The present invention refers to the medical field. Particularly, it refers to an in vitro method for obtaining clinical data in patients suffering from an inflammatory disease, preferably, for predicting gravity and mortality risk among patients suffering from sepsis or for deciding whether to administer a medical treatment to a patient suffering from an autoinflammatory syndrome.
Sepsis is a life-threatening condition that arises when the body's response to infection causes injury to its own tissues and organs.
The response of the immune system during sepsis is a complex dynamic process involving an initial inflammatory response followed by an immune deactivation. Exacerbation of any of these responses compromises the life of septic patients. The initial production of proinflammatory cytokines aims to protect the host from the invading pathogens, but it can also damage non-infected tissues and lead to the dysfunction of different organs and systems. Immunosuppression after the inflammatory response is a compensatory response aimed at protecting the host from the excess production of cytokines and other inflammatory factors; however, in sepsis there is a profound leukocyte deactivation that is related to complications in critically ill patients and which leads to secondary fatal infections and the majority of deaths associated with sepsis. Changes in the metabolism of leukocytes during sepsis are associated with the different immune responses in sepsis, whereby the glycolytic pathway is upregulated during the inflammatory response while defects in both glycolysis and mitochondrial respiration lead to immunosuppression. However, the molecular mechanisms in the host that initiate and regulate immunoparalysis in human sepsis are still poorly understood.
Pattern recognition receptors in innate immune cells are able to recognize microbial- or damage-associated molecules and initiate a proinflammatory response. Inflammasomes are important signaling complexes formed by a subgroup of intracellular pattern recognition receptors that activate caspase-1. Caspase-1 promotes a specific type of cell death called pyroptosis and the release of the proinflammatory cytokines IL-113, IL-18 and the alarmin high mobility group protein B1 (HMGB1), which are all involved in the inflammatory response of sepsis. A promiscuous type of inflammasome is formed by the nucleotide-binding domain, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) that can be activated in response to different microbial- or damage-associated molecules, as elevated extracellular concentrations of the nucleotide adenosine triphosphate (ATP) signaling through the P2X purinoceptor 7 (P2X7). Upon activation, NLRP3 oligomerizes and recruits the apoptotic speck-like protein with a caspase activating domain (ASC) into large oligomers that can be found in biological fluids upon inflammasome activation and pyroptosis execution.
The present invention if focused on assessing the activation of NLRP3 inflammasome by the P2X7 receptor in blood leukocytes from a cohort of clinically relevant intra-abdominal origin septic patients and then carry out a follow-up analysis of the same individuals after sepsis resolution. Based on this assessment, the present invention provides an in vitro method for predicting mortality risk among patients suffering from sepsis or for deciding whether to administer a medical treatment to a patient suffering from an autoinflammatory syndrome. Regarding, autoinflammatory diseases, this is a group of genetically diverse but clinically similar disorders characterized by recurrent fever associated with rash, serositis, and musculoskeletal involvement. Although they are considered as rare diseases, the increase in the knowledge about their immunopathogenesis has set the bases for a broad spectrum of diseases not only of immunological nature, but also of metabolic, chronic degenerative and inflammatory nature. Despite the limited clinical and genetic diagnostic, there are no functional or biochemistry diagnostic method for autoinflammatory syndromes. We developed a novel functional diagnostic test for these patients that will help the clinician to quickly tailor the treatment for these syndromes. This method will improve the detection and tailor treatment for autoinflammatory syndromes that could also be expanded to other inflammatory, metabolic and chronic degenerative diseases.
Systemic infection during sepsis induces an exacerbated inflammatory response that is followed by an immunosuppression of the host and thus compromising the life of critically ill patients. Defects in the metabolism and mitochondrial failure are associated with immunocompromised septic patients. The NLRP3 inflammasome is important for establishing an inflammatory response after activation by the purinergic P2X7 receptor. After studying a cohort of individuals with intra-abdominal origin sepsis, we found that monocytes from these patients presented impaired NLRP3 activation when the P2X7 receptor was stimulated. Furthermore, most of the sepsis-related deaths were among patients whose NLRP3 activation was profoundly altered. In monocytes from septic patients, P2X7 receptor was associated with mitochondrial dysfunction, and activation of the P2X7 receptor resulted in mitochondrial damage, which in turn inhibited the NLRP3 activation mediated by HIF-1α. Mortality increased in a mouse model of sepsis when the P2X7 receptor was activated in vivo. Thus, the present invention reveals a molecular mechanism initiated by the P2X7 receptor that contributes to NLRP3 impairment during infection.
Such as it is shown in the results provided by the present invention, a Receiver operating characteristic (ROC) analysis showed that the release of IL-1β and the formation of ASC specks from monocytes activated in vitro from septic patients and healthy individuals, as well as the release of HMGB1, IL-6 and TNF-α, were sufficiently significant in the ROC analysis to identify a group of septic patients with impaired NLRP3 inflammasome activation, all with an area under the curve from 0.9 to 1. Particularly, the ROC analysis provided by the present invention for IL-1β, ASC-speck formation, HMBG1, IL-6, TNF-α, comparing control vs septic individuals and septic individuals vs those septic individuals with a profound NLRP3 deactivation, reveals that said biomarkers from monocytes stimulated in vitro can be efficiently used according to the present invention for predicting mortality risk among patients suffering from sepsis.
Moreover, according to the present invention, IL-1β, IL-18, ASC-speck formation, HMBG1, IL-6 and/or TNF-α can be efficiently used according to the present invention for deciding whether to administer a medical treatment to a patient suffering from an autoinflammatory syndrome.
Thus, in a preferred embodiment, any of the biomarkers IL-1β, IL-18, ASC-speck formation, HMBG1, IL-6, IL-8 and/or TNF-α can be individually used, after the activation of NLRP3 inflammasome, for predicting mortality risk among patients suffering from sepsis and/or for deciding whether to administer a medical treatment to a patient suffering from an autoinflammatory syndrome. Nevertheless the present invention also comprises the possible use of any combination of at least two, at least three, at least four, at least five or at least six of said biomarkers. In other words, the present invention also refers to a biomarker signature comprising any individual biomarker of the group: IL-1β, ASC-speck formation, HMBG1, IL-6, IL-8, TNF-α, or any combination thereof.
Consequently, the special technical feature which provides unity of invention is the activation of NLRP3 inflammasome. Once the NLRP3 inflammasome is activated, the decreased production of any of the biomarkers IL-1β, IL-18, ASC-speck formation, HMBG1, IL-6, IL-8 and/or TNF-α can be determined in order to implement the present invention.
Particularly, the first embodiment of the present invention refers to an in vitro method for obtaining clinical data in patients suffering from an inflammatory disease which comprises determining the concentration level of at least one biomarker selected from the group comprising: IL-1β, IL-18, ASC-specks, HMGB1, IL-6 and/or TNF-α, once the NLRP3 inflammasome has been activated in vitro in blood cells obtained from the patient.
In a preferred embodiment, the present invention refers to an in vitro method for predicting mortality risk among patients suffering from sepsis which comprises determining the concentration level of at least one biomarker selected from the group comprising: IL-1β, ASC-specks, HMGB1, IL-6 and/or TNF-α, once the NLRP3 inflammasome has been activated in blood cells obtained from the patient, wherein a variation, preferably a decrease of the concentration level of at least one of said biomarkers with respect to the concentration level determined in control healthy patients, is an indication of mortality risk.
In a preferred embodiment, the present invention refers to an in vitro method for deciding whether to administer a medical treatment to a patient suffering from an autoinflammatory syndrome which comprises determining the concentration level of at least one biomarker selected from the group comprising: IL-1β, IL-18, ASC-specks, HMGB1, IL-6 and/or TNF-α, once the NLRP3 inflammasome has been activated in blood cells obtained from the patient, wherein a variation, preferably decrease of the concentration level of at least one of said biomarkers with respect to the concentration level determined in healthy patients, is an indication that a treatment directed to the inhibition of IL-1β, IL-18, ASC-specks, HMGB1, IL-6 and/or TNF-α can be discarded or wherein a variation, preferably an increase of the concentration level of said biomarkers with respect to the concentration level determined in healthy patients, is an indication that a treatment directed to the inhibition of IL-1β, IL-18, ASC-specks, HMGB1, IL-6 and/or TNF-α can be implemented.
In a preferred embodiment, the determination of the concentration level of the biomarkers is carried out in a suitable media comprising NaCl, HEPES, d-glucose, KCl, CaCl2 and MgCl2, preferably 147 mM NaCl, 10 mM HEPES, 13 mM d-glucose, 2 mM KCl, 2 mM CaCl2, and 1 mM MgCl2 and pH 7.4 once the NLRP3 inflammasome has been activated in blood cells obtained from the patient.
In a preferred embodiment, the NLRP3 inflammasome activation is carried out by means of the activation of P2X7 receptor.
In a preferred embodiment, the NLRP3 inflammasome activation is carried out by other means of the activation of P2X7 receptor with LPS and nigericin, uric acid crystals, alum crystals, intracellular lipids or other inflammasomes as NLRC4 or Pyrin inflammasomes are also activated.
In a preferred embodiment, the blood cells in which the NLRP3 inflammasome is activated are leukocytes, preferably monocytes.
In a preferred embodiment, the blood cells in which the NLRP3 inflammasome is activated are peripheral blood mononuclear cells (PBMC) extracted from whole blood samples.
In a preferred embodiment, the above cited method is performed within 5 days from the patient admission.
In a preferred embodiment, the mortality risk is evaluated within a period of 10-90 days.
In a preferred embodiment, the method of the invention comprises the following steps:
The second embodiment of the present invention refers to the in vitro use of the concentration level of at least one biomarker selected from the group comprising: IL-1β, IL-18, ASC-specks, HMGB1, IL-6 and/or TNF-α, once the NLRP3 inflammasome has been activated in blood cells comprised in blood samples obtained from the patient, for obtaining clinical data in patients suffering from an inflammatory disease.
In a preferred embodiment the present invention refers to the in vitro use of at least one biomarker selected from the group comprising: IL-1β, IL-18, ASC-specks, HMGB1, IL-6 and/or TNF-α, once the NLRP3 inflammasome has been activated in blood cells obtained from the patient, for predicting mortality risk among patients suffering from sepsis or for deciding whether to administer a medical treatment to a patient suffering from an autoinflammatory syndrome.
The third embodiment of the present invention refers to a kit of parts, adapted for performing any of the methods described in the present invention, which comprises at least three components:
In a preferred embodiment, the buffer of the component a) comprises: LPS, ATP, nigericin, uric acid crystals, alum crystals, or other compounds to activate the inflammasomes NLRC4 or Pyrin.
The fourth embodiment of the present invention refer to a method for treating septic patients or patients suffering from an inflammatory disease which initially comprises determining the concentration level of at least one biomarker selected from the group comprising: IL-1β, IL-18, ASC-specks, HMGB1, IL-6 and/or TNF-α, once the NLRP3 inflammasome has been activated in blood cells obtained from the patient.
For the purpose of the present invention the following terms are defined:
The clinical ethics committee of the Clinical University Hospital Virgen de la Arrixaca (Murcia, Spain) approved this study and its procedures (reference number PI13/00174). The samples and data from patients included in this study were provided by the Biobanco en Red de la Region de Murcia (PT13/0010/0018), which is integrated into the Spanish National Biobanks Network (B.000859). All study procedures were conducted in accordance with the declaration of Helsinki. Whole peripheral blood samples were collected after receiving written informed consent from intraabdominal septic patients (n=35, Table 1 showing demographics and clinical characteristics of enrolled patients with intra-abdominal origin sepsis and control groups) at the Surgical Critical Unit from the Clinical University Hospital Virgen de la Arrixaca (Murcia, Spain) after 1, 3, 5 and 120 days of sepsis development, day 1 being the blood sample obtained within 24 hours of the diagnosis of sepsis.
Acute physiology and chronic health evaluation II (APACHE II) and sequential organ failure assessment (SOFA), different clinical, microbiological, hemodynamic and biochemical determinations were routinely evaluated in all septic patients at different days by the Clinical University Hospital Virgen de la Arrixaca Surgical Critical Unit, Clinical Analysis and Microbiology Units. All the septic patients included in this study met the definition for severe sepsis or septic shock that was valid at the time of sample and data collection. The inclusion criteria for septic patients were patients diagnosed with intra-abdominal origin sepsis confirmed by exploratory laparotomy, with at least two diagnostic criteria for sepsis (fever or hypothermia; heart rate greater than 90 beats per minute; tachypnea, leukocytosis or leukopenia) and multiple organ dysfunction defined as physiological dysfunction in two or more organs or organ systems. We excluded patients who were immunocompromised or presented immunodeficiency (including antineoplastic treatments during the month previous to the septic episode). We also excluded terminal oncologic and hematologic neoplastic patients, as well as patients that had a delay of >24 h from intra-abdominal sepsis diagnosis to surgery, patients who spent less than 24 h in the Surgical Critical Unit, those whose infection was not cleared by the surgery and patients who presented another septic focus different from the abdominal focus. We also analyzed whole peripheral blood samples from healthy volunteers (n=11) and abdominal surgery patients who did not developed sepsis (n=14, Table 1).
nsno significant difference (p > 0.05);
The University of Murcia Animal Research Ethical Committee approved animal procedures (ref 5/2014) and then the Animal Health Service of the General Directorate of Fishing and Farming of the Council of Murcia (Servicio de Sanidad Animal, Dirección General de Ganaderia y Pesca, Consejeria de Agricultura y Agua Región de Murcia) approved animal procedures with ref A1320140201. Cecal ligation and puncture (CLP)-induced sepsis was performed in C57BL/6 (WT, wild-type) and P2X7R-deficient (P2rx7−/−) mice in C57BL/6 background. 30 min before CLP, a group of mice received an intraperitoneal injection of ATP (0.5 mg/g) or saline vehicle. Laparotomy was performed to isolate the cecum of mice anesthetized with isoflurane. Approximately ⅔ of the cecum was ligated with a 6-0 silk suture and punctured twice through-and-through with a 21 gauge needle. The abdominal wall and incision were then closed with 6-0 silk suture. Sham operated animals underwent laparotomy without ligation or puncture of the cecum. Buprenorphine (0.3 mg/kg) was administered intraperitoneally at the time of surgery and mice were monitored continuously until recovery from anesthesia. For sample collection, 24 h after the procedure, animals were euthanized with CO2 inhalation and peritoneal lavages were performed with 4 ml of sterile saline and then blood was collected from the thoracic aorta. Serum and centrifuged (cell free) peritoneal lavages were stored at −80° C. until further analysis. Serum was diluted serially in sterile physiologic saline and plated and cultured on agar plates at 37° C. for 24 h. Then the number of bacterial colonies was counted and expressed as CFU/ml of serum.
Human peripheral blood mononuclear cells (PBMCs) were isolated from blood within one hour after extraction using Ficoll histopaque 1077 (Sigma-Aldrich). BMDM were obtained from wild-type, Nlrp3−/−, Casp1−/− and P2rx7−/− mice as previously described. THP-1 cells were maintained in RPMI1640 media with 10% FCS and 2 mM GlutaMAX (Invitrogen). Cells were treated with ATP (Sigma-Aldrich), antimycin A (Sigma-Aldrich) or FCCP (Sigma-Aldrich) in the presence or absence of PDTC (Sigma-Aldrich) or echinomycin (Sigma-Aldrich) in E-total buffer (147 mM NaCl, 10 mM HEPES, 13 mM glucose, 2 mM CaCl2), 1 mM MgCl2, and 2 mM KCl) and then washed and stimulated with E. coli LPS O55:B5 in their respective complete media. In some experiments, PBMCs were activated with recombinant human IL-6, TNF-α or IFNγ (PeproTech). After LPS treatment, cells were incubated with P2X7 modulating nanobodies, the specific P2X7 receptor antagonist A438079 or AZ11645373 (Tocris), and then subsequently stimulated with ATP or nigericin (Sigma-Aldrich) in E-total buffer. Times and concentrations for the reagents used are specified in the figure legends. 23 out of the 35 septic patients included in this study (65.7%) were able to provide enough blood samples for in vitro PBMC stimulation.
Cytokines and soluble P2X7 in plasma were measured by ELISAs from eBioscience, IBL International, R&D Systems, Cusabio (for soluble P2X7) and MBL following the manufacturers' indications and read in a Synergy Mx (BioTek) plate reader. Multiplexing was performed using the Cytometric Bead Array from Becton Dickinson Biosciences following the manufacturer indications, and were analyzed in a BD FACS Canto.
Intracellular ASC-speck formation was evaluated by the Time of Flight Inflammasome Evaluation in CD14+ monocytes using a polyclonal unconjugated rabbit anti-ASC (N-15)-R antibody (SantaCruz Biotechnology) and a secondary monoclonal donkey anti-rabbit antibody Alexa Fluor-488 (Thermofisher Scientific), both at 1:1000 dilution. HIF-1α expression was detected in CD14+ monocytes using Alexa Fluor-647 mouse anti-human HIF-1α (BD Biosciences, Clone 54) at 1:1000 dilution. Monocytes were determined from PBMCs were determined by CD3− CD14+ selection and P2X7 receptor surface expression was determined in CD16−/+/++ monocytes using the monoclonal anti-P2X7 L4 clone conjugated with APC62. Active caspase-1 was measured in monocytes using the specific fluorescent probe FLICA-660 Caspase-1 Assay Kit (Immunochemistry Technologies) following the manufacturer's instructions. Production of ROS was measured in monocytes using the red mitochondrial superoxide indicator MitoSOX (Invitrogen) following the manufacturer's instructions. The detection of extracellular particles of ASC was performed on 0.5 ml of human plasma. For the determination of ASC specks, HIF-1α expression and FLICA, monocytes were selected using anti-CD14 PE (clone 61D3, Tonbo biosciencies) or anti-CD14 APC-H7 (clone MφP9, BD Biosciences). For MitoSOX determination monocytes were selected using anti-CD33 APC-vio770 (clone REA775, Miltenyi). Samples were analyzed by flow cytometry using FACS Canto (BD Biosciences) and the FCS express software (De Novo Software).
Mitochondrial membrane potential was measured using the JC-10 dye (Abcam) in BMDMs with a Sinergy Mx plate reader (BioTek) or by flow cytometry using the BD FACS Celesta flow cytometer (BD Biosciences), or in human monocytes (CD14+ labelled with anti-CD14 APC-H7, clone MφP9, BD Biosciences) using FACS Canto (BD Biosciences), following the indications of manufacturer.
Presence of LDH in cell-free supernatants was measured using the Cytotoxicity Detection kit (Roche) following the manufacturer's instructions.
Detailed methods used for qRT-PCR have been described in the prior art. Relative gene expression levels were calculated using the 2−ΔCt method normalizing to Hprt1 expression as endogenous control.
Statistics were calculated with Prism software (GraphPad Software Inc.). Normality of the samples was determined with D'Agostino and Pearson omnibus K2 normality test and samples did not follow a Gaussian distribution. Outliers from data sets were identified by the ROUT method with Q=1%. Non-parametric Mann-Whitney test was used to analyze differences between two non-paired groups, Wilcoxon test was used to compare two paired groups, and Kruskal-Wallis test was used to analyze differences among three or more groups. The χ2 test was used to determine whether there was a significant difference between different clinical variables among groups of septic patients. Kaplan-Meier was used to estimate the survival of septic patients and the log-rank test was used to compare the survival distributions of samples.
We analyzed a cohort of intra-abdominal origin septic patients (n=35, Table 1) who presented elevated levels of C-reactive protein (CRP) and procalcitonin (PCT) in their plasma 24 hours after sepsis initiation when compared to a control group of abdominal surgery patients that had not developed sepsis (
nsno significant difference (p > 0.05) with Chi-square (χ2) test.
In contrast, the amounts of hemoglobin and bicarbonate were lower than the standards found in healthy individuals. As expected, IL-6 and IL-8 were also higher in the plasma of septic patients when compared to control surgery and healthy groups (
On the other hand, the inflammasome-related cytokines IL-1β, IL-18 and the alarmin HMGB1 were higher in septic patients than in the control groups (
Despite the elevated concentration of IL-1β found in the plasma of septic patients, PBMCs from septic patients presented a decreased release of IL-1β when LPS and ATP stimulation activated NLRP3 inflammasome (
We next found that the septic patients with immunocompromised NLRP3 accounted for over 80% of the deaths registered in the group of septic patients during their stay at the Surgical Critical Unit (
nsno significant difference (p > 0.05).
Escherichia coli
Prevotella sp.
Klebsiella sp.
Bacteroides sp.
Pseudomonas aeruginosa
Enterobacter sp.
Citrobacter sp.
Proteus mirabilis
Clostridium perfringens
Streptococcus sp.
Enterococcus sp.
Staphylococcus sp.
Candida sp.
nsno significant difference (p > 0.05) with Chi-square (χ2) test.
Plasma concentration of CRP, PCT and IL-6 decreased after 3 and 5 days of sepsis onset (
In order to investigate the possible causes of NLRP3 impairment during sepsis, we aimed to study the P2X7 receptor in monocytes as this is the receptor for extracellular ATP, the ligand we used to activate the inflammasome in monocytes from septic patients. We first found that the surface expression of P2X7 receptor was higher in the monocytes of septic patients than in the control groups (
We next found that P2X7 receptor expression positively correlated with the release of IL-1β after ATP stimulation in surgery control patients and non-compromised NLRP3 septic patients (
Having found that stimulating P2X7 receptors in unprimed monocytes and macrophages induced mitochondrial membrane depolarization, we then found simultaneously that NLRP3 inflammasome activation was impaired after LPS-priming and subsequent ATP or nigericin treatment (
To determine if P2X7-receptor activation before bacterial priming was had a significant effect on decreasing survival rates during sepsis, we performed cecal ligation and puncture (CLP) in wild type and P2rx7−/− mice with an initial i.p. ATP injection. We found a significant reduction in the survival of P2rx7−/− mice compared to wild type animals after CLP. Injection of ATP before CLP significantly decreased the survival rates of wild type mice, but not of P2rx7−/− mice (
NLRP3 impairment induced by P2X7 receptor activation in cultured macrophages was transitory, and mitochondrial membrane potential was restored after washing extracellular ATP for 4-12 h (
Similar to septic patients, blood samples from patients with autoinflammatory syndromes is processed and PBMCs are purified (alternatively, we also have preliminary data showing that this test can be performed in whole blood samples without any purification). Blood cells (either whole blood, PBMCs or monocytes) are stimulated with different triggers LPS, LPS+ATP, LPS+Nigericin, LPS+CdtB, or left untreated. Cell supernatants are collected for cytokine determination (IL-1β, IL-18, IL-6, TNFa) and cells are fixed for ASC speck determination by flow cytometry (the same methodology than for septic patients). As it can be observed in
The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications and publications to provide yet further embodiments.
These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.
Number | Date | Country | Kind |
---|---|---|---|
19382184.0 | Mar 2019 | EP | regional |
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/EP2020/056729 | 3/12/2020 | WO |