Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Minerva Surg ; 79(2): 147-154, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38252400

ABSTRACT

BACKGROUND: Open Abdomen (OA) is gaining popularity in damage control surgery (DCS) but there is not an absolute prognostic score to identify patients that may benefit from it. Our study investigates the correlation between the clinical frailty scale score (CFSS) and postoperative morbidity and mortality in patients undergoing OA. METHODS: Patients ≥65 yo undergoing OA in two referral centres between 2015 and 2020 were included and stratified according to CFSS in non-frail (NF), frail (F) and highly-frail (HF). The primary endpoint was 30-day mortality. Secondary endpoints were postoperative morbidity and 1- year survival. RESULTS: One hundred and thirty-six patients were included: 35 NF (25.7%), 56 F (41.2%), 45 HF (33.1%). Average age 76.8. The 73.5% of cases were non-traumatic diseases with no difference in preoperative characteristics. 95 (71.4%) had one complication, 26 NF (74.3%), 34 F (63.2%), 35 HF (77.8%) (P=0.301) and 59.4% had a complication with a CD≥3, 57.1% NF, 56.6% F and 64.4 HF. The 30-day mortality was 32.4%, higher in HF (46.7%) and F (30.4%) compared to NF (17.1%, P=0.018). The Overall 1-year survival was 41% (SE ±4) with statistically significant difference between HF vs. NF and HF vs. F (P=0.009 and P=0.029, respectively). In the univariate analysis, the only significant prognostic factor impacting mortality was CFSS, with HF having an HR of 1.948 (95% CI 1.097-3.460, P=0.023). CONCLUSIONS: When OA is a surgical option, frail patients should not be precluded, while HF should be carefully evaluated. The CFSS might be a good prognostic score for patients that may safely benefit from OA.


Subject(s)
Abdominal Cavity , Frailty , Humans , Aged , Frailty/diagnosis , Retrospective Studies , Frail Elderly , Abdomen/surgery
2.
Sci Rep ; 7(1): 17388, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29234123

ABSTRACT

The host antiviral response involves the induction of interferons and proinflammatory cytokines, but also the activation of cell death pathways, including apoptosis, to limit viral replication and spreading. This host defense is strictly regulated to eliminate the infection while limiting tissue damage that is associated with virus pathogenesis. Post-translational modifications, most notably phosphorylation, are key regulators of the antiviral defense implying an important role of protein phosphatases. Here, we investigated the role of the dual-specificity phosphatase 1 (DUSP1) in the host defense against human respiratory syncytial virus (RSV), a pathogenic virus of the Pneumoviridae family, and Sendai virus (SeV), a model virus being developed as a vector for anti-RSV vaccine. We found that DUSP1 is upregulated before being subjected to proteasomal degradation. DUSP1 does not inhibit the antiviral response, but negatively regulates virus-induced JNK/p38 MAPK phosphorylation. Interaction with the JNK-interacting protein 1 scaffold protein prevents dephosphorylation of JNK by DUSP1, likely explaining that AP-1 activation and downstream cytokine production are protected from DUSP1 inhibition. Importantly, DUSP1 promotes SeV-induced apoptosis and suppresses cell migration in RSV-infected cells. Collectively, our data unveils a previously unrecognized selective role of DUSP1 in the regulation of tissue damage and repair during infections by RSV and SeV.


Subject(s)
Apoptosis , Cell Movement , Dual Specificity Phosphatase 1/metabolism , MAP Kinase Signaling System , Respiratory Syncytial Virus Infections/metabolism , Respirovirus Infections/metabolism , A549 Cells , Adaptor Proteins, Signal Transducing/metabolism , Dual Specificity Phosphatase 1/genetics , Gene Expression Regulation , Humans , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus, Human , Respirovirus Infections/genetics , Sendai virus
4.
Nat Immunol ; 17(11): 1291-1299, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27618553

ABSTRACT

Innate lymphoid cells (ILCs) have potent immunological functions in experimental conditions in mice, but their contributions to immunity in natural conditions in humans have remained unclear. We investigated the presence of ILCs in a cohort of patients with severe combined immunodeficiency (SCID). All ILC subsets were absent in patients with SCID who had mutation of the gene encoding the common γ-chain cytokine receptor subunit IL-2Rγ or the gene encoding the tyrosine kinase JAK3. T cell reconstitution was observed in patients with SCID after hematopoietic stem cell transplantation (HSCT), but the patients still had considerably fewer ILCs in the absence of myeloablation than did healthy control subjects, with the exception of rare cases of reconstitution of the ILC1 subset of ILCs. Notably, the ILC deficiencies observed were not associated with any particular susceptibility to disease, with follow-up extending from 7 years to 39 years after HSCT. We thus report here selective ILC deficiency in humans and show that ILCs might be dispensable in natural conditions, if T cells are present and B cell function is preserved.


Subject(s)
Immunity, Innate , Lymphocytes/immunology , Adolescent , Adult , Animals , Biomarkers , Child , Disease Models, Animal , Graft Survival , Hematopoietic Stem Cell Transplantation , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Interleukin Receptor Common gamma Subunit/deficiency , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Janus Kinase 3/deficiency , Lymphocyte Count , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes/metabolism , Lymphopenia/blood , Lymphopenia/etiology , Mice , Mice, Knockout , Phenotype , Severe Combined Immunodeficiency/blood , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/metabolism , Severe Combined Immunodeficiency/therapy , Skin/immunology , Skin/pathology
5.
Cancer Immunol Res ; 3(10): 1109-14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26438443

ABSTRACT

The world of lymphocytes has recently expanded. A group of cells, innate lymphoid cells (ILC), has been defined. It includes lymphoid cells that have been known for decades, such as natural killer (NK) cells and lymphoid tissue-inducer (LTi) cells. NK cells recognize a vast array of tumor cells, which they help to eliminate through cytotoxicity and the production of cytokines, such as IFNγ. Advances in our understanding of NK-cell biology have led to a growing interest in the clinical manipulation of these cells in cancer. The other ILCs are found mostly in the mucosae and mucosal-associated lymphoid tissues, where they rapidly initiate immune responses to pathogens without the need for specific sensitization. Here, we outline the basic features of ILCs and review the role of ILCs other than NK cells in cancer. Much of the role of these ILCs in cancer remains unknown, but several findings should lead to further efforts to dissect the contribution of different ILC subsets to the promotion, maintenance, or elimination of tumors at various anatomic sites. This will require the development of standardized reagents and protocols for monitoring the presence and function of ILCs in human blood and tissue samples.


Subject(s)
Immunity, Innate , Lymphocyte Subsets/immunology , Neoplasms/immunology , Animals , Humans , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Subsets/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Phenotype
6.
J Innate Immun ; 6(5): 650-62, 2014.
Article in English | MEDLINE | ID: mdl-24800889

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are the main cytosolic sensors of single-stranded RNA viruses, including paramyxoviruses, and are required to initiate a quick and robust innate antiviral response. Despite different ligand-binding properties, the consensus view is that RIG-I and MDA5 trigger common signal(s) to activate interferon regulatory factor 3 (IRF-3) and NF-κB, and downstream antiviral and proinflammatory cytokine expression. Here, we performed a thorough analysis of the temporal involvement of RIG-I and MDA5 in the regulation of IRF-3 during respiratory syncytial virus (RSV) infection. Based on specific RNA interference-mediated knockdown of RIG-I and MDA5 in A549 cells, we confirmed that RIG-I is critical for the initiation of IRF-3 phosphorylation, dimerization and downstream gene expression. On the other hand, our experiments yielded the first evidence that knockdown of MDA5 leads to early ubiquitination and proteasomal degradation of active IRF-3. Conversely, ectopic expression of MDA5 prolonged RIG-I-induced IRF-3 activation. Altogether, we provide novel mechanistic insight into the temporal involvement of RIG-I and MDA5 in the innate antiviral response. While RIG-I is essential for initial IRF-3 activation, engagement of induced MDA5 is essential to prevent early degradation of IRF-3, thereby sustaining IRF-3-dependent antiviral gene expression. MDA5 plays a similar role during Sendai virus infection suggesting that this model is not restricted to RSV amongst paramyxoviruses.


Subject(s)
DEAD-box RNA Helicases/metabolism , Epithelial Cells/immunology , Interferon Regulatory Factor-3/metabolism , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Epithelial Cells/virology , Gene Expression Regulation/genetics , Humans , Immunity, Innate/genetics , Interferon Regulatory Factor-3/genetics , Interferon-Induced Helicase, IFIH1 , Phosphorylation/genetics , Proteolysis , RNA, Small Interfering/genetics , Receptors, Immunologic , Signal Transduction/genetics , Ubiquitination/genetics
7.
Cell Host Microbe ; 12(4): 571-84, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-23084923

ABSTRACT

Type I interferons (IFNs) are central to antiviral defense, but how they orchestrate immune cell function is incompletely understood. We determined that IFNs produced during murine cytomegalovirus (MCMV) infection differentially affect dendritic cells (DCs) and natural killer (NK) cells. IFNs induce cell-intrinsic responses in DCs, activating antiproliferative, antiviral, and lymphocyte-activating gene networks, consistent with high activity of the transcription factor STAT1 in these cells. By comparison, NK cells exhibit lower STAT1 expression and reduced IFN responsiveness. Rather, IFNs indirectly affect NK cells by inducing IL-15, which activates the transcription factor E2F and stimulates genes promoting cell expansion. IFN cell-intrinsic responses are necessary in DCs, but not NK cells, for MCMV resistance. Thus, sensitivity to IFN-induced cytokines and differences in IFN receptor signaling program immune cells to mount distinct responses that promote viral control.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , Animals , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Herpesviridae Infections/immunology , Herpesviridae Infections/veterinary , Mice , Mice, Inbred C57BL , Models, Biological , Signal Transduction
8.
J Interferon Cytokine Res ; 32(9): 393-400, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22817838

ABSTRACT

The interferon regulatory factor (IRF)-3 transcription factor plays a central role in the capacity of the host to mount an efficient innate antiviral immune defense, mainly through the regulation of type I Interferon genes. A tight regulation of IRF-3 is crucial for an adapted intensity and duration of the response. Redox-dependent processes are now well known to regulate signaling cascades. Recent reports have revealed that signaling molecules upstream of IRF-3, including the mitochondrial antiviral-signalling protein (MAVS) and the TNF receptor associated factors (TRAFs) adaptors, are sensitive to redox regulation. In the present study, we assessed whether redox regulation of thiol residues contained in IRF-3, which are priviledged redox sensors, play a role in its regulation following Sendai virus infection, using a combination of mutation of Cysteine (Cys) residues into Alanine and thiols alkylation using N-ethyl maleimide. Alkylation of IRF-3 on Cys289 appears to destabilize IRF-3 dimer in vitro. However, a detailed analysis of IRF-3 phosphorylation, dimerization, nuclear accumulation, and induction of target gene promoter in vivo led us to conclude that IRF-3 specific, individual Cys residues redox status does not play an essential role in its activation in vivo.


Subject(s)
Cell Nucleus/metabolism , Interferon Regulatory Factor-3/metabolism , Protein Multimerization , Respirovirus Infections/metabolism , Sendai virus/metabolism , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Alkylation , Amino Acid Substitution , Animals , Cell Line, Transformed , Cell Nucleus/genetics , Cell Nucleus/immunology , Ethylmaleimide/chemistry , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Type I/biosynthesis , Interferon Type I/genetics , Interferon Type I/immunology , Mice , Mice, Knockout , Mutation, Missense , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Respirovirus Infections/genetics , Respirovirus Infections/immunology , Sendai virus/immunology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/immunology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
9.
Viruses ; 1(3): 383-419, 2009 12.
Article in English | MEDLINE | ID: mdl-21994554

ABSTRACT

Type-I interferons (IFN-I) are cytokines essential for vertebrate antiviral defense, including against herpesviruses. IFN-I have potent direct antiviral activities and also mediate a multiplicity of immunoregulatory functions, which can either promote or dampen antiviral adaptive immune responses. Plasmacytoid dendritic cells (pDCs) are the professional producers of IFN-I in response to many viruses, including all of the herpesviruses tested. There is strong evidence that pDCs could play a major role in the initial orchestration of both innate and adaptive antiviral immune responses. Depending on their activation pattern, pDC responses may be either protective or detrimental to the host. Here, we summarize and discuss current knowledge regarding pDC implication in the physiopathology of mouse and human herpesvirus infections, and we discuss how pDC functions could be manipulated in immunotherapeutic settings to promote health over disease.

10.
Expert Rev Anti Infect Ther ; 6(6): 867-85, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19053900

ABSTRACT

Following the discovery of innate immune receptors, the topics of innate immunity and its role in defense against infective agents have recently blossomed into very active research fields, after several decades of neglect. Among innate immune cells, natural killer (NK) cells are endowed with the unique ability to recognize and kill cells infected with a variety of pathogens, irrespective of prior sensitization to these microbes. NK cells have a number of other functions, including cytokine production and immunoregulatory activities. Major advances have recently been made in the understanding of the role of NK cells in the physiopathology of infectious diseases. The cellular and molecular mechanisms regulating the acquisition of effector functions by NK cells and their triggering upon pathogenic encounters are being unraveled. The possibility that the power of NK cells could be harnessed for the design of innovative treatments against infections is a major incentive for biologists to further explore NK cell subset complexity and to identify the ligands that activate NK cell receptors.


Subject(s)
Immunity, Innate/physiology , Killer Cells, Natural/immunology , Animals , Humans , Immunity, Innate/genetics , Immunotherapy , Infections/immunology , Infections/pathology , Killer Cells, Natural/physiology , Lymphocyte Activation/physiology , Mice
11.
J Immunol ; 180(9): 5799-803, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18424698

ABSTRACT

As initially demonstrated with murine cytomegalovirus (MCMV), plasmacytoid dendritic cells (pDCs) are the major source of IFN-alpha/beta in response to a variety of viruses in vivo. However, contradictory results have been obtained pertaining to the mechanisms promoting IFN-alpha/beta production by pDCs in response to MCMV. In this study we show that TLR7 and TLR9 exert redundant functions for IFN-alpha/beta, IL-12p40, and TNF-alpha production by pDCs in vivo during MCMV infection. In contrast, we confirm that systemic production of IL-12p70 strictly depends on TLR9. The combined loss of TLR7 and TLR9 recapitulates critical features of the phenotype of MyD88-deficient mice, including a dramatic decrease in systemic IFN-alpha/beta levels, an increase in viral load, and increased susceptibility to MCMV-induced mortality. This is the first demonstration of the implication of TLR7 in the recognition of a DNA virus.


Subject(s)
Herpesviridae Infections/immunology , Immunity, Innate , Membrane Glycoproteins/immunology , Muromegalovirus/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology , Animals , Dendritic Cells/immunology , Herpesviridae Infections/genetics , Immunity, Innate/genetics , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Viral Load
12.
Int Immunol ; 20(1): 45-56, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18000008

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are an important source of IFN-alpha/beta in response to a variety of viruses in vivo, including murine cytomegalovirus (MCMV). However, the respective contributions of various infected organs, and within these of pDCs, conventional dendritic cells and other cells, to the systemic production of IFN-alpha/beta or other innate cytokines during viral infections in vivo is largely unknown. Whether a specialization of pDC subsets in the production of different patterns of innate cytokines exists in vivo in response to a viral infection has not been investigated. Here, by analyzing for the first time directly ex vivo, at the single-cell level, the simultaneous production of up to three cytokines in pDCs isolated from MCMV-infected mice, we show that (i) pDCs are the quasi-exclusive source of IFN-alpha/beta, IL-12 and tumor necrosis factor (TNF)-alpha, early during MCMV infection, in two immunocompetent mouse lines and with two viral strains, (ii) pDC activation for IFN-alpha/beta production is organ specific and (iii) a significant proportion of pDCs simultaneously produce IFN-alpha/beta, TNF-alpha and IL-12, although TNF-alpha and IFN-alpha/beta appear more often co-expressed with one another than each of them with IL-12. Altogether, these results show a broad and non-redundant role of pDCs in early innate detection of, and defense against, viral infection. The data also show differences in the responsiveness of pDCs from different tissues and suggest distinct molecular requirements for pDC production of various cytokines. These observations must be taken into account when designing new antiviral vaccination strategies aimed at harnessing pDC responses.


Subject(s)
Cytokines/biosynthesis , Dendritic Cells/cytology , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Muromegalovirus/pathogenicity , Animals , Cytokines/genetics , Dendritic Cells/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Immunity, Innate , Interferon-alpha/genetics , Interferon-beta/genetics , Interleukin-12/biosynthesis , Interleukin-12/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/immunology , Organ Specificity , Specific Pathogen-Free Organisms , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
13.
PLoS Pathog ; 3(8): e123, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17722980

ABSTRACT

Understanding the mechanisms that help promote protective immune responses to pathogens is a major challenge in biomedical research and an important goal for the design of innovative therapeutic or vaccination strategies. While natural killer (NK) cells can directly contribute to the control of viral replication, whether, and how, they may help orchestrate global antiviral defense is largely unknown. To address this question, we took advantage of the well-defined molecular interactions involved in the recognition of mouse cytomegalovirus (MCMV) by NK cells. By using congenic or mutant mice and wild-type versus genetically engineered viruses, we examined the consequences on antiviral CD8 T cell responses of specific defects in the ability of the NK cells to control MCMV. This system allowed us to demonstrate, to our knowledge for the first time, that NK cells accelerate CD8 T cell responses against a viral infection in vivo. Moreover, we identify the underlying mechanism as the ability of NK cells to limit IFN-alpha/beta production to levels not immunosuppressive to the host. This is achieved through the early control of cytomegalovirus, which dramatically reduces the activation of plasmacytoid dendritic cells (pDCs) for cytokine production, preserves the conventional dendritic cell (cDC) compartment, and accelerates antiviral CD8 T cell responses. Conversely, exogenous IFN-alpha administration in resistant animals ablates cDCs and delays CD8 T cell activation in the face of NK cell control of viral replication. Collectively, our data demonstrate that the ability of NK cells to respond very early to cytomegalovirus infection critically contributes to balance the intensity of other innate immune responses, which dampens early immunopathology and promotes optimal initiation of antiviral CD8 T cell responses. Thus, the extent to which NK cell responses benefit the host goes beyond their direct antiviral effects and extends to the prevention of innate cytokine shock and to the promotion of adaptive immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cytotoxicity, Immunologic , Dendritic Cells/cytology , Dendritic Cells/immunology , Disease Models, Animal , Herpesviridae Infections/metabolism , Immunity, Innate , Interferon-alpha/metabolism , Interferon-beta/metabolism , Killer Cells, Natural/cytology , Lymphocyte Activation , Mice , Mice, Congenic , Mice, Inbred BALB C , Specific Pathogen-Free Organisms , Spleen/cytology , Spleen/virology , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...