Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nat Rev Mol Cell Biol ; 19(9): 594-610, 2018 09.
Article in English | MEDLINE | ID: mdl-29858605

ABSTRACT

Adult tissue stem cells have a pivotal role in tissue maintenance and regeneration throughout the lifespan of multicellular organisms. Loss of tissue homeostasis during post-reproductive lifespan is caused, at least in part, by a decline in stem cell function and is associated with an increased incidence of diseases. Hallmarks of ageing include the accumulation of molecular damage, failure of quality control systems, metabolic changes and alterations in epigenome stability. In this Review, we discuss recent evidence in support of a novel concept whereby cell-intrinsic damage that accumulates during ageing and cell-extrinsic changes in ageing stem cell niches and the blood result in modifications of the stem cell epigenome. These cumulative epigenetic alterations in stem cells might be the cause of the deregulation of developmental pathways seen during ageing. In turn, they could confer a selective advantage to mutant and epigenetically drifted stem cells with altered self-renewal and functions, which contribute to the development of ageing-associated organ dysfunction and disease.


Subject(s)
Cellular Senescence/genetics , Cellular Senescence/physiology , Epigenesis, Genetic/genetics , Stem Cells/physiology , Animals , Epigenomics/methods , Homeostasis/genetics , Homeostasis/physiology , Humans
2.
Cell ; 133(2): 235-49, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18423196

ABSTRACT

Multiple lung pathogens such as chemical agents, H5N1 avian flu, or SARS cause high lethality due to acute respiratory distress syndrome. Here we report that Toll-like receptor 4 (TLR4) mutant mice display natural resistance to acid-induced acute lung injury (ALI). We show that TLR4-TRIF-TRAF6 signaling is a key disease pathway that controls the severity of ALI. The oxidized phospholipid (OxPL) OxPAPC was identified to induce lung injury and cytokine production by lung macrophages via TLR4-TRIF. We observed OxPL production in the lungs of humans and animals infected with SARS, Anthrax, or H5N1. Pulmonary challenge with an inactivated H5N1 avian influenza virus rapidly induces ALI and OxPL formation in mice. Loss of TLR4 or TRIF expression protects mice from H5N1-induced ALI. Moreover, deletion of ncf1, which controls ROS production, improves the severity of H5N1-mediated ALI. Our data identify oxidative stress and innate immunity as key lung injury pathways that control the severity of ALI.


Subject(s)
Oxidative Stress , Respiratory Distress Syndrome/metabolism , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Humans , Influenza, Human/metabolism , Interleukin-6/metabolism , Lung , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Orthomyxoviridae Infections/metabolism , Phospholipids/metabolism , Severe Acute Respiratory Syndrome/metabolism , Signal Transduction
3.
Int J Mol Sci ; 22(24)2021 Dec 13.
Article in English | MEDLINE | ID: mdl-34948198

ABSTRACT

Green rods (GRs) represent a unique type of photoreceptor to be found in the retinas of anuran amphibians. These cells harbor a cone-specific blue-sensitive visual pigment but exhibit morphology of the outer segment typical for classic red rods (RRs), which makes them a perspective model object for studying cone-rod transmutation. In the present study, we performed detailed electrophysiological examination of the light sensitivity, response kinetics and parameters of discrete and continuous dark noise in GRs of the two anuran species: cane toad and marsh frog. Our results confirm that anuran GRs are highly specialized nocturnal vision receptors. Moreover, their rate of phototransduction quenching appeared to be about two-times slower than in RRs, which makes them even more efficient single photon detectors. The operating intensity ranges for two rod types widely overlap supposedly allowing amphibians to discriminate colors in the scotopic region. Unexpectedly for typical cone pigments but in line with some previous reports, the spontaneous isomerization rate of the GR visual pigment was found to be the same as for rhodopsin of RRs. Thus, our results expand the knowledge on anuran GRs and show that these are even more specialized single photon catchers than RRs, which allows us to assign them a status of "super-rods".


Subject(s)
Light Signal Transduction/physiology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Anura/anatomy & histology , Isomerism , Kinetics , Light , Night Vision/physiology , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/physiology , Retina/anatomy & histology , Retina/metabolism , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Rhodopsin , Rod Opsins , Vision, Ocular/physiology
4.
Nat Immunol ; 9(9): 1037-46, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18641654

ABSTRACT

Tumor necrosis factor receptor 1 (TNFR1) and Toll-like receptors (TLRs) regulate immune and inflammatory responses. Here we show that the TNFR1-associated death domain protein (TRADD) is critical in TNFR1, TLR3 and TLR4 signaling. TRADD deficiency abrogated TNF-induced apoptosis, prevented recruitment of the ubiquitin ligase TRAF2 and ubiquitination of the adaptor RIP1 in the TNFR1 signaling complex, and considerably inhibited but did not completely abolish activation of the transcription factor NF-kappaB and mitogen-activated protein kinases 'downstream' of TNFR1. TRIF-dependent cytokine production induced by the synthetic double-stranded RNA poly(I:C) and lipopolysaccharide was lower in TRADD-deficient mice than in wild-type mice. Moreover, TRADD deficiency inhibited poly(I:C)-mediated RIP1 ubiquitination and activation of NF-kappaB and mitogen-activated protein kinase signaling in fibroblasts but not in bone marrow macrophages. Thus, TRADD is an essential component of TNFR1 signaling and has a critical but apparently cell type-specific function in TRIF-dependent TLR responses.


Subject(s)
Signal Transduction , TNF Receptor-Associated Death Domain Protein/deficiency , TNF Receptor-Associated Factor 1/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/physiology , Animals , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Death Domain Protein/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Ubiquitin/metabolism
5.
Immunity ; 35(4): 572-82, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22000287

ABSTRACT

Epidermal keratinocytes provide an essential structural and immunological barrier forming the first line of defense against potentially pathogenic microorganisms. Mechanisms regulating barrier integrity and innate immune responses in the epidermis are important for the maintenance of skin immune homeostasis and the pathogenesis of inflammatory skin diseases. Here, we show that epidermal keratinocyte-restricted deficiency of the adaptor protein FADD (FADD(E-KO)) induced severe inflammatory skin lesions in mice. The development of skin inflammation in FADD(E-KO) mice was triggered by RIP kinase 3 (RIP3)-mediated programmed necrosis (termed necroptosis) of FADD-deficient keratinocytes, which was partly dependent on the deubiquitinating enzyme CYLD and tumor necrosis factor (TNF)-TNF receptor 1 signaling. Collectively, our findings provide an in vivo experimental paradigm that regulation of necroptosis in keratinocytes is important for the maintenance of immune homeostasis and the prevention of chronic inflammation in the skin.


Subject(s)
Apoptosis , Dermatitis/immunology , Dermatitis/pathology , Epidermis/immunology , Fas-Associated Death Domain Protein/immunology , Keratinocytes/immunology , Animals , Cells, Cultured , Dermatitis/metabolism , Fas-Associated Death Domain Protein/deficiency , Homeostasis , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Necrosis , Signal Transduction
6.
J Biol Inorg Chem ; 24(8): 1285-1303, 2019 12.
Article in English | MEDLINE | ID: mdl-31773268

ABSTRACT

Alzheimer's disease (AD) is the main cause of dementia in elderly. Increasing life expectancy is behind the growing prevalence of AD worldwide with approximately 45 million cases currently documented and projection studies suggesting a triplication of this number by 2050. Mexico does not have an accurate AD registry, but 860,000 cases were reported in 2014 and the prediction reaches 3.5 million cases by 2050. Amyloid plaques and neurofibrillary tangles represent the main hallmarks of AD, being constituted of amyloid beta (Aß) peptide and phosphorylated tau, respectively. The risk factors for AD include genetic mutations, lifestyle and environmental pollution. Particularly, lead (Pb) has attracted attention due to its ability to target multiple pathways involved in the pathophysiology of AD. Although the epidemiological data are limiting, animal and in vitro studies show growing evidence of causal effects of Pb exposure on AD-linked features including Aß aggregation and tau phosphorylation. Interestingly, many Pb effects occur selectively following early-life exposure to the metal, suggesting an epigenetic mechanism. This hypothesis is supported by changes in DNA methylation and microRNA expression patterns inflicted by early-life Pb exposure. Pb pollution in Mexico represents a significant problem because past and current mining activities, historical use of Pb as fuel additive and culturally rooted use of Pb in glazed ceramics, contribute to high levels of Pb pollution in Mexico. In this review we will discuss potential risks of AD development in Mexican populations chronically exposed to Pb in their childhood.


Subject(s)
Alzheimer Disease/etiology , Child Development/drug effects , Environmental Exposure/adverse effects , Lead/toxicity , Alzheimer Disease/genetics , Amyloid beta-Peptides/drug effects , Animals , Child , DNA/metabolism , DNA Methylation/drug effects , Environmental Pollutants/toxicity , Epigenesis, Genetic/drug effects , Humans , Mexico , Risk Factors , tau Proteins/drug effects
7.
Nature ; 501(7467): 416-20, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23975097

ABSTRACT

DNA damage responses have been well characterized with regard to their cell-autonomous checkpoint functions leading to cell cycle arrest, senescence and apoptosis. In contrast, systemic responses to tissue-specific genome instability remain poorly understood. In adult Caenorhabditis elegans worms germ cells undergo mitotic and meiotic cell divisions, whereas somatic tissues are entirely post-mitotic. Consequently, DNA damage checkpoints function specifically in the germ line, whereas somatic tissues in adult C. elegans are highly radio-resistant. Some DNA repair systems such as global-genome nucleotide excision repair (GG-NER) remove lesions specifically in germ cells. Here we investigated how genome instability in germ cells affects somatic tissues in C. elegans. We show that exogenous and endogenous DNA damage in germ cells evokes elevated resistance to heat and oxidative stress. The somatic stress resistance is mediated by the ERK MAP kinase MPK-1 in germ cells that triggers the induction of putative secreted peptides associated with innate immunity. The innate immune response leads to activation of the ubiquitin-proteasome system (UPS) in somatic tissues, which confers enhanced proteostasis and systemic stress resistance. We propose that elevated systemic stress resistance promotes endurance of somatic tissues to allow delay of progeny production when germ cells are genomically compromised.


Subject(s)
Adaptation, Physiological/physiology , Caenorhabditis elegans/physiology , DNA Damage , Germ Cells/immunology , Germ Cells/metabolism , Immunity, Innate , Stress, Physiological/immunology , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/immunology , Caenorhabditis elegans Proteins/metabolism , DNA Damage/genetics , Enzyme Activation , Genomic Instability/genetics , Germ Cells/enzymology , Hot Temperature , Immunity, Innate/genetics , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Oxidative Stress , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism
8.
BMC Biol ; 16(1): 82, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30068331

ABSTRACT

BACKGROUND: Mammals display a wide range of variation in their lifespan. Investigating the molecular networks that distinguish long- from short-lived species has proven useful to identify determinants of longevity. Here, we compared the livers of young and old long-lived naked mole-rats (NMRs) and the phylogenetically closely related, shorter-lived, guinea pigs using an integrated omics approach. RESULTS: We found that NMR livers display a unique expression pattern of mitochondrial proteins that results in distinct metabolic features of their mitochondria. For instance, we observed a generally reduced respiration rate associated with lower protein levels of respiratory chain components, particularly complex I, and increased capacity to utilize fatty acids. Interestingly, we show that the same molecular networks are affected during aging in both NMRs and humans, supporting a direct link to the extraordinary longevity of both species. Finally, we identified a novel detoxification pathway linked to longevity and validated it experimentally in the nematode Caenorhabditis elegans. CONCLUSIONS: Our work demonstrates the benefits of integrating proteomic and transcriptomic data to perform cross-species comparisons of longevity-associated networks. Using a multispecies approach, we show at the molecular level that livers of NMRs display progressive age-dependent changes that recapitulate typical signatures of aging despite the negligible senescence and extraordinary longevity of these rodents.


Subject(s)
Aging , Liver/metabolism , Longevity , Mole Rats/physiology , Proteome , Adult , Aged , Aged, 80 and over , Animals , Caenorhabditis elegans/physiology , Guinea Pigs , Humans , Male , Middle Aged , Species Specificity
9.
Semin Immunol ; 26(4): 303-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24856329

ABSTRACT

The nematode worm Caenorhabditis elegans comprises an ancestral immune system. C. elegans recognizes and responds to viral, bacterial, and fungal infections. Components of the RNA interference machinery respond to viral infection, while highly conserved MAPK signaling pathways activate the innate immune response to bacterial infection. C. elegans has been particularly important for exploring the role of innate immunity in organismal stress resistance and the regulation of longevity. Also functions of neuronal sensing of infectious bacteria have recently been uncovered. Studies on nematode immunity can be instructive in exploring innate immune signaling in the absence of specialized immune cells and adaptive immunity.


Subject(s)
Caenorhabditis elegans/immunology , Immunity, Innate , Models, Animal , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans Proteins/metabolism , Humans , Regeneration
10.
Trends Genet ; 30(3): 95-102, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24439457

ABSTRACT

DNA damage checkpoints are important tumor-suppressor mechanisms that halt cell cycle progression to allow time for DNA repair, or induce senescence and apoptosis to remove damaged cells permanently. Non-cell-autonomous DNA damage responses activate the innate immune system in multiple metazoan species. These responses not only enable clearance of damaged cells and contribute to tissue remodeling and regeneration but can also result in chronic inflammation and tissue damage. Germline DNA damage-induced systemic stress resistance (GDISR) is mediated by an ancestral innate immune response and results in organismal adjustments to the presence of damaged cells. We discuss GDISR as an organismal DNA damage checkpoint mechanism through which elevated somatic endurance can extend reproductive lifespan when germ cells require extended time for restoring genome stability.


Subject(s)
Adaptation, Physiological/genetics , DNA Damage , Genomic Instability/genetics , Animals , Humans , Inflammation/genetics , Inflammation/pathology , Neoplasms/genetics , Neoplasms/pathology , Regeneration/genetics
11.
Immunity ; 28(5): 651-61, 2008 May.
Article in English | MEDLINE | ID: mdl-18439848

ABSTRACT

Upon detection of viral RNA, the helicases RIG-I and/or MDA5 trigger, via their adaptor Cardif (also known as IPS-1, MAVS, or VISA), the activation of the transcription factors NF-kappaB and IRF3, which collaborate to induce an antiviral type I interferon (IFN) response. FADD and RIP1, known as mediators of death-receptor signaling, are implicated in this antiviral pathway; however, the link between death-receptor and antiviral signaling is not known. Here we showed that TRADD, a crucial adaptor of tumor necrosis factor receptor (TNFRI), was important in RIG-like helicase (RLH)-mediated signal transduction. TRADD is recruited to Cardif and orchestrated complex formation with the E3 ubiquitin ligase TRAF3 and TANK and with FADD and RIP1, leading to the activation of IRF3 and NF-kappaB. Loss of TRADD prevented Cardif-dependent activation of IFN-beta, reduced the production of IFN-beta in response to RNA viruses, and enhanced vesicular stomatitis virus replication. Thus, TRADD is not only an essential component of proinflammatory TNFRI signaling, but is also required for RLH-Cardif-dependent antiviral immune responses.


Subject(s)
DNA Helicases/metabolism , Interferon Regulatory Factor-3/metabolism , Rhabdoviridae Infections/immunology , TNF Receptor-Associated Death Domain Protein/metabolism , Vesiculovirus/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Fas-Associated Death Domain Protein/metabolism , GTPase-Activating Proteins/metabolism , Humans , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface , Rhabdoviridae Infections/virology , Signal Transduction , TNF Receptor-Associated Factor 3/immunology , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/metabolism , Vesiculovirus/physiology
12.
Nature ; 477(7364): 330-4, 2011 Jul 31.
Article in English | MEDLINE | ID: mdl-21804564

ABSTRACT

Intestinal immune homeostasis depends on a tightly regulated cross talk between commensal bacteria, mucosal immune cells and intestinal epithelial cells (IECs). Epithelial barrier disruption is considered to be a potential cause of inflammatory bowel disease; however, the mechanisms regulating intestinal epithelial integrity are poorly understood. Here we show that mice with IEC-specific knockout of FADD (FADD(IEC-KO)), an adaptor protein required for death-receptor-induced apoptosis, spontaneously developed epithelial cell necrosis, loss of Paneth cells, enteritis and severe erosive colitis. Genetic deficiency in RIP3, a critical regulator of programmed necrosis, prevented the development of spontaneous pathology in both the small intestine and colon of FADD(IEC-KO) mice, demonstrating that intestinal inflammation is triggered by RIP3-dependent death of FADD-deficient IECs. Epithelial-specific inhibition of CYLD, a deubiquitinase that regulates cellular necrosis, prevented colitis development in FADD(IEC-KO) but not in NEMO(IEC-KO) mice, showing that different mechanisms mediated death of colonic epithelial cells in these two models. In FADD(IEC-KO) mice, TNF deficiency ameliorated colon inflammation, whereas MYD88 deficiency and also elimination of the microbiota prevented colon inflammation, indicating that bacteria-mediated Toll-like-receptor signalling drives colitis by inducing the expression of TNF and other cytokines. However, neither CYLD, TNF or MYD88 deficiency nor elimination of the microbiota could prevent Paneth cell loss and enteritis in FADD(IEC-KO) mice, showing that different mechanisms drive RIP3-dependent necrosis of FADD-deficient IECs in the small and large bowel. Therefore, by inhibiting RIP3-mediated IEC necrosis, FADD preserves epithelial barrier integrity and antibacterial defence, maintains homeostasis and prevents chronic intestinal inflammation. Collectively, these results show that mechanisms preventing RIP3-mediated epithelial cell death are critical for the maintenance of intestinal homeostasis and indicate that programmed necrosis of IECs might be implicated in the pathogenesis of inflammatory bowel disease, in which Paneth cell and barrier defects are thought to contribute to intestinal inflammation.


Subject(s)
Colitis/pathology , Colon/pathology , Enteritis/pathology , Epithelial Cells/pathology , Fas-Associated Death Domain Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Chronic Disease , Colitis/enzymology , Colitis/metabolism , Colon/enzymology , Colon/metabolism , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzyme CYLD , Enteritis/enzymology , Enteritis/metabolism , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Fas-Associated Death Domain Protein/deficiency , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/metabolism , Metagenome/physiology , Mice , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/metabolism , Necrosis , Paneth Cells/pathology , Signal Transduction , Tumor Necrosis Factors/deficiency
13.
Trends Mol Med ; 2024 May 13.
Article in English | MEDLINE | ID: mdl-38744580

ABSTRACT

Hormesis is a phenomenon whereby low-level stress can improve cellular, organ, or organismal fitness in response to a subsequent similar or other stress insult. Whereas hormesis is thought to contribute to the fitness benefits arising from symbiotic host-microbe interactions, the putative benefits of hormesis in host-pathogen interactions have yet to be explored. Hormetic responses have nonetheless been reported in experimental models of infection, a common feature of which is regulation of host mitochondrial function. We propose that these mitohormetic responses could be harnessed therapeutically to limit the severity of infectious diseases.

14.
Nat Commun ; 15(1): 1702, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38402241

ABSTRACT

Ribosome biogenesis is initiated by RNA polymerase I (Pol I)-mediated synthesis of pre-ribosomal RNA (pre-rRNA). Pol I activity was previously linked to longevity, but the underlying mechanisms were not studied beyond effects on nucleolar structure and protein translation. Here we use multi-omics and functional tests to show that curtailment of Pol I activity remodels the lipidome and preserves mitochondrial function to promote longevity in Caenorhabditis elegans. Reduced pre-rRNA synthesis improves energy homeostasis and metabolic plasticity also in human primary cells. Conversely, the enhancement of pre-rRNA synthesis boosts growth and neuromuscular performance of young nematodes at the cost of accelerated metabolic decline, mitochondrial stress and premature aging. Moreover, restriction of Pol I activity extends lifespan more potently than direct repression of protein synthesis, and confers geroprotection even when initiated late in life, showcasing this intervention as an effective longevity and metabolic health treatment not limited by aging.


Subject(s)
Caenorhabditis elegans Proteins , Longevity , Animals , Humans , Longevity/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , RNA Precursors/metabolism , Aging/genetics
15.
Front Oncol ; 11: 687371, 2021.
Article in English | MEDLINE | ID: mdl-34408980

ABSTRACT

Hepatocellular carcinoma (HCC) remains a devastating malignancy worldwide due to lack of effective therapy. The immune-rich contexture of HCC tumor microenvironment (TME) makes this tumor an appealing target for immune-based therapies; however, the immunosuppressive TME is still a major challenge for more efficient immunotherapy in HCC. Using bioinformatics analysis based on the TCGA database, here we found that MAPK10 is frequently down-regulated in HCC tumors and significantly correlates with poor survival of HCC patients. HCC patients with low MAPK10 expression have lower expression scores of tumor infiltration lymphocytes (TILs) and stromal cells in the TME and increased scores of tumor cells than those with high MAPK10 expression. Further transcriptomic analyses revealed that the immune activity in the TME of HCC was markedly reduced in the low-MAPK10 group of HCC patients compared to the high-MAPK10 group. Additionally, we identified 495 differentially expressed immune-associated genes (DIGs), with 482 genes down-regulated and 13 genes up-regulated in parallel with the decrease of MAPK10 expression. GO enrichment and KEGG pathway analyses indicated that the biological functions of these DIGs included cell chemotaxis, leukocyte migration and positive regulation of the response to cytokine-cytokine receptor interaction, T cell receptor activation and MAPK signaling pathway. Protein-protein interaction (PPI) analyses of the 495 DIGs revealed five potential downstream hub genes of MAPK10, including SYK, CBL, VAV1, LCK, and CD3G. Several hub genes such as SYK, LCK, and VAV1 could respond to the immunological costimulatory signaling mediated by the transmembrane protein ICAM1, which was identified as a down-regulated DIG associated with low-MAPK10 expression. Moreover, ectopic overexpression or knock-down of MAPK10 could up-regulate or down-regulate ICAM1 expression via phosphorylation of c-jun at Ser63 in HCC cell lines, respectively. Collectively, our results demonstrated that MAPK10 down-regulation likely contributes to the immunosuppressive TME of HCC, and this gene might serve as a potential immunotherapeutic target and a prognostic factor for HCC patients.

16.
Gut Microbes ; 13(1): 1993598, 2021.
Article in English | MEDLINE | ID: mdl-34793277

ABSTRACT

Antibiotics are commonly used in the Intensive Care Unit (ICU); however, several studies showed that the impact of antibiotics to prevent infection, multi-organ failure, and death in the ICU is less clear than their benefit on course of infection in the absence of organ dysfunction. We characterized here the compositional and metabolic changes of the gut microbiome induced by critical illness and antibiotics in a cohort of 75 individuals in conjunction with 2,180 gut microbiome samples representing 16 different diseases. We revealed an "infection-vulnerable" gut microbiome environment present only in critically ill treated with antibiotics (ICU+). Feeding of Caenorhabditis elegans with Bifidobacterium animalis and Lactobacillus crispatus, species that expanded in ICU+ patients, revealed a significant negative impact of these microbes on host viability and developmental homeostasis. These results suggest that antibiotic administration can dramatically impact essential functional activities in the gut related to immune responses more than critical illness itself, which might explain in part untoward effects of antibiotics in the critically ill.


Subject(s)
Anti-Bacterial Agents/adverse effects , Critical Illness , Gastrointestinal Microbiome/drug effects , Metabolome/drug effects , Animals , Bacteria/classification , Bacteria/drug effects , Bacteria/metabolism , Bacteria/pathogenicity , Bile Acids and Salts/metabolism , Candida/classification , Candida/drug effects , Candida/metabolism , Candida/pathogenicity , Drug Resistance, Fungal/drug effects , Fatty Acids, Volatile/metabolism , Humans , Infections/microbiology , Intensive Care Units , Moths
17.
Genomics Proteomics Bioinformatics ; 18(4): 430-442, 2020 08.
Article in English | MEDLINE | ID: mdl-33309863

ABSTRACT

Aging is a complex process that can be characterized by functional and cognitive decline in an individual. Aging can be assessed based on the functional capacity of vital organs and their intricate interactions with one another. Thus, the nature of aging can be described by focusing on a specific organ and an individual itself. However, to fully understand the complexity of aging, one must investigate not only a single tissue or biological process but also its complex interplay and interdependencies with other biological processes. Here, using RNA-seq, we monitored changes in the transcriptome during aging in four tissues (including brain, blood, skin and liver) in mice at 9 months, 15 months, and 24 months, with a final evaluation at the very old age of 30 months. We identified several genes and processes that were differentially regulated during aging in both tissue-dependent and tissue-independent manners. Most importantly, we found that the electron transport chain (ETC) of mitochondria was similarly affected at the transcriptome level in the four tissues during the aging process. We also identified the liver as the tissue showing the largest variety of differentially expressed genes (DEGs) over time. Lcn2 (Lipocalin-2) was found to be similarly regulated among all tissues, and its effect on longevity and survival was validated using its orthologue in Caenorhabditis elegans. Our study demonstrated that the molecular processes of aging are relatively subtle in their progress, and the aging process of every tissue depends on the tissue's specialized function and environment. Hence, individual gene or process alone cannot be described as the key of aging in the whole organism.


Subject(s)
Aging , Longevity , Aging/genetics , Animals , Caenorhabditis elegans/genetics , Longevity/genetics , Mice , Mitochondria/genetics , Transcriptome
18.
Nat Metab ; 2(11): 1316-1331, 2020 11.
Article in English | MEDLINE | ID: mdl-33139960

ABSTRACT

Current clinical trials are testing the life-extending benefits of the diabetes drug metformin in healthy individuals without diabetes. However, the metabolic response of a non-diabetic cohort to metformin treatment has not been studied. Here, we show in C. elegans and human primary cells that metformin shortens lifespan when provided in late life, contrary to its positive effects in young organisms. We find that metformin exacerbates ageing-associated mitochondrial dysfunction, causing respiratory failure. Age-related failure to induce glycolysis and activate the dietary-restriction-like mobilization of lipid reserves in response to metformin result in lethal ATP exhaustion in metformin-treated aged worms and late-passage human cells, which can be rescued by ectopic stabilization of cellular ATP content. Metformin toxicity is alleviated in worms harbouring disruptions in insulin-receptor signalling, which show enhanced resilience to mitochondrial distortions at old age. Together, our data show that metformin induces deleterious changes of conserved metabolic pathways in late life, which could bring into question its benefits for older individuals without diabetes.


Subject(s)
Aging , Caenorhabditis elegans , Hypoglycemic Agents/toxicity , Metabolism/drug effects , Metformin/toxicity , Adenosine Triphosphate/metabolism , Animals , Caloric Restriction , Glycolysis , Humans , Life Expectancy , Lipid Metabolism , Microbiota , Mitochondrial Diseases/metabolism , Primary Cell Culture , Receptor, Insulin/metabolism , Signal Transduction
19.
Nat Metab ; 4(4): 407-409, 2022 04.
Article in English | MEDLINE | ID: mdl-35388211
20.
Mech Ageing Dev ; 165(Pt A): 47-53, 2017 07.
Article in English | MEDLINE | ID: mdl-27317629

ABSTRACT

The impact of DNA damage-induced immune responses on aging and disease development is a topic of growing scientific interest and debate. While abundant data links persistent genotoxic stress and associated inflammatory activity to organ decline and cancer development, evidence of pro-homeostatic nature of immune responses triggered by transient DNA damage gradually accumulates. Current review focuses on comparing systemic outcomes of transient genotoxicity with effects of persistent DNA damage from the angle of associated immune activity. We discuss genotoxic stress as a potential damage associated molecular pattern (DAMP) which alerts the organism of the upcoming systemic dysfunction and pre-conditions the body for damage tolerance and repair.


Subject(s)
DNA Damage/immunology , DNA Repair/immunology , Neoplasms/immunology , Animals , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Neoplasms/genetics , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL