019 Dec 05.
Article En | MEDLINE | ID: mdl-31817314

Iron is essential for almost all organisms, being involved in oxygen transport, DNA synthesis, and respiration; however, it is also potentially toxic via the formation of free radicals [...].

6.
Pharmaceuticals (Basel) ; 12(3)2019 Aug 29.
Article En | MEDLINE | ID: mdl-31470556

Iron is a critical element for most organisms, which plays a fundamental role in the great majority of physiological processes. So much so, that disruption of iron homeostasis has severe multi-organ impacts with the brain being particularly sensitive to such modifications. More specifically, disruption of iron homeostasis in the brain can affect neurophysiological mechanisms, cognition, and social behavior, which eventually contributes to the development of a diverse set of neuro-pathologies. This article starts by exploring the mechanisms of iron action in the brain and follows with a discussion on cognitive and behavioral implications of iron deficiency and overload and how these are framed by the social context. Subsequently, we scrutinize the implications of the disruption of iron homeostasis for the onset and progression of psychosocial disorders. Lastly, we discuss the links between biological, psychological, and social dimensions and outline potential avenues of research. The study of these interactions could ultimately contribute to a broader understanding of how individuals think and act under physiological and pathophysiological conditions.

7.
Proc Natl Acad Sci U S A ; 116(12): 5681-5686, 2019 03 19.
Article En | MEDLINE | ID: mdl-30833408

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.


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
8.
J Infect Dis ; 216(7): 907-918, 2017 10 17.
Article En | MEDLINE | ID: mdl-28973651

Background: Recent evidence indicates a robust competition between the host and mycobacteria for iron acquisition during mycobacterial infection. Variable effects of iron supplementation on the susceptibility to mycobacterial infection have been reported. In this study, we revisited the effects of an experimental iron-enriched diet on Mycobacterium bovis bacille Calmette-Guerin (BCG) infection. Methods: Mice fed a standard diet or a diet moderately enriched with iron were infected with M. bovis BCG expressing green fluorescent protein. Colony-forming unit numbers, host myeloid cell counts, cell recruitment, cytokine production, and iron gene expression were determined at different stages of infection. Bone marrow-derived macrophages incubated with or without iron were also used to measure bacterial uptake, levels of inflammation markers, and iron gene expression. Results: In vivo analysis of BCG-infected mice revealed that moderate iron supplementation reduced inflammation, as measured by decreased proinflammatory cytokine levels and neutrophil recruitment and enhanced T-cell recruitment in granulomas, and decreased the bacterial load. Enhanced bacterial clearance in the liver correlated with upregulation of the gene encoding hepcidin, which is known to have antimicrobial proprieties, and with sequestration of iron in tissues. In cultured macrophages, iron supplementation induced reactive oxygen species and reduced uptake and intracellular growth of BCG. Conclusion: Moderate iron diet supplementation diminished inflammation and growth of M. bovis BCG via enhanced reactive oxygen species production, immune cell activation, and local hepcidin expression.


Cytokines/metabolism , Hepcidins/metabolism , Iron, Dietary/pharmacology , Mycobacterium bovis/immunology , T-Lymphocytes/physiology , Tuberculosis/microbiology , Animals , Cytokines/genetics , Hepcidins/genetics , Iron/metabolism , Liver/metabolism , Liver/microbiology , Lung/metabolism , Lung/microbiology , Mice , Tuberculosis/immunology , Up-Regulation
9.
IUBMB Life ; 69(6): 442-450, 2017 06.
Article En | MEDLINE | ID: mdl-28474474

Iron (Fe) is essential to almost all organisms, as required by cells to satisfy metabolic needs and accomplish specialized functions. Its ability to exchange electrons between different substrates, however, renders it potentially toxic. Fine tune-mechanisms are necessary to maintain Fe homeostasis and, as such, to prevent its participation into the Fenton reaction and generation of oxidative stress. These are particularly important in the context of inflammation/infection, where restricting Fe availability to invading pathogens is one, if not, the main host defense strategy against microbial growth. The ability of Fe to modulate several aspects of the immune response is associated with a number of "costs" and "benefits", some of which have been described in this review. © 2017 IUBMB Life, 69(6):442-450, 2017.


Bacterial Infections/metabolism , Dendritic Cells/metabolism , Iron/metabolism , Lymphocytes/metabolism , Macrophages/metabolism , Mycoses/metabolism , Animals , Bacteria/metabolism , Bacteria/pathogenicity , Bacterial Infections/immunology , Bacterial Infections/microbiology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Fungi/metabolism , Fungi/pathogenicity , Gastrointestinal Absorption/physiology , Homeostasis/physiology , Humans , Immunity, Innate , Inflammation , Lymphocytes/immunology , Lymphocytes/microbiology , Macrophages/immunology , Macrophages/microbiology , Mycoses/immunology , Mycoses/microbiology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism
10.
Int J Mol Sci ; 17(1)2016 Jan 20.
Article En | MEDLINE | ID: mdl-26805813

Iron is required for the survival of most organisms, including bacteria, plants, and humans. Its homeostasis in mammals must be fine-tuned to avoid iron deficiency with a reduced oxygen transport and diminished activity of Fe-dependent enzymes, and also iron excess that may catalyze the formation of highly reactive hydroxyl radicals, oxidative stress, and programmed cell death. The advance in understanding the main players and mechanisms involved in iron regulation significantly improved since the discovery of genes responsible for hemochromatosis, the IRE/IRPs machinery, and the hepcidin-ferroportin axis. This review provides an update on the molecular mechanisms regulating cellular and systemic Fe homeostasis and their roles in pathophysiologic conditions that involve alterations of iron metabolism, and provides novel therapeutic strategies to prevent the deleterious effect of its deficiency/overload.


Aging/metabolism , Anemia, Iron-Deficiency/genetics , Hemochromatosis/genetics , Homeostasis/genetics , Iron Overload/genetics , Aging/genetics , Anemia, Iron-Deficiency/metabolism , Anemia, Iron-Deficiency/pathology , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Gene Expression Regulation , Heme/metabolism , Hemochromatosis/metabolism , Hemochromatosis/pathology , Hepcidins/genetics , Hepcidins/metabolism , Humans , Iron/metabolism , Iron Overload/metabolism , Iron Overload/pathology , Iron Regulatory Protein 1/genetics , Iron Regulatory Protein 1/metabolism , Iron Regulatory Protein 2/genetics , Iron Regulatory Protein 2/metabolism , Response Elements , Signal Transduction
11.
Curr Alzheimer Res ; 13(2): 174-84, 2016.
Article En | MEDLINE | ID: mdl-26391040

Heme is essential for the survival of most organisms, despite the fact of being potentially toxic. This dual effect is due to the ability of the iron (Fe) atom contained within the protoporphyrin ring of the heme molecule to participate in redox reactions and exchange electrons with a variety of substrates. Therefore, the pro-oxidant reactivity of heme needs to be kept under control, an effect achieved by its incorporation into the heme pockets of hemoproteins, i.e. proteins required to exert vital biological functions in which heme acts as prosthetic group. The release of heme from hemoproteins and the participation of Fe in the Fenton reaction lead to the generation of unfettered oxidative stress and programmed cell death. Although further investigations would be required to elucidate the regulation of heme in the brain, this molecule appears to be critically involved in the pathogenesis of different neurodegenerative diseases, as heme accumulation or deficiency is associated with impaired brain activity and neuronal death. Thus, the aim of this review is to provide an overview on the importance of heme in the brain and the pathophysiologic consequences associated with its accumulation.


Brain/physiopathology , Heme/metabolism , Neurodegenerative Diseases/physiopathology , Animals , Humans
12.
Biochem J ; 472(1): 1-15, 2015 Nov 15.
Article En | MEDLINE | ID: mdl-26518749

Ferritins, the main intracellular iron storage proteins, have been studied for over 60 years, mainly focusing on the mammalian ones. This allowed the elucidation of the structure of these proteins and the mechanisms regulating their iron incorporation and mineralization. However, ferritin is present in most, although not all, eukaryotic cells, comprising monocellular and multicellular invertebrates and vertebrates. The aim of this review is to provide an update on the general properties of ferritins that are common to various eukaryotic phyla (except plants), and to give an overview on the structure, function and regulation of ferritins. An update on the animal models that were used to characterize H, L and mitochondrial ferritins is also provided. The data show that ferritin structure is highly conserved among different phyla. It exerts an important cytoprotective function against oxidative damage and plays a role in innate immunity, where it also contributes to prevent parenchymal tissue from the cytotoxicity of pro-inflammatory agonists released by the activation of the immune response activation. Less clear are the properties of the secretory ferritins expressed by insects and molluscs, which may be important for understanding the role played by serum ferritin in mammals.


Apoferritins/metabolism , Cytoprotection , Eukaryota/metabolism , Ferritins/metabolism , Iron/metabolism , Amino Acid Sequence , Animals , Apoferritins/genetics , Eukaryota/classification , Eukaryota/genetics , Ferritins/genetics , Humans , Molecular Sequence Data , Oxidoreductases , Sequence Homology, Amino Acid
14.
Cell ; 159(6): 1277-89, 2014 Dec 04.
Article En | MEDLINE | ID: mdl-25480293

Glycosylation processes are under high natural selection pressure, presumably because these can modulate resistance to infection. Here, we asked whether inactivation of the UDP-galactose:ß-galactoside-α1-3-galactosyltransferase (α1,3GT) gene, which ablated the expression of the Galα1-3Galß1-4GlcNAc-R (α-gal) glycan and allowed for the production of anti-α-gal antibodies (Abs) in humans, confers protection against Plasmodium spp. infection, the causative agent of malaria and a major driving force in human evolution. We demonstrate that both Plasmodium spp. and the human gut pathobiont E. coli O86:B7 express α-gal and that anti-α-gal Abs are associated with protection against malaria transmission in humans as well as in α1,3GT-deficient mice, which produce protective anti-α-gal Abs when colonized by E. coli O86:B7. Anti-α-gal Abs target Plasmodium sporozoites for complement-mediated cytotoxicity in the skin, immediately after inoculation by Anopheles mosquitoes. Vaccination against α-gal confers sterile protection against malaria in mice, suggesting that a similar approach may reduce malaria transmission in humans.


Escherichia coli/physiology , Immunoglobulin M/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/transmission , Plasmodium/physiology , Polysaccharides/immunology , Adult , Animals , Anopheles/parasitology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Autoantigens/immunology , Cell Line, Tumor , Child , Escherichia coli/classification , Escherichia coli/immunology , Female , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Gastrointestinal Tract/microbiology , Germ-Free Life , Humans , Immunoglobulin M/blood , Malaria, Falciparum/microbiology , Malaria, Falciparum/parasitology , Mice , Plasmodium/classification , Plasmodium/growth & development , Plasmodium/immunology , Plasmodium falciparum/immunology , Plasmodium falciparum/physiology , Sporozoites/immunology , Toll-Like Receptor 9/agonists
15.
Trends Immunol ; 35(10): 483-94, 2014 Oct.
Article En | MEDLINE | ID: mdl-25182198

Immune-driven resistance mechanisms are the prevailing host defense strategy against infection. By contrast, disease tolerance mechanisms limit disease severity by preventing tissue damage or ameliorating tissue function without interfering with pathogen load. We propose here that tissue damage control underlies many of the protective effects of disease tolerance. We explore the mechanisms of cellular adaptation that underlie tissue damage control in response to infection as well as sterile inflammation, integrating both stress and damage responses. Finally, we discuss the potential impact of targeting these mechanisms in the treatment of disease.


Immune Tolerance/immunology , Animals , Host-Pathogen Interactions/immunology , Humans , Inflammation/immunology
16.
Front Pharmacol ; 5: 155, 2014.
Article En | MEDLINE | ID: mdl-25071574

Plasmodium infection during gestation may lead to severe clinical manifestations including abortion, stillbirth, intrauterine growth retardation, and low birth weight. Mechanisms underlying such poor pregnancy outcomes are still unclear. In the animal model of severe placental malaria (PM), in utero fetal death frequently occurs and mothers often succumb to infection before or immediately after delivery. Plasmodium berghei-infected erythrocytes (IEs) continuously accumulate in the placenta, where they are then phagocytosed by fetal-derived placental cells, namely trophoblasts. Inside the phagosomes, disruption of IEs leads to the release of non-hemoglobin bound heme, which is subsequently catabolized by heme oxygenase-1 into carbon monoxide, biliverdin, and labile iron. Fine-tuned regulatory mechanisms operate to maintain iron homeostasis, preventing the deleterious effect of iron-induced oxidative stress. Our preliminary results demonstrate that iron overload in trophoblasts of P. berghei-infected placenta is associated with fetal death. Placentas which supported normally developing embryos showed no iron accumulation within the trophoblasts. Placentas from dead fetuses showed massive iron accumulation, which was associated with parasitic burden. Here we present preliminary data suggesting that disruption of iron homeostasis in trophoblasts during the course of PM is a consequence of heme accumulation after intense IE engulfment. We propose that iron overload in placenta is a pathogenic component of PM, contributing to fetal death. The mechanism through which it operates still needs to be elucidated.

17.
Antioxid Redox Signal ; 20(11): 1754-69, 2014 Apr 10.
Article En | MEDLINE | ID: mdl-24124891

SIGNIFICANCE: Inflammation and immunity can be associated with varying degrees of heme release from hemoproteins, eventually leading to cellular and tissue iron (Fe) overload, oxidative stress, and tissue damage. Presumably, these deleterious effects contribute to the pathogenesis of systemic infections. RECENT ADVANCES: Heme release from hemoglobin sensitizes parenchyma cells to undergo programmed cell death in response to proinflammatory cytokines, such as tumor necrosis factor. This cytotoxic effect is driven by a mechanism involving intracellular accumulation of free radicals, which sustain the activation of the c-Jun N-terminal kinase (JNK) signaling transduction pathway. While heme catabolism by heme oxygenase-1 (HO-1) prevents programmed cell death, this cytoprotective effect requires the co-expression of ferritin H (heart/heavy) chain (FTH), which controls the pro-oxidant effect of labile Fe released from the protoporphyrin IX ring of heme. This antioxidant effect of FTH restrains JNK activation, whereas JNK activation inhibits FTH expression, a cross talk that controls metabolic adaptation to cellular Fe overload associated with systemic infections. CRITICAL ISSUES AND FUTURE DIRECTIONS: Identification and characterization of the mechanisms via which FTH provides metabolic adaptation to tissue Fe overload should provide valuable information to our current understanding of the pathogenesis of systemic infections as well as other immune-mediated inflammatory diseases.


Apoferritins/physiology , Heme/metabolism , Iron/metabolism , Animals , Heme Oxygenase-1/physiology , Homeostasis , Humans , Immunity, Innate
18.
Immunity ; 39(5): 874-84, 2013 Nov 14.
Article En | MEDLINE | ID: mdl-24184056

Severe sepsis remains a poorly understood systemic inflammatory condition with high mortality rates and limited therapeutic options in addition to organ support measures. Here we show that the clinically approved group of anthracyclines acts therapeutically at a low dose regimen to confer robust protection against severe sepsis in mice. This salutary effect is strictly dependent on the activation of DNA damage response and autophagy pathways in the lung, as demonstrated by deletion of the ataxia telangiectasia mutated (Atm) or the autophagy-related protein 7 (Atg7) specifically in this organ. The protective effect of anthracyclines occurs irrespectively of pathogen burden, conferring disease tolerance to severe sepsis. These findings demonstrate that DNA damage responses, including the ATM and Fanconi Anemia pathways, are important modulators of immune responses and might be exploited to confer protection to inflammation-driven conditions, including severe sepsis.


Anthracyclines/pharmacology , Anti-Bacterial Agents/pharmacology , DNA Repair/drug effects , Lung/drug effects , Peritonitis/drug therapy , Sepsis/prevention & control , Adenoviridae Infections/immunology , Animals , Anthracyclines/therapeutic use , Anti-Bacterial Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/physiology , Autophagy-Related Protein 7 , Cecum/injuries , DNA Damage , Epirubicin/administration & dosage , Epirubicin/pharmacology , Epirubicin/therapeutic use , Fanconi Anemia Complementation Group D2 Protein/physiology , Inflammation , Inflammation Mediators/analysis , Injections, Intraperitoneal , Lung/metabolism , Meropenem , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/physiology , Organ Specificity , Peritonitis/etiology , Peritonitis/genetics , Peritonitis/immunology , Peritonitis/physiopathology , Respiratory Tract Infections/immunology , Shock, Septic/prevention & control , Thienamycins/therapeutic use , Whole-Body Irradiation
19.
Biochim Biophys Acta ; 1833(5): 1085-95, 2013 May.
Article En | MEDLINE | ID: mdl-23369735

TNFα can promote either cell survival or cell death. The activation of NF-κB plays a central role in cell survival while its inhibition makes TNFα-triggered cytotoxicity possible. Here, we report that the overexpression of a non-degradable mutant of the inhibitor of NF-κB (super-repressor (SR)-IκBα) sensitizes HeLa cells towards TNFα-induced apoptosis, involving caspases activation and cytocrome C release from the mitochondria. Interestingly, we describe that the specific knockdown of Bcl-xL, but not that of Bcl-2, Bcl-w or Mcl-1, renders cells sensitive to TNFα-induced apoptosis. This cytotoxic effect occurs without altering the activation of NF-κB. Then, the activation of the NF-κB pathway is not sufficient to protect Bcl-xL-downregulated cells from TNFα-induced cell death, meaning that TNFα is not able to promote cell survival in the absence of Bcl-xL. In addition, Bcl-xL silencing does not potentiate the cytotoxicity afforded by the cytokine in SR-IκBα-overexpressing cells. This indicates that TNFα-induced apoptosis in SR-IκBα-overexpressing cells relies on the protein levels of Bcl-xL. We have corroborated these findings using RD and DU-145 cells, which also become sensitive to TNFα-induced apoptosis after Bcl-xL knockdown despite that NF-κB remains activated. Altogether, our results point out that the impairment of the anti-apoptotic function of Bcl-xL should make cells sensitive towards external insults circumventing the TNFα-triggered NF-κB-mediated cytoprotective effect. Hence, the specific inhibition of Bcl-xL could be envisaged as a promising alternative strategy against NF-κB-dependent highly chemoresistant proliferative malignancies.


Apoptosis Regulatory Proteins , Proto-Oncogene Proteins c-bcl-2 , Tumor Necrosis Factor-alpha/metabolism , bcl-X Protein , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , HeLa Cells , Humans , I-kappa B Proteins/pharmacology , Mitochondria , Myeloid Cell Leukemia Sequence 1 Protein , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
20.
Cell Host Microbe ; 12(5): 693-704, 2012 Nov 15.
Article En | MEDLINE | ID: mdl-23159058

Disease tolerance is a defense strategy that limits the fitness costs of infection irrespectively of pathogen burden. While restricting iron (Fe) availability to pathogens is perceived as a host defense strategy, the resulting tissue Fe overload can be cytotoxic and promote tissue damage to exacerbate disease severity. Examining this interplay during malaria, the disease caused by Plasmodium infection, we find that expression of the Fe sequestering protein ferritin H chain (FtH) in mice, and ferritin in humans, is associated with reduced tissue damage irrespectively of pathogen burden. FtH protection relies on its ferroxidase activity, which prevents labile Fe from sustaining proapoptotic c-Jun N-terminal kinase (JNK) activation. FtH expression is inhibited by JNK activation, promoting tissue Fe overload, tissue damage, and malaria severity. Mimicking FtH's antioxidant effect or inhibiting JNK activation pharmacologically confers therapeutic tolerance to malaria in mice. Thus, FtH provides metabolic adaptation to tissue Fe overload, conferring tolerance to malaria.


Apoferritins/metabolism , Ferritins/metabolism , Iron Overload/metabolism , Malaria/metabolism , Malaria/parasitology , Plasmodium chabaudi/immunology , Plasmodium chabaudi/physiology , Animals , Antioxidants/metabolism , Cells, Cultured , Ceruloplasmin/metabolism , Cytoprotection , Enzyme Activation , Hepatocytes/metabolism , Host-Parasite Interactions , Humans , Iron/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Plasmodium berghei/physiology , Plasmodium vivax/physiology
...