ABSTRACT
Sepsis is characterized by a dysregulated inflammatory response to infection. Despite studies in mice, the cellular and molecular basis of human sepsis remains unclear and effective therapies are lacking. Blood monocytes serve as the first line of host defense and are equipped to recognize and respond to infection by triggering an immune-inflammatory response. However, the response of these cells in human sepsis and their contribution to sepsis pathogenesis is poorly understood. To investigate this, we performed a transcriptomic, functional, and mechanistic analysis of blood monocytes from patients during sepsis and after recovery. Our results revealed the functional plasticity of monocytes during human sepsis, wherein they transited from a pro-inflammatory to an immunosuppressive phenotype, while enhancing protective functions like phagocytosis, anti-microbial activity, and tissue remodeling. Mechanistically, hypoxia inducible factor-1α (HIF1α) mediated this functional re-programming of monocytes, revealing a potential mechanism for their therapeutic targeting to regulate human sepsis.
Subject(s)
Cellular Reprogramming/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immunocompromised Host , Interleukin-1 Receptor-Associated Kinases/immunology , Sepsis/immunology , Adaptive Immunity , Convalescence , Cytokines/genetics , Cytokines/immunology , Gene Expression Profiling , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunity, Innate , Interleukin-1 Receptor-Associated Kinases/genetics , Monocytes/immunology , Monocytes/pathology , Phagocytosis , Sepsis/genetics , Sepsis/pathology , Signal Transduction , Transcriptome/immunologyABSTRACT
Patients with acute ischemic stroke (AIS) suffer from infections associated with mortality. The relevance of the innate immune system, and monocytes in particular, has emerged as an important factor in the evolution of these infections. The study enrolled 14 patients with AIS, without previous treatment, and 10 healthy controls. In the present study, we show that monocytes from patients with AIS exhibit a refractory state or endotoxin tolerance. The patients were unable to orchestrate an inflammatory response against LPS and expressed three factors reported to control the evolution of human monocytes into a refractory state: IL-1R-associated kinase-M, NFkB2/p100, and hypoxia-inducible factor-1α. The levels of circulating mitochondrial DNA (mtDNA) in patients with AIS correlated with impaired inflammatory response of isolated monocytes. Interestingly, the patients could be classified into two groups: those who were infected and those who were not, according to circulating mtDNA levels. This finding was validated in an independent cohort of 23 patients with AIS. Additionally, monocytes from healthy controls, cultured in the presence of both sera from patients and mtDNA, reproduced a refractory state after endotoxin challenge. This effect was negated by either a TLR9 antagonist or DNase treatment. The present data further extend our understanding of endotoxin tolerance implications in AIS. A putative role of mtDNA as a new biomarker of stroke-associated infections, and thus a clinical target for preventing poststroke infection, has also been identified.
Subject(s)
Biomarkers/blood , Blood Cells/immunology , DNA, Mitochondrial/blood , Infections/immunology , Ischemia/immunology , Monocytes/immunology , Stroke/immunology , Acute Disease , Aged , Aged, 80 and over , Cells, Cultured , Endotoxins/immunology , Female , Humans , Immune Tolerance , Immunity, Innate , Infections/etiology , Ischemia/complications , Male , Middle Aged , Stroke/complicationsABSTRACT
Sepsis, among other pathologies, is an endotoxin tolerance (ET)-related disease. On admission, we classified 48 patients with sepsis into 3 subgroups according to the ex vivo response to lipopolysaccharide. This response correlates with the Acute Physiology and Chronic Health Evaluation (APACHE) II score and the ET degree. Moreover, the ET-related classification determines the outcome of these patients. Programmed cell death-ligand 1 (PD-L1) expression on septic monocytes is also linked with ET status. In addition to the regulation of cytokine production, one of the hallmarks of ET that significantly affects patients with sepsis is T-cell proliferation impairment or a poor switch to the adaptive response. PD-L1/programmed cell death-1 (PD-1) blocking and knockdown assays on tolerant monocytes from both patients with sepsis and the in vitro model reverted the impaired adaptive response. Mechanistically, the transcription factor hypoxia-inducible factor-1α (HIF1α) has been translocated into the nucleus and drives PD-L1 expression during ET in human monocytes. This fact, together with patient classification according to the ex vivo lipopolysaccharide response, opens an interesting field of study and potential personalized clinical applications, not only for sepsis but also for all ET-associated pathologies.
Subject(s)
Adaptive Immunity , B7-H1 Antigen/biosynthesis , Endotoxins/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immune Tolerance , Sepsis/pathology , APACHE , Adult , Aged , Aged, 80 and over , Cells, Cultured , Female , Humans , Male , Middle Aged , Monocytes/immunologyABSTRACT
BACKGROUND: The analysis of tumour-infiltrating immune cells within patients' tumour samples in colorectal cancer (CRC) has become an independent predictor of patient survival. The tumour microenvironment and the immune checkpoints, such as PD-L1/PD-1, are relevant to the prognoses and also appear to be relevant for further CRC therapies. METHODS: We analysed the presence and features of the infiltrated monocyte/macrophage and lymphocyte populations in both tumour and peritumour samples from patients with CRC (n = 15). RESULTS: We detected a large number of CD14+ monocytes/macrophages with an alternative phenotype (CD64+CD163+) and CD4+ lymphocytes that infiltrated the tumour, but not the peritumour area. The monocytes/macrophages expressed PD-L1, whereas the lymphocytes were PD-1+; however, we did not find high PD-L1 levels in the tumour cells. Coculture of circulating naïve human monocytes/macrophages and lymphocytes with tumour cells from patients with proficient mismatch repair CRC induced both an alternative phenotype with higher expression of PD-L1 in CD14+ cells and the T-cell exhaustion phenomenon. The addition of an α-PD-1 antibody restored lymphocyte proliferation. CONCLUSION: These results emphasise the interesting nature of immune checkpoint shifting therapies, which have potential clinical applications in the context of colorectal cancer.
Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor , Colorectal Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Aged , Aged, 80 and over , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Female , Humans , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Microsatellite Instability , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Phenotype , Protein Binding , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolismABSTRACT
Obstructive sleep apnea (OSA) is a syndrome characterized by repeated pauses in breathing induced by a partial or complete collapse of the upper airways during sleep. Intermittent hypoxia (IH), a hallmark characteristic of OSA, has been proposed to be a major determinant of cancer development, and patients with OSA are at a higher risk of tumors. Both OSA and healthy monocytes have been found to show enhanced HIF1α expression under IH. Moreover, these cells under IH polarize toward a tumor-promoting phenotype in a HIF1α-dependent manner and influence tumor growth via vascular endothelial growth factor (VEGF). Monocytes from patients with OSA increased the tumor-induced microenvironment and exhibited an impaired cytotoxicity in a 3D tumor in vitro model as a result of the increased HIF1α secretion. Adequate oxygen restoration both in vivo (under continuous positive airway pressure treatment, CPAP) and in vitro leads the monocytes to revert the tumor-promoting phenotype, demonstrating the plasticity of the innate immune system and the oxygen recovery relevance in this context.
Subject(s)
Leukocytes, Mononuclear/metabolism , Sleep Apnea, Obstructive/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Aged , Biomarkers/metabolism , Cell Survival/physiology , Cells, Cultured , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Middle Aged , Oxygen/metabolism , Prospective Studies , Spheroids, Cellular/metabolismABSTRACT
The opioid system is well conserved among species and plays a critical role in pain and addiction systems. The use of zebrafish as an experimental model to study development and genetics is extraordinary and has been proven to be relevant for the study of different diseases. The main drawback to its use for the analysis of different pathologies is the lack of protein tools. Antibodies that work in other models are not suitable for zebrafish due to the low degree of homology that exists among the opioid receptor protein sequences in different species. Here we report the successful generation and characterization of antibodies against the mu, delta 1 and delta 2 opioid receptors in zebrafish. The antibodies obtained, which are specific for each receptor due to the use of the C-terminus as antigens, work for Western blotting and immunohistochemistry. In addition, the antibodies against mu and delta 1 opioid receptors, but not those against delta 2, are able to immunoprecipitate the corresponding receptor from zebrafish lysates. The development of opioid receptor antibodies is an asset to the further study of the endogenous opioid system in zebrafish.
Subject(s)
Antibodies/metabolism , Receptors, Opioid/immunology , Zebrafish/metabolism , Amino Acid Sequence , Animals , Antibody Specificity , Female , HEK293 Cells , Humans , Larva/metabolism , Rabbits , Receptors, Opioid/chemistry , Receptors, Opioid, delta/metabolism , Sequence AlignmentABSTRACT
Obstructive sleep apnoea (OSA) is associated with higher cancer incidence, tumour aggressiveness and cancer mortality, as well as greater severity of infections, which have been attributed to an immune deregulation. We studied the expression of programmed cell death (PD)-1 receptor and its ligand (PD-L1) on immune cells from patients with OSA, and its consequences on immune-suppressing activity. We report that PD-L1 was overexpressed on monocytes and PD-1 was overexpressed on CD8+ T-cells in a severity-dependent manner. PD-L1 and PD-1 overexpression were induced in both the human in vitro and murine models of intermittent hypoxia, as well as by hypoxia-inducible factor-1α transfection. PD-L1/PD-1 crosstalk suppressed T-cell proliferation and activation of autologous T-lymphocytes and impaired the cytotoxic activity of CD8+ T-cells. In addition, monocytes from patients with OSA exhibited high levels of retinoic acid related orphan receptor, which might explain the differentiation of myeloid-derived suppressor cells. Intermittent hypoxia upregulated the PD-L1/PD-1 crosstalk in patients with OSA, resulting in a reduction in CD8+ T-cell activation and cytotoxicity, providing biological plausibility to the increased incidence and aggressiveness of cancer and the higher risk of infections described in these patients.
Subject(s)
B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/physiology , Monocytes/physiology , Programmed Cell Death 1 Receptor/metabolism , Sleep Apnea, Obstructive/metabolism , Adult , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Signal Transduction , Sleep Apnea, Obstructive/diagnosis , Up-RegulationABSTRACT
Obstructive sleep apnoea (OSA) is associated with cancer incidence and mortality. The contribution of the immune system appears to be crucial; however, the potential role of monocytes and natural killer (NK) cells remains unclear.Quantitative reverse transcriptase PCR, flow cytometry and in vitro assays were used to analyse the phenotype and immune response activity in 92 patients with OSA (60 recently diagnosed untreated patients and 32 patients after 6â months of treatment with continuous positive airway pressure (CPAP)) and 29 healthy volunteers (HV).We determined that monocytes in patients with OSA exhibit an immunosuppressive phenotype, including surface expression of glycoprotein-A repetitions predominant protein (GARP) and transforming growth factor-ß (TGF-ß), in contrast to those from the HV and CPAP groups. High levels of TGF-ß were detected in OSA sera. TGF-ß release by GARP+ monocytes impaired NK cytotoxicity and maturation. This altered phenotype correlated with the hypoxic severity clinical score (CT90). Reoxygenation eventually restored the altered phenotypes and cytotoxicity.This study demonstrates that GARP+ monocytes from untreated patients with OSA have an NK-suppressing role through their release of TGF-ß. Our findings show that monocyte plasticity immunomodulates NK activity in this pathology, suggesting a potential role in cancer incidence.
Subject(s)
Continuous Positive Airway Pressure/methods , Hypoxia , Killer Cells, Natural/physiology , Membrane Proteins/metabolism , Monocytes/physiology , Sleep Apnea, Obstructive , Transforming Growth Factor beta/metabolism , Cytotoxicity, Immunologic , Female , Humans , Hypoxia/etiology , Hypoxia/metabolism , Hypoxia/therapy , Male , Middle Aged , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/immunology , Sleep Apnea, Obstructive/therapy , Treatment Outcome , Tumor EscapeABSTRACT
Infections are a significant cause of morbidity and mortality in patients with chronic lymphocytic leukemia (CLL). The pathogenesis of infections is multifactorial and includes hypogammaglobulinemia, conventional therapy with alkylating drugs, and recently, purine analogs and mAb-associated T cells. Patients without these risk factors also suffer from infections, although the mechanism remains unknown. In a cohort of 70 patients with CLL, we demonstrated that their monocytes were locked into a refractory state and were unable to mount a classic inflammatory response to pathogens. In addition, they exhibited the primary features of endotoxin tolerance, including low cytokine production, high phagocytic activity, and impaired Ag presentation. The involvement of miR-146a in this phenomenon was suspected. We found miR-146a target genes, such as IRAK1 and TRAF6, were manifestly downregulated. Our study provides a new explanation for infections in patients with CLL and describes a cross-tolerance between endotoxins and tumors.
Subject(s)
Immune Tolerance , Immunity, Innate , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Monocytes/immunology , T-Lymphocytes/immunology , Aged , Aged, 80 and over , Cytokines/immunology , Down-Regulation/immunology , Endotoxins/immunology , Female , Gene Expression Regulation, Leukemic/immunology , Humans , Interleukin-1 Receptor-Associated Kinases/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , MicroRNAs/immunology , Middle Aged , Monocytes/pathology , T-Lymphocytes/pathology , TNF Receptor-Associated Factor 6/immunologyABSTRACT
B-cell lymphoma 10 (BCL10) is not essential for actin polymerisation after FcγR stimulation in human fibroblasts.
Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Fibroblasts/metabolism , Immunologic Deficiency Syndromes/genetics , Polymerization , Receptors, IgG/immunology , Adaptor Proteins, Signal Transducing/immunology , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/immunology , Case-Control Studies , Caspases/immunology , Fibroblasts/immunology , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Microscopy, Fluorescence , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/immunology , Neoplasm Proteins/immunology , Signal Transduction/immunologyABSTRACT
A series of 31 pentacyclic triterpenoids isolated from the root barks of Celastrus vulcanicola and Maytenus jelskii were tested for cytotoxicity and inhibitory activity against lipopolysaccharide (LPS)-induced nitric oxide (NO) production in RAW 264.7 macrophages. Compounds 18 (C18) and 25 (C25) exhibited significant inhibition of LPS-induced NO release at 50 and 25µM concentrations, respectively, and decreased mRNAs of pro-inflammatory cytokines. At the molecular level, C18 neither inhibited LPS-mediated phosphorylation of mitogen activated protein kinases (MAPKs) nor nuclear translocation of nuclear factor kappa beta (NFκB). Instead, C18 enhanced and prolonged nuclear translocation of nuclear factor-erythroid 2-related factor 2 (Nrf2) and increased the expression of its target genes including hemeoxigenase 1 (HO1). C25 efficiently inhibited LPS-mediated phosphorylation of JNK, p38 and ERK, without affecting NFκB or Nrf2 signaling pathways. Both compounds reduced LPS-mediated processing of caspase-1 and the cleavage of interleukin 1ß (IL1ß) proform, reflecting their ability to target the inflammasome. C25 also counteracted LPS effects on iNOS expression and pro-inflammatory cytokines mRNA levels in Bv-2 microglial cells. The anti-inflammatory effect of both compounds was also assessed in human macrophages. Our results suggest that triterpenoids C18 and C25 possess anti-inflammatory effects, which may be therapeutically relevant for diseases linked to inflammation.
Subject(s)
Anti-Inflammatory Agents/pharmacology , Lipopolysaccharides/toxicity , Macrophages/drug effects , Signal Transduction/drug effects , Triterpenes/pharmacology , Animals , Caspase 1/metabolism , Cell Line , Enzyme Induction , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , RNA, Messenger/geneticsABSTRACT
Endotoxin tolerance (ET) is a state of reduced responsiveness to endotoxin stimulation after a primary bacterial insult. This phenomenon has been described in several pathologies, including sepsis, in which an endotoxin challenge results in reduced cytokine production. In this study, we show that the NFκ L chain enhancer of activated B cells 2 (NFκB2)/p100 was overexpressed and accumulated in a well-established in vitro human monocyte model of ET. The p100 accumulation in these cells inversely correlated with the inflammatory response after LPS stimulation. Knocking down NFκB2/p100 using small interfering RNA in human monocytes further indicated that p100 expression is a crucial factor in the progression of ET. The monocytes derived from patients with sepsis had high levels of p100, and a downregulation of NFκB2/p100 in these septic monocytes reversed their ET status.
Subject(s)
Endotoxins/immunology , Immune Tolerance , Monocytes/immunology , NF-kappa B p52 Subunit/biosynthesis , Sepsis/immunology , Aged , Down-Regulation , Gene Knockout Techniques , Humans , Inflammation/immunology , NF-kappa B p52 Subunit/genetics , RNA Interference , RNA, Small InterferingABSTRACT
UNLABELLED: Due to the essential role macrophages play in antiviral immunity, it is important to understand the intracellular and molecular processes that occur in macrophages following infection with various strains of vaccinia virus, particularly those used as vaccine vectors. Similarities as well as differences were found in macrophages infected with different poxvirus strains, particularly at the level of virus-induced apoptosis and the expression of immunomodulatory genes, as determined by microarray analyses. Interestingly, the attenuated modified vaccinia Ankara virus (MVA) was particularly efficient in triggering apoptosis and beta interferon (IFN-ß) secretion and in inducing changes in the expression of genes associated with increased activation of innate immunity, setting it apart from the other five vaccinia virus strains tested. Taken together, these results increase our understanding of how these viruses interact with human macrophages, at the cellular and molecular levels, and suggest mechanisms that may underlie their utility as recombinant vaccine vectors. IMPORTANCE: Our studies clearly demonstrate that there are substantial biological differences in the patterns of cellular gene expression between macrophages infected with different poxvirus strains and that these changes are due specifically to infection with the distinct viruses. For example, a clear induction in IFN-ß mRNA was observed after infection with MVA but not with other poxviruses. Importantly, antiviral bioassays confirmed that MVA-infected macrophages secreted a high level of biologically active type I IFN. Similarly, the phagocytic capacity of macrophages was also specifically increased after infection with MVA. Although the main scope of this study was not to test the vaccine potential of MVA as there are several groups in the field working extensively on this aspect, the characteristics/phenotypes we observed at the in vitro level clearly highlight the inherent advantages that MVA possesses in comparison to other poxvirus strains.
Subject(s)
Apoptosis , Interferons/immunology , Macrophages/immunology , Macrophages/virology , Phagocytosis , Poxviridae/immunology , Signal Transduction , Cells, Cultured , Gene Expression Profiling , Humans , Microarray AnalysisABSTRACT
BACKGROUND AIMS: Natural killer cell (NK) cytotoxic activity plays a major role in natural immunologic defences against malignancies. NK cells are emerging as a tool for adoptive cancer immunotherapies. Arabinoxylan rice bran (MGN-3/Biobran) has been described as a biological response modifier that can enhance the cytotoxic activity of NK cells. This study evaluated the effect of MGN-3/Biobran on NK cell activation, expansion and cytotoxicity against neuroblastoma cells. METHODS: NK cells were enriched with magnetic beads and stimulated with MGN-3/Biobran. NK cell activation was evaluated via analysis of their phenotype, and their expansion capability was tracked. The in vitro cytotoxic ability of the activated NK cells was tested against K562, Jurkat, A673, NB1691, A-204, RD and RH-30 cell lines and the in vivo cytotoxic ability against the NB1691 cell line. RESULTS: MGN-3/Biobran stimulation of NK cells induced a higher expression of the activation-associated receptors CD25 and CD69 than in unstimulated cells (P < 0.05). The expression of NKG2D, DNAM, NCRs and TLRs remained unchanged. Overnight MGN-3/Biobran stimulation increased NK cell cytotoxic activity against all cell lines tested in vitro and decelerated neuroblastoma growth in vivo. The mechanism is not mediated by lipopolysaccharide contamination in MGN-3/Biobran. Furthermore, the addition of MGN-3/Biobran promoted NK cell expansion and decreased T cells in vitro. CONCLUSIONS: Our data show that MGN-3/Biobran upregulates NK cell activation markers, stimulates NK cell cytotoxic activity against neuroblastoma in vitro and in vivo and selectively augments the expansion of NK cells. These results may be useful for future NK cell therapeutic strategies of the treatment of neuroblastoma.
Subject(s)
Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/cytology , Neuroblastoma/immunology , Oryza/chemistry , Xylans/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/metabolism , Disease Models, Animal , Fluorescence , Humans , Immunophenotyping , Killer Cells, Natural/drug effects , Kinetics , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice, Inbred NOD , Mice, SCID , Neuroblastoma/pathology , Receptors, Natural Killer Cell/metabolismABSTRACT
We show that galactomannan, a polysaccharide consisting of a mannose backbone with galactose side groups present on the cell wall of several fungi, induces a reprogramming of the inflammatory response in human macrophages through dectin-1 receptor. The nuclear factor kappa-light-chain-enhancer of activated B cells 2 (NFκB2)/p100 was overexpressed after galactomannan challenge. Knocking down NFκB2/p100 using small interfering RNA (siRNA) indicated that NFκB2/p100 expression is a crucial factor in the progression of the galactomannan-induced refractoriness. The data presented in this study could be used as a modulator of inflammatory response in clinical situations where refractory state is required.
Subject(s)
Inflammation/drug therapy , Macrophages/drug effects , Macrophages/immunology , Mannans/therapeutic use , NF-kappa B p52 Subunit/metabolism , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Galactose/analogs & derivatives , Humans , Inflammation/chemically induced , Lipopolysaccharides/pharmacology , RNA, Small InterferingABSTRACT
Although the natural mode of bacterial growth in nature is as biofilm, almost all antimicrobial and immunological tests are routinely developed using planktonic inoculums. Bacterial biofilms protect the microbial community from external damage and promote the persistence of chronic infections. In this study, interactions between human macrophages and bacterial inoculums of planktonic and biofilm modes of growth have been explored using Escherichia coli (E. coli) K12. Human macrophages phagocytize planktonic E. coli more efficiently than bacteria grown in a biofilm. Moreover, they prefer to phagocytize planktonic bacteria. In this context, CD64 expression is involved. Our data indicate that bacteria with "a biofilm background" avoid phagocytosis by naïve macrophages, which could create a favorable environment for chronic infection. Our findings were corroborated in a clinical O25b-ST131 ESBL-producer E. coli isolate, which caused urinary tract infections.
Subject(s)
Bacteria/growth & development , Biofilms , Macrophages/immunology , Macrophages/microbiology , Phagocytosis , Plankton , Cells, Cultured , Escherichia coli/enzymology , Escherichia coli/growth & development , Humans , Receptors, IgG/immunology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , beta-Lactamases/metabolismABSTRACT
BACKGROUND: Currently, good prognosis and management of critically ill patients with COVID-19 are crucial for developing disease management guidelines and providing a viable healthcare system. We aimed to propose individual outcome prediction models based on binary logistic regression (BLR) and artificial neural network (ANN) analyses of data collected in the first 24 h of intensive care unit (ICU) admission for patients with COVID-19 infection. We also analysed different variables for ICU patients who survived and those who died. METHODS: Data from 326 critically ill patients with COVID-19 were collected. Data were captured on laboratory variables, demographics, comorbidities, symptoms and hospital stay related information. These data were compared with patient outcomes (survivor and non-survivor patients). BLR was assessed using the Wald Forward Stepwise method, and the ANN model was constructed using multilayer perceptron architecture. RESULTS: The area under the receiver operating characteristic curve of the ANN model was significantly larger than the BLR model (0.917 vs 0.810; p < 0.001) for predicting individual outcomes. In addition, ANN model presented similar negative predictive value than the BLR model (95.9% vs 94.8%). Variables such as age, pH, potassium ion, partial pressure of oxygen, and chloride were present in both models and they were significant predictors of death in COVID-19 patients. CONCLUSIONS: Our study could provide helpful information for other hospitals to develop their own individual outcome prediction models based, mainly, on laboratory variables. Furthermore, it offers valuable information on which variables could predict a fatal outcome for ICU patients with COVID-19.
Subject(s)
COVID-19/diagnosis , Aged , Critical Illness , Female , Hospitalization , Humans , Intensive Care Units , Logistic Models , Male , Middle Aged , Models, Statistical , Neural Networks, Computer , Predictive Value of Tests , Prognosis , ROC Curve , Time FactorsABSTRACT
Most breast cancers exhibit low immune infiltration and are unresponsive to immunotherapy. We hypothesized that inhibition of the receptor activator of nuclear factor-κB (RANK) signaling pathway may enhance immune activation. Here we report that loss of RANK signaling in mouse tumor cells increases leukocytes, lymphocytes, and CD8+ T cells, and reduces macrophage and neutrophil infiltration. CD8+ T cells mediate the attenuated tumor phenotype observed upon RANK loss, whereas neutrophils, supported by RANK-expressing tumor cells, induce immunosuppression. RANKL inhibition increases the anti-tumor effect of immunotherapies in breast cancer through a tumor cell mediated effect. Comparably, pre-operative single-agent denosumab in premenopausal early-stage breast cancer patients from the Phase-II D-BEYOND clinical trial (NCT01864798) is well tolerated, inhibits RANK pathway and increases tumor infiltrating lymphocytes and CD8+ T cells. Higher RANK signaling activation in tumors and serum RANKL levels at baseline predict these immune-modulatory effects. No changes in tumor cell proliferation (primary endpoint) or other secondary endpoints are observed. Overall, our preclinical and clinical findings reveal that tumor cells exploit RANK pathway as a mechanism to evade immune surveillance and support the use of RANK pathway inhibitors to prime luminal breast cancer for immunotherapy.