Next Article in Journal
Cordyceps militaris Grown on Germinated Rhynchosia nulubilis (GRC) Encapsulated in Chitosan Nanoparticle (GCN) Suppresses Particulate Matter (PM)-Induced Lung Inflammation in Mice
Previous Article in Journal
Evaluation of Retinal Structure and Visual Function in Blue Cone Monochromacy to Develop Clinical Endpoints for L-opsin Gene Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Modulation of Septic Shock: A Proteomic Approach

by
Patrícia Terra Alves
1,*,
Aline Gomes de Souza
2,
Victor Alexandre F. Bastos
3,
Eduarda L. Miguel
4,
Augusto César S. Ramos
4,
L. C. Cameron
5,6,
Luiz Ricardo Goulart
1 and
Thúlio M. Cunha
1,4
1
Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil
2
Department of Medical Imaging, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14040-900, SP, Brazil
3
Laboratory of Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil
4
School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil
5
Arthritis Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
6
Lorraine Protein Biochemistry Group, Graduate Program in Neurology, Gaffrée e Guinle University Hospital, Rio de Janeiro 20270-004, RJ, Brazil
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(19), 10641; https://doi.org/10.3390/ijms251910641
Submission received: 28 August 2024 / Revised: 15 September 2024 / Accepted: 18 September 2024 / Published: 3 October 2024
(This article belongs to the Special Issue Role of Proteomics in Human Diseases and Infections)

Abstract

:
Sepsis poses a significant challenge due its lethality, involving multiple organ dysfunction and impaired immune responses. Among several factors affecting sepsis, monocytes play a crucial role; however, their phenotype, proteomic profile, and function in septic shock remain unclear. Our aim was to fully characterize the subpopulations and proteomic profiles of monocytes seen in septic shock cases and discuss their possible impact on the disease. Peripheral blood monocyte subpopulations were phenotype based on CD14/CD16 expression by flow cytometry, and proteins were extracted from the monocytes of individuals with septic shock and healthy controls to identify changes in the global protein expression in these cells. Analysis using 2D-nanoUPLC-UDMSE identified 67 differentially expressed proteins in shock patients compared to controls, in which 44 were upregulated and 23 downregulated. These proteins are involved in monocyte reprogramming, immune dysfunction, severe hypotension, hypo-responsiveness to vasoconstrictors, vasodilation, endothelial dysfunction, vascular injury, and blood clotting, elucidating the disease severity and therapeutic challenges of septic shock. This study identified critical biological targets in monocytes that could serve as potential biomarkers for the diagnosis, prognosis, and treatment of septic shock, providing new insights into the pathophysiology of the disease.

1. Introduction

Sepsis is a potentially fatal clinical syndrome characterized by organ dysfunction caused by the host’s deregulated response to infection [1]. This syndrome evolves into septic shock when the pathophysiological changes are intensified. This entails circulatory, cellular, and metabolic abnormalities becoming severe enough to cause disturbance, and is associated with an increased risk of death [2,3,4].
Arterial hypotension, a hallmark of septic shock, results from hemodynamic changes, including alterations in the degree of hypovolemic response, a decreased vascular tone, and myocardial depression [5]. One of the characteristics of this alteration is the increase in generalized vascular permeability, generating progressive subcutaneous and cavitary edema. This results in blood hypovolemia. Endothelial injury, causing vascular leakage and edema, may be responsible for shock, microvascular thrombosis, and organic failure [6,7]. In septic shock, systemic vascular dysfunction is observed as described above; however, these pivotal changes are essential to controlling the infection. The endothelial lesions trigger inflammatory activation and coagulation. Both are important to activating an immune response against the pathogens and to compartmentalizing the infection, preventing a systemic response. Subsequently, the rolling and adhesion of the white blood cells, and the increase in the vascular permeability, allow these cells to infiltrate tissues and combat invading microorganisms [3,8].
In this context, monocytes play an essential role since they are involved in systemic blood circulation. These cells are also part of the immune system’s first line of scouting defense, with the function of combating microorganisms, performing dead cell clearance, and undertaking injured tissue regeneration [9,10].
Monocytes are present in three distinct populations: classical (CD14high:CD16low), non-classical (CD14low:CD16high), and intermediate (CD14high:CD16high). Classical monocytes are pro-inflammatory, performing phagocytosis and releasing large amounts of reactive oxygen species. Non-classical monocytes are anti-inflammatory, primarily affecting tissue regeneration, immunological surveillance, and vascular endothelial patrol. Intermediate monocytes are monocytes that are transitioning from a classical to a non-classical profile and can display the characteristics of both profiles [11,12].
Understanding the complex biological and clinical roles of monocytes in septic shock remains a challenge. Many researchers have focused their findings and efforts on the cell response to microorganisms, including phagocytic capacity, microbicide, antigen presentation (HLA-DR), and cytokine production, in order to determine if the monocytes display any dysfunction during septic shock. Moreover, different monocyte expression patterns can be observed by examining a patient’s clinical history [10,13,14,15].
In the present study, we intended to evaluate the global protein profile from septic shock patients’ circulating monocytes in order to determine the existent alterations in these cells and to investigate their possible contribution to complete immune collapse and the subsequent mortality.

2. Results

2.1. Monocyte Profile

Following PBMC extraction, three monocyte populations were identified: CD14high:CD16low, CD14high:CD16high and CD14low:CD16high. Control subjects present about 50% CD14high:CD16low monocytes, 12% CD14high:CD16high monocytes, and 12% CD14low:CD16high monocytes. In septic shock patients, the CD14low:CD16high monocyte population was the most abundant in peripheral blood (approximately 70%); 2% of the cells were CD14high:CD16low, and 3% were CD14high:CD16low (Figure S1).

2.2. Mass Spectrometry and Bioinformatics

A total of 3472 proteins were identified in our study (1705 in control samples and 1727 in the septic shock samples), with an average of 7 peptides per protein. For the entire study, the coefficients of variance for all quantitative protein measurements were less than 6%. Sixty-seven proteins were differentially expressed in the monocytes from septic shock patients compared to controls. Considering at least 3 folds in the protein expression levels, 48 proteins were found to be increased, while 23 protein levels were decreased. The details of these proteins, including statistical values and cellular locations, are presented in Table 1.
Gene ontology, FunRich, and Pathway Studio were used to analyze the changes present in the monocytes of septic shock patients. Major cellular components and biological processes involved in the altered proteins were identified using gene ontology analysis in FunRich (Figure 1).
Although differentially expressed proteins did not correlate directly with each other, they participated in common pathways, forming an interactome, with only one molecule required for communication between differentially expressed proteins (Figure 1). The Supplementary Table S2 describes the proteins that are interconnected.
The major regulatory molecules of proteins that are augmented are nuclear proteins with Enhancer-binding protein action, such as CEBPA, CEBPE, and CEBPP. The inflammasome and S100A3 protein in the cytoplasm and signaling pathways are activated by HIF-1a, interleukin 17, and lactoferrin (Table 2).
Proteins that have decreased expressions are mainly influenced by transcriptional factors GRHL2 and NFX1; growth factors TGFA and FGF2; integrins ITGA3 and ILK; and PLAT and Oncostatin M proteins, with actions described in Table 3.
To assess whether these protein changes influence monocyte function, we examined their effects on phenotype, cellular activation, phagocytosis, migration, senescence, cell viability, and apoptosis/necrosis.
Among the 67 differentially expressed proteins, 46 (29 upregulated and 17 downregulated) were associated with these functions, but it remains unclear which changes are most relevant for monocyte dysfunction (Table S3).
Resistin, S100A8, S100A9, MPO, LCN2, PDPK1, CSTG, LTF, PTP1, and EPRS are proteins that may act directly in the collapse of sepsis since they are increased in septic shock cases and can be secreted by monocytes. These proteins have been correlated with direct effects in situations of endotoxemia, chronic inflammation, vascular damage, and thrombosis (Table 4).

3. Discussion

Monocytes play a crucial role in orchestrating the host immune response and contribute to the pathogenesis of sepsis. In the early stages of this syndrome, monocytes exacerbate the immune response through a cytokine storm. As the disease progresses, they contribute to immunological exhaustion and adopt an immunosuppressive phenotype, being incapable of responding to secondary infections [103,104,105].
Classical, intermediate, and non-classical monocytes have distinct predominant functions [106]. Disturbance in the proportion of these monocytes subsets in the blood has been linked to several serious pathologies with poor clinical outcomes [107,108,109].
In this study, patients with septic shock exhibited a predominance of non-classical monocytes in circulation. Non-classical monocytes play a crucial role in septic shock by serving as potential diagnostic markers and indicators of disease severity. Research has shown that the proportion of non-classical monocytes is significantly higher in septic patients compared to non-septic individuals, indicating their potential value in diagnosing sepsis [110]. Additionally, the frequency of classical and intermediate monocytes at the time of ICU admission can predict short-term survival in patients with septic shock, with distinct differences observed between survivors and non-survivors [107]. Furthermore, the dynamic nature of sepsis involves a shift in monocyte polarization, with an increase in intermediate monocytes in patients with suspected or confirmed sepsis, highlighting their role in the immune response during disease progression [111]. Additionally, an increase in non-classical monocytes in patients with clinical symptoms of sepsis was reported to be associated with negative bacterial blood cultures, while an increase in intermediate monocytes population was associated with positive blood cultures [112,113], suggesting that non-classical monocytes may play a role in controlling intermediate infection disruptions. These findings underscore the significance of non-classical monocytes in septic shock, and as potential diagnostic and prognostic markers.
Patients with high proportions of non-classical classical monocytes and low proportions of classical monocytes exhibit impaired endothelial function; as such, it is recognized that high levels of non-classical monocytes are associated with increased vascular superoxide production, leading to vascular dysfunction [11,114,115].
Although immunophenotyping indicated the predominance of non-classical monocytes in septic shock patients, the S100A9, CTSG, MPO and RNAS3 proteins, which are upregulated, have been identified in intermediate and classical monocytes [11,116,117]. The large increase in non-classical monocytes in the circulation of the analyzed patients may be the result of rapid migration without complete differentiation.
As noted, there is an apparent paradox between the predominance of non-classical monocytes, which are typically associated with anti-inflammatory and tissue repair functions in healthy individuals, and the pro-inflammatory and prothrombotic protein expression observed in our septic shock cohort. This shift in phenotype could be explained by the extreme inflammatory environment of septic shock, which may induce atypical activation states in these cells. The rapid recruitment and incomplete differentiation of non-classical monocytes may drive them into a more pro-inflammatory role, as evidenced by the upregulation of proteins like S100A9, CTSG, and MPO. This atypical behavior mirrors the findings of [118], who described monocytes with M2-like features as exhibiting inflammatory activity in septic patients. Such a shift suggests a maladaptive immune response, where non-classical monocytes contribute to immune dysregulation under the severe conditions of septic shock.
The Enhancer-binding protein, HIF-1a, and the inflammasome are major regulatory molecules in the transcription of various genes, culminating in increased expression of different proteins. They play key roles in the regulation of monocytes. (Table 2).
The Enhancer-binding protein regulates almost all listed proteins. In addition, it has been associated with essential proteins for the differentiation of classical monocytes into non-classical ones [119,120,121]. On the other hand, hypoxia-inducible factor-1α (HIF-1α) induces immunosuppression and enhances repair characteristics in classical and intermediate monocytes during active sepsis. It regulates metabolic pathways, promotes glycolysis, and reduces oxidative phosphorylation, adapting monocyte metabolism to hypoxic conditions. HIF-1α also supports cellular survival, repair, and differentiation into anti-inflammatory subtypes, while enhancing phagocytic activity in order to effectively clear pathogens [122,123].
The inflammasome, a crowded multiprotein complex, finely regulates caspase-1 to induce pro-inflammatory protein expression. During septic shock, the activity of the inflammasome, particularly the NLRP3 inflammasome, is dysregulated, which contributes to immune system imbalance and monocyte deactivation [124,125,126,127].
Many studies have identified monocyte dysfunction in septic shock cases, particularly regarding changes in their ability to present antigens and produce cytokines. However, it is known that their phagocytic and microbicidal functions remain conserved [128,129].
In response to pathogens, monocytes can secrete molecules that have a direct effect on the collapse of septic shock (Table 4).
Defense mechanisms are beneficial when activated locally, but harmful when systemically activated. Septic shock results from generalized consequences of the immune response to infection, which involves a cascade of events including endotoxemia, inflammation, vascular damage, and thrombosis [130,131].
Endotoxemia is correlated with infection per se. It involves components of the pathogen’s cell wall being present in abundance in the systemic circulation. While Gram-negative bacteria are frequently associated with endotoxemia, high levels of endotoxin have also been found in the plasma of patients with septic shock caused by Gram-positive bacteria, fungi and, in some cases, viruses [132,133,134].
The presence of endotoxins activates the immune system cells. This process leads to utter inflammation and disturbance in the production of pro-inflammatory cytokines, such as TNF-α, IL-6, IL-1β, IL-8, and IL-12, as well as in the production of platelet activation factors, reactive oxygen species, and microbicidal components (myeloperoxidase, cationic proteins, acid hydrolases, cathepsin G, and lactoferrin), which struggle against microorganisms [135,136,137,138]. However, the exacerbation of these factors causes damage to the vascular endothelium [6,7,8].
Prolonged inflammation and the presence of endothelial injury push homeostasis toward a prothrombotic and anti-fibrinolytic state, which can lead to disseminated microvascular thrombosis, organ ischemia, and multiple organ dysfunction syndrome, common and well-known circumstances that follow septic shock [139,140].

4. Materials and Methods

4.1. Study Population

The Ethics Committee of the Federal University of Uberlândia (UFU) approved this study under protocol number 153.331. Prior to blood collection, written informed consent was obtained from all controls and patients or their guardians.

4.1.1. Healthy Volunteers

The control group consisted of 10 healthy individuals, including 5 women and 5 men (age range, 19–55 years; median age, 27 years). Exclusion criteria included HIV infection, autoimmune disease, or any condition compromising full health.

4.1.2. Patients

Five patients, who were in treatment at the Intensive Care Unit in the Clinical Hospital—Federal University of Uberlândia (Uberlândia—Brazil), were included in this study. All patients were over than 18 years and fulfilled the defining criteria of septic shock [4]. Samples were collected within 48 h of diagnosis. Relevant clinical and laboratory data for septic shock patients are displayed in Supplementary Table S1.

4.2. Peripheral Blood Monocytes Isolation

Approximately 40 mL of blood was collected from patients and control subjects in tubes containing Heparin (an anti-coagulant). Fresh blood samples were immediately processed to isolate peripheral blood mononuclear cells (PBMCs) using density gradient centrifugation through Histopaque-1077 (Sigma, Sigma, St. Louis, MO, USA, Catalog n° H1077-1). Subsequently, monocytes were isolated from PBMCs using the Dynabeads untouched human monocytes kit, (Thermo Fisher Scientific, Waltham, MA, USA, Catalog n°11350D) according to the manufacturer’s instructions. The kit contains antibodies and magnetic beads to capture T cells, B cells, NK cells, dendritic cells, erythrocytes, granulocytes, and macrophages, leaving monocytes untouched and free of surface-bound antibodies and beads. Monocyte populations in PBMCs were characterized by flow cytometry using CD14 and CD16 antibodies (Bio-legend: San Diego, CA, USA: CD14 PECy7, catalog: 325618/400126, and CD16 Alexa Fluor 647, catalog: 302020/400130).

4.3. Protein Extraction and Sample Preparation

Monocytes total proteins were extracted using Complete Lysis-M, EDTA-free (Sigma Aldrich, St. Louis, MO, USA, catalog: 04719964001), according to the manufacturer’s protocol. Proteins were concentrated using Amicon ultra-filtration devices with 10 kDa molecular weight cutoff membranes (Merck-Millipore, Darmstadt, Germany, catalog: UFC501096) and quantified by the Micro BCA Protein assay kit (Thermo Fisher Scientific, Waltham, MA, USA, catalog: 23235). Samples were adjusted to the same protein concentration and digested with trypsin, as described below. Each pooled sample (50 μg of total protein) was buffer-exchanged in 50 mM ammonium bicarbonate, denatured in the presence of 0.2% RapiGEST SF (Waters, Milford, MA, USA) at 80 °C for 15 min in a dry bath, reduced with 100 mM dithiothreitol at 60 °C for 60 min, and then alkylated with 300 mM iodoacetamide for 30 min in the dark at room temperature. Samples were then digested with modified trypsin (Promega, Madison, WI, USA) at an enzyme-to-protein ratio of 1:100 (w/w) at 37 °C overnight. The reaction was stopped using 10 μL of 5% (v/v) trifluoroacetic acid (TFA), mixed, and incubated for 90 min at 37 °C. Samples were centrifuged at 14,000 rpm at 4 °C for 30 min (10). The supernatant was recovered and then reconstituted in ammonium hydroxide (NH4OH) previously prepared at 1 N and then transferred to a Waters Total Recovery vial (Waters) prior to 2D-nanoUPLC-UDMSE analyses. Tryptic digested peptides from yeast alcohol dehydrogenase (ADH) (Waters, Milford, MA, USA) were added to a final concentration of 25 fmol.μL−1 as an internal standard for relative quantification and column loading capacity estimation. The quantitative method utilized in the above estimation is described in detail by Jeffrey Silva et al. (2006) and was performed accordingly [141]. Each sample were injected during “scouting” runs for stoichiometry purposes between assessments of the conditions using an integrated total ion current, as previously described [142].

4.4. Mass Spectrometry of Complex Digested Samples

Using a 2D-nanoUPLC-tandem nanoESI-UDMSE instrument platform, proteomic analyses were performed through multiplexed data-independent acquisition experiments [143].
Samples were fractionated using a dual reversed-phase (RP) approach. In first-dimension chromatography, peptides (5 µg) were loaded onto an M-Class BEH C18 Column (130 Å, 5 µm, 300 µm × 50 mm, Waters Corporation, Milford, MA, USA). Fractionation was performed through 10 discontinuous steps involving acetonitrile (8.7%, 11.4%, 13.2%, 14.7%, 16.0%, 17.4%, 18.9%, 20.7%, 23.4%, 50%) and via high-pH fractionation over 10 min at a flow rate of 2 µL.min−1. After each step, peptide loads were carried through to second-dimension separation on a nanoACQUITY UPLC HSS T3 Column (1.8 µm, 75 µm × 150 mm, Waters Corporation).
Peptide elution was performed using a continuous acetonitrile gradient from 7% to 40% (v/v) over 54 min at a flow rate of 0.4 µL.min−1 directly into a Synapt G2-S HDMS. For every measurement, the mass spectrometer was operated in resolution mode with the scan time (0.5 s) adjusted to obtain at least 10 points per chromatographic peak for each low and elevated energy level (σ10%:20). The m/z resolving power was approximately 1,800,000 FWHM when considering a conjoined stacked-ring ion method such as the T-wave ion mobility, which operates with a cross-section resolving power of at least 40 Ω/ΔΩ. LC–MS/MS multiplex data were collected using ion-mobility-enhanced MSE [144,145].
The exact mass retention time (EMRT) signals from multiplexed ion-mobility DIA scanning (UDMSE) were detected in an alternating low-energy and elevated-energy acquisition mode. In the low-energy mode, data were collected at 6 eV. In the elevated collision energy, quasi m/z-specific collision energies were applied at the traveling-wave stacked-ring ion guide transfer lens (TWIG) to the different drift time bins [146]. These were used to fragment precursor ions prior to orthogonal acceleration time-of-flight (oaTOF) analysis, at the transfer TWIG cell filled with argon gas, using collision-induced dissociation (CID) [147].
The mass spectrometer was calibrated with an MS/MS spectrum of human [Glu1]-Fibrinopeptide B (Glu-Fib) that was delivered every 30 s through the reference sprayer of the NanoLock Spray source. Quadrupole profiles were adjusted if an m/z less than 400 arose from dissociations in the collision cell. Global quality control of the obtained data and the figures of merit (FOMs) are displayed as described at Souza et al., 2017 (Figure S2) [147].

4.5. Database Searching and Bioinformatics

Proteins were identified and quantified using dedicated algorithms and searched against the UniProt human proteomic database, version 2016_02 [147,148]. For proper spectra processing and database searching conditions, Progenesis QI V3.0 for Proteomics software with Apex3D, Peptide 3D, and Ion Accounting informatics were exploited (Waters Corporation). Processing parameters included 150 counts for the low-energy threshold, 50.0 counts for the elevated energy threshold, and 750 counts for the intensity threshold as a default, such as peak picking and alignment processing [149]. The following parameters were considered for protein identification and quantitation: (1) digestion by trypsin with at least one missed cleavage; (2) variable modifications by oxidation (M) and fixed modification by carbamidomethyl (C); and a (3) false discovery rate (FDR) of less than 1%. We used at least three peptides per protein, with one unique aspect, and performed quantitation based on non-conflicted peptides per condition. Only reproducible proteins were investigated across all the replicates obtained. Identifications and quantitative values that did not satisfy these criteria were rejected. The raw files and the database search tables were deposited in the ProteomeXchange repository under accession number PXD004696.

Functional Annotation and In Silico Analysis of Monocyte Function

Functional annotations (gene ontology and chromosome annotations) of the dataset were performed using DAVID Bioinformatics Resources 6.8 (https://david.ncifcrf.gov/home.jsp (accessed on 4 March 2017)). Gene ontology (GO) functional classifications were analyzed with Blast2GO software V6.0.1 (www.blast2go.org (accessed on 6 April 2017)) and GO enrichment analysis was conducted to identify GO terms that were significantly enriched in differentially expressed proteins. We also utilized FUNRICH software V3.1.4 (http://www.funrich.org/ (accessed on 18 May 2017)) to evaluate the interactome and assess the functional enrichment of pathways associated with monocyte activities.
In silico analysis was performed to predict the potential impact of the identified protein changes on key monocyte functions, including phenotype, cellular activation, phagocytosis, migration, senescence, cell viability, and apoptosis/necrosis. Using gene ontology, we categorized the proteins based on their associated biological processes, molecular functions, and cellular components, which are linked to monocyte behavior. FunRich was employed to identify enriched pathways and potential protein interactions relevant to monocyte activation and migration. Furthermore, Pathway Studio Software V21.0 (www.pathwaystudio.com (accessed on 22 June 2017)) was used to explore regulatory networks and signaling pathways, allowing us to predict the influence of the identified proteins on processes such as cellular activation, apoptosis, and necrosis. These tools enabled us to model how protein alterations observed in monocytes during septic shock might affect their function.

5. Study Limitations

This study has limitations, such as the small sample size, and the confirmation regarding the impact of global proteome alterations in monocytes being based solely on in silico analyses and supported only by a literature review. However, this is the first study to demonstrate how monocytes can modify the microenvironment and contribute to the immunopathological collapse in septic shock, providing new insights into the role of these cells in the progression of the disease.

6. Conclusions

Sepsis is a syndrome in which the human system undergoes extreme immune and pathophysiological stress, characterized by initial immune system exacerbation and later physiological response silencing. Monocyte changes in sepsis shock suggest a “biological reprogramming” rather than a simple loss of function. In this study, we presented robust evidence on how the monocyte systemic effect occurs upon the septic shock collapse. Despite limitations, such as pool samples and the absence of separation of the monocytes in subtypes, we demonstrated the potential contribution of monocytes to the collapse of the septic shock. Further studies are needed to validate these findings and to ascertain the contribution of each individual protein in monocyte functional alterations during sepsis. However, these findings could have important clinical implications. The observed monocytes alterations may serve to evaluate the progression in septic shock. By addressing immune dysfunction and vascular damage, these insights may contribute to the future development of personalized treatment strategies, potentially improving patient outcomes and reducing mortality rates.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms251910641/s1.

Author Contributions

Conceptualization: P.T.A., L.R.G. and T.M.C.; Methodology: P.T.A., L.C.C., L.R.G. and T.M.C.; Software: P.T.A., A.G.d.S. and L.C.C.; Validation: P.T.A. and A.G.d.S.; Formal analysis: P.T.A., A.G.d.S. and L.C.C.; Investigation: P.T.A., A.G.d.S., V.A.F.B., E.L.M. and A.C.S.R.; Resources: L.R.G.; Data Curation: P.T.A., L.R.G. and T.M.C.; writing—original draft preparation: P.T.A., L.R.G. and T.M.C.; writing—review and editing: P.T.A., V.A.F.B., L.R.G. and T.M.C.; visualization: P.T.A., L.R.G. and T.M.C.; supervision: L.R.G. and T.M.C.; project administration: L.R.G. and T.M.C.; funding acquisition: L.R.G. and T.M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Institutes of Science and Technology in Theranostics and Biotechnology (INCT-TeraNano, CNPq Process nº 403193/2022-2 and Minas Gerais State Research Support Foundation (FAPEMIG) Process nº CBB—APQ-03613-17).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of Federal University of Uberlandia (protocol code 153.331 and 23 November 2012).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The original contributions presented in this study are included in this article/Supplementary Material; further inquiries can be directed to the corresponding author.

Acknowledgments

The authors thank and pay respect to Luiz Ricardo Goulart Filho a brilliant scientist who passed away due to COVID-19 complications. Your departure was premature. The authors would also like to thank Gustavo Henrique Martins Ferreira de Souza, from Waters Corporation for the proteomic analysis. We are also grateful to all the individuals who participated in this work, the medical staff from the Intensive Unit Care of the University Hospital at UFU, for providing the biological samples and the clinical parameters.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Evans, L.; Rhodes, A.; Alhazzani, W.; Antonelli, M.; Coopersmith, C.M.; French, C.; Machado, F.R.; Mcintyre, L.; Ostermann, M.; Prescott, H.C.; et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock 2021. Intensiv. Care Med. 2021, 47, 1181–1247. [Google Scholar] [CrossRef] [PubMed]
  2. Marques, A.; Torre, C.; Pinto, R.; Sepodes, B.; Rocha, J. Treatment Advances in Sepsis and Septic Shock: Modulating Pro- and Anti-Inflammatory Mechanisms. J. Clin. Med. 2023, 12, 2892. [Google Scholar] [CrossRef]
  3. Jarczak, D.; Kluge, S.; Nierhaus, A. Sepsis—Pathophysiology and Therapeutic Concepts. Front. Med. 2021, 8, 628302. [Google Scholar] [CrossRef] [PubMed]
  4. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.-D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef]
  5. Backer, D.D.; Hajjar, L.; Monnet, X. Vasoconstriction in Septic Shock. Intensiv. Care Med. 2024, 50, 459–462. [Google Scholar] [CrossRef] [PubMed]
  6. Fernández-Sarmiento, J.; Schlapbach, L.J.; Acevedo, L.; Santana, C.R.; Acosta, Y.; Diana, A.; Monsalve, M.; Carcillo, J.A. Endothelial Damage in Sepsis: The Importance of Systems Biology. Front. Pediatr. 2022, 10, 828968. [Google Scholar] [CrossRef] [PubMed]
  7. Dolmatova, E.V.; Wang, K.; Mandavilli, R.; Griendling, K.K. The Effects of Sepsis on Endothelium and Clinical Implications. Cardiovasc. Res. 2020, 117, 60–73. [Google Scholar] [CrossRef]
  8. Joffre, J.; Hellman, J.; Ince, C.; Ait-Oufella, H. Endothelial Responses in Sepsis. Am. J. Respir. Crit. Care Med. 2019, 202, 361–370. [Google Scholar] [CrossRef]
  9. Han, Z.; Liu, Q.; Li, H.; Zhang, M.; You, L.; Lin, Y.; Wang, K.; Gou, Q.; Wang, Z.; Zhou, S.; et al. The Role of Monocytes in Thrombotic Diseases: A Review. Front. Cardiovasc. Med. 2023, 10, 1113827. [Google Scholar] [CrossRef]
  10. Ning, D.; Garg, K.; Mayer, B.; Schick, B.; Bracht, H.; Barth, E.; Weiss, M.; Li, C.; Schneider, J.; Schneider, E.M. Monocyte Subtype Expression Patterns in Septic Patients with Diabetes Are Distinct from Patterns Observed in Obese Patients. Front. Med. 2023, 9, 1026298. [Google Scholar] [CrossRef]
  11. Kapellos, T.S.; Bonaguro, L.; Gemünd, I.; Reusch, N.; Saglam, A.; Hinkley, E.R.; Schultze, J.L. Human Monocyte Subsets and Phenotypes in Major Chronic Inflammatory Diseases. Front. Immunol. 2019, 10, 2035. [Google Scholar] [CrossRef] [PubMed]
  12. Teh, Y.C.; Ding, J.L.; Ng, L.G.; Chong, S.Z. Capturing the Fantastic Voyage of Monocytes through Time and Space. Front. Immunol. 2019, 10, 834. [Google Scholar] [CrossRef]
  13. Beltrán-García, J.; Osca-Verdegal, R.; Jávega, B.; Herrera, G.; O’Connor, J.-E.; García-López, E.; Casabó-Vallés, G.; Rodriguez-Gimillo, M.; Ferreres, J.; Carbonell, N.; et al. Characterization of Early Peripheral Immune Responses in Patients with Sepsis and Septic Shock. Biomedicines 2022, 10, 525. [Google Scholar] [CrossRef] [PubMed]
  14. Gorecki, G.; Cochior, D.; Moldovan, C.; Rusu, E. Molecular Mechanisms in Septic Shock (Review). Exp. Ther. Med. 2021, 22, 1161. [Google Scholar] [CrossRef] [PubMed]
  15. Wiersinga, W.J.; van der Poll, T. Immunopathophysiology of Human Sepsis. eBioMedicine 2022, 86, 104363. [Google Scholar] [CrossRef]
  16. Verbeek, W.; Lekstrom-Himes, J.; Park, D.J.; Dang, P.M.-C.; Vuong, P.T.; Kawano, S.; Babior, B.M.; Xanthopoulos, K.; Koeffler, H.P. Myeloid Transcription Factor C/EBPɛ Is Involved in the Positive Regulation of Lactoferrin Gene Expression in Neutrophils. Blood 1999, 94, 3141–3150. [Google Scholar] [CrossRef]
  17. Wolff, L.; Ackerman, S.J.; Nucifora, G. Meeting Report: Seventh International Workshop on Molecular Aspects of Myeloid Stem Cell Development and Leukemia, Annapolis, MD, May 13–16, 2007. Exp. Hematol. 2008, 36, 523–532. [Google Scholar] [CrossRef]
  18. Asou, N. The Role of a Runt Domain Transcription Factor AML1/RUNX1 in Leukemogenesis and Its Clinical Implications. Crit. Rev. Oncol. Hematol. 2003, 45, 129–150. [Google Scholar] [CrossRef]
  19. Velusamy, S.K.; Poojary, R.; Ardeshna, R.; Alabdulmohsen, W.; Fine, D.H.; Velliyagounder, K. Protective Effects of Human Lactoferrin during Aggregatibacter Actinomycetemcomitans-Induced Bacteremia in Lactoferrin-Deficient Mice. Antimicrob. Agents Chemother. 2014, 58, 397–404. [Google Scholar] [CrossRef]
  20. Natarajan, R.; Salloum, F.N.; Fisher, B.J.; Ownby, E.D.; Kukreja, R.C.; Fowler, A.A. Activation of Hypoxia-Inducible Factor-1 via Prolyl-4 Hydoxylase-2 Gene Silencing Attenuates Acute Inflammatory Responses in Postischemic Myocardium. Am. J. Physiol.-Heart Circ. Physiol. 2007, 293, H1571–H1580. [Google Scholar] [CrossRef]
  21. Zhang, Z.; Mahajan, S.; Zhang, X.; Stanley, S.L. Tumor Necrosis Factor Alpha Is a Key Mediator of Gut Inflammation Seen in Amebic Colitis in Human Intestine in the SCID Mouse-Human Intestinal Xenograft Model of Disease. Infect. Immun. 2003, 71, 5355–5359. [Google Scholar] [CrossRef] [PubMed]
  22. Katzenback, B.A.; Katakura, F.; Belosevic, M. Goldfish (Carassius auratus L.) as a Model System to Study the Growth Factors, Receptors and Transcription Factors That Govern Myelopoiesis in Fish. Dev. Comp. Immunol. 2016, 58, 68–85. [Google Scholar] [CrossRef] [PubMed]
  23. Gaviria-Agudelo, C.; Carter, K.; Tareen, N.; Pascual, V.; Copley, L.A. Gene Expression Analysis of Children with Acute Hematogenous Osteomyelitis Caused by Methicillin-Resistant Staphylococcus Aureus: Correlation with Clinical Severity of Illness. PLoS ONE 2014, 9, e103523. [Google Scholar] [CrossRef]
  24. Zhang, Z.; Zhang, Z.; Kang, Y.; Hou, C.; Duan, X.; Sheng, P.; Sandell, L.; Liao, W. Resistin Stimulates Expression of Chemokine Genes in Chondrocytes via Combinatorial Regulation of C/EBPβ and NF-ΚB. Int. J. Mol. Sci. 2014, 15, 17242–17255. [Google Scholar] [CrossRef] [PubMed]
  25. Bagi, Z.; Broskova, Z.; Feher, A. Obesity and Coronary Microvascular Disease—Implications for Adipose Tissue-Mediated Remote Inflammatory Response. Curr. Vasc. Pharmacol. 2014, 12, 453–461. [Google Scholar] [CrossRef] [PubMed]
  26. Sauter, N.S.; Schulthess, F.T.; Galasso, R.; Castellani, L.W.; Maedler, K. The Antiinflammatory Cytokine Interleukin-1 Receptor Antagonist Protects from High-Fat Diet-Induced Hyperglycemia. Endocrinology 2008, 149, 2208–2218. [Google Scholar] [CrossRef] [PubMed]
  27. Singh, A.K.; Battu, A.; Mohareer, K.; Hasnain, S.E.; Ehtesham, N.Z. Transcription of Human Resistin Gene Involves an Interaction of Sp1 with Peroxisome Proliferator-Activating Receptor Gamma (PPARγ). PLoS ONE 2010, 5, e9912. [Google Scholar] [CrossRef]
  28. Gombart, A.F.; Kwok, S.H.; Anderson, K.L.; Yamaguchi, Y.; Torbett, B.E.; Koeffler, H.P. Regulation of Neutrophil and Eosinophil Secondary Granule Gene Expression by Transcription Factors C/EBPε and PU.1. Blood 2003, 101, 3265–3273. [Google Scholar] [CrossRef]
  29. Hu, S.; Peng, L.; Kwak, Y.-T.; Tekippe, E.M.; Pasare, C.; Malter, J.S.; Hooper, L.V.; Zaki, M.H. The DNA Sensor AIM2 Maintains Intestinal Homeostasis via Regulation of Epithelial Antimicrobial Host Defense. Cell Rep. 2015, 13, 1922–1936. [Google Scholar] [CrossRef]
  30. Guo, H.; Jin, D.; Chen, X. Lipocalin 2 Is a Regulator of Macrophage Polarization and NF-ΚB/STAT3 Pathway Activation. Mol. Endocrinol. 2014, 28, 1616–1628. [Google Scholar] [CrossRef]
  31. Chakraborty, S.; Kaur, S.; Guha, S.; Batra, S.K. The Multifaceted Roles of Neutrophil Gelatinase Associated Lipocalin (NGAL) in Inflammation and Cancer. Biochim. Biophys. Acta (BBA)—Rev. Cancer 2012, 1826, 129–169. [Google Scholar] [CrossRef] [PubMed]
  32. Gaffen, S.L.; Jain, R.; Garg, A.V.; Cua, D.J. The IL-23–IL-17 Immune Axis: From Mechanisms to Therapeutic Testing. Nat. Rev. Immunol. 2014, 14, 585–600. [Google Scholar] [CrossRef] [PubMed]
  33. Gebhardt, C.; Németh, J.; Angel, P.; Hess, J. S100A8 and S100A9 in Inflammation and Cancer. Biochem. Pharmacol. 2006, 72, 1622–1631. [Google Scholar] [CrossRef] [PubMed]
  34. Curran, C.S.; Bertics, P.J. Lactoferrin Regulates an Axis Involving CD11b and CD49d Integrins and the Chemokines MIP-1α and MCP-1 in GM-CSF-Treated Human Primary Eosinophils. J. Interf. Cytokine Res. 2012, 32, 450–461. [Google Scholar] [CrossRef]
  35. Bando, M.; Zou, X.; Hiroshima, Y.; Kataoka, M.; Ross, K.F.; Shinohara, Y.; Nagata, T.; Herzberg, M.C.; Kido, J. Mechanism of Interleukin-1α Transcriptional Regulation of S100A9 in a Human Epidermal Keratinocyte Cell Line. Biochim. Biophys. Acta (BBA)—Gene Regul. Mech. 2013, 1829, 954–962. [Google Scholar] [CrossRef]
  36. Miao, L.; Grebhardt, S.; Shi, J.; Peipe, I.; Zhang, J.; Mayer, D. Prostaglandin E2 Stimulates S100A8 Expression by Activating Protein Kinase A and CCAAT/Enhancer-Binding-Protein-Beta in Prostate Cancer Cells. Int. J. Biochem. Cell Biol. 2012, 44, 1919–1928. [Google Scholar] [CrossRef]
  37. Voigtländer, T.; Wlecke, J.; Negm, A.A.; Lenzen, H.; Manns, M.P.; Lankisch, T.O. Calprotectin in Bile. J. Clin. Gastroenterol. 2014, 48, 866–869. [Google Scholar] [CrossRef]
  38. Sorenson, B.S.; Khammanivong, A.; Guenther, B.D.; Ross, K.F.; Herzberg, M.C. IL-1 Receptor Regulates S100A8/A9-Dependent Keratinocyte Resistance to Bacterial Invasion. Mucosal Immunol. 2012, 5, 66–75. [Google Scholar] [CrossRef]
  39. Hemdan, N.Y.A.; El-Saad, A.M.A.; Sack, U. The Role of T Helper (T H)17 Cells as a Double-Edged Sword in the Interplay of Infection and Autoimmunity with a Focus on Xenobiotic-Induced Immunomodulation. J. Immunol. Res. 2013, 2013, 374769. [Google Scholar] [CrossRef]
  40. Nagareddy, P.R.; Murphy, A.J.; Stirzaker, R.A.; Hu, Y.; Yu, S.; Miller, R.G.; Ramkhelawon, B.; Distel, E.; Westerterp, M.; Huang, L.-S.; et al. Hyperglycemia Promotes Myelopoiesis and Impairs the Resolution of Atherosclerosis. Cell Metab. 2013, 17, 695–708. [Google Scholar] [CrossRef]
  41. Deng, M.; Zhang, W.; Tang, H.; Ye, Q.; Liao, Q.; Zhou, Y.; Wu, M.; Xiong, W.; Zheng, Y.; Guo, X.; et al. Lactotransferrin Acts as a Tumor Suppressor in Nasopharyngeal Carcinoma by Repressing AKT through Multiple Mechanisms. Oncogene 2013, 32, 4273–4283. [Google Scholar] [CrossRef] [PubMed]
  42. Lindholm, M.E.; Fischer, H.; Poellinger, L.; Johnson, R.S.; Gustafsson, T.; Sundberg, C.J.; Rundqvist, H. Negative Regulation of HIF in Skeletal Muscle of Elite Endurance Athletes: A Tentative Mechanism Promoting Oxidative Metabolism. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2014, 307, R248–R255. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, Y.-Y.; Li, X.; Qian, S.-W.; Guo, L.; Huang, H.-Y.; He, Q.; Liu, Y.; Ma, C.-G.; Tang, Q.-Q. Transcriptional Activation of Histone H4 by C/EBPβ during the Mitotic Clonal Expansion of 3T3-L1 Adipocyte Differentiation. Mol. Biol. Cell 2011, 22, 2165–2174. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, J.; Hong, S.J.; Lim, E.K.; Yu, Y.-S.; Kim, S.W.; Roh, J.H.; Do, I.-G.; Joh, J.-W.; Kim, D.S. Expression of Nicotinamide N-Methyltransferase in Hepatocellular Carcinoma Is Associated with Poor Prognosis. J. Exp. Clin. Cancer Res. 2009, 28, 20. [Google Scholar] [CrossRef] [PubMed]
  45. Denizot, J.; Desrichard, A.; Agus, A.; Uhrhammer, N.; Dreux, N.; Vouret-Craviari, V.; Hofman, P.; Darfeuille-Michaud, A.; Barnich, N. Diet-Induced Hypoxia Responsive Element Demethylation Increases CEACAM6 Expression, Favouring Crohn’s Disease-Associated Escherichia coli Colonisation. Gut 2015, 64, 428–437. [Google Scholar] [CrossRef]
  46. Zhu, H.; Hou, L.; Liu, J.; Li, Z. MiR-217 Is down-Regulated in Psoriasis and Promotes Keratinocyte Differentiation via Targeting GRHL2. Biochem. Biophys. Res. Commun. 2016, 471, 169–176. [Google Scholar] [CrossRef]
  47. Vliet-Gregg, P.A.; Hamilton, J.R.; Katzenellenbogen, R.A. Human Papillomavirus 16E6 and NFX1-123 Potentiate Notch Signaling and Differentiation without Activating Cellular Arrest. Virology 2015, 478, 50–60. [Google Scholar] [CrossRef]
  48. Dubash, A.D.; Koetsier, J.L.; Amargo, E.V.; Najor, N.A.; Harmon, R.M.; Green, K.J. The GEF Bcr Activates RhoA/MAL Signaling to Promote Keratinocyte Differentiation via Desmoglein-1. J. Cell Biol. 2013, 202, 653–666. [Google Scholar] [CrossRef]
  49. Lian, X.; Yang, T.; Xiang, M. Regulation of Calcium to Tissue Plasminogen Activator Secretion in Mouse Epidermal Keratinocytes. Colloids Surf. B Biointerfaces 2003, 28, 279–284. [Google Scholar] [CrossRef]
  50. Rabeony, H.; Petit-Paris, I.; Garnier, J.; Barrault, C.; Pedretti, N.; Guilloteau, K.; Jegou, J.-F.; Guillet, G.; Huguier, V.; Lecron, J.-C.; et al. Inhibition of Keratinocyte Differentiation by the Synergistic Effect of IL-17A, IL-22, IL-1α, TNFα and Oncostatin M. PLoS ONE 2014, 9, e101937. [Google Scholar] [CrossRef]
  51. Yang, S.; Sun, Y.; Geng, Z.; Ma, K.; Sun, X.; Fu, X. Abnormalities in the Basement Membrane Structure Promote Basal Keratinocytes in the Epidermis of Hypertrophic Scars to Adopt a Proliferative Phenotype. Int. J. Mol. Med. 2016, 37, 1263–1273. [Google Scholar] [CrossRef] [PubMed]
  52. Cheng, X.; Jin, J.; Hu, L.; Shen, D.; Dong, X.; Samie, M.A.; Knoff, J.; Eisinger, B.; Liu, M.; Huang, S.M.; et al. TRP Channel Regulates EGFR Signaling in Hair Morphogenesis and Skin Barrier Formation. Cell 2010, 141, 331–343. [Google Scholar] [CrossRef] [PubMed]
  53. Sun, X.; Fu, X.; Han, W.; Zhao, Y.; Liu, H.; Sheng, Z. Dedifferentiation of Human Terminally Differentiating Keratinocytes into Their Precursor Cells Induced by Basic Fibroblast Growth Factor. Biol. Pharm. Bull. 2011, 34, 1037–1045. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, Y.; Petreaca, M.; Yao, M.; Martins-Green, M. Cell and Molecular Mechanisms of Keratinocyte Function Stimulated by Insulin during Wound Healing. BMC Cell Biol. 2009, 10, 1. [Google Scholar] [CrossRef]
  55. Giot, J.-P.; Paris, I.; Levillain, P.; Huguier, V.; Charreau, S.; Delwail, A.; Garcia, M.; Garnier, J.; Bernard, F.-X.; Dagregorio, G.; et al. Involvement of IL-1 and Oncostatin M in Acanthosis Associated with Hypertensive Leg Ulcer. Am. J. Pathol. 2013, 182, 806–818. [Google Scholar] [CrossRef]
  56. Tanimizu, N.; Nakamura, Y.; Ichinohe, N.; Mizuguchi, T.; Hirata, K.; Mitaka, T. Hepatic Biliary Epithelial Cells Acquire Epithelial Integrity but Lose Plasticity to Differentiate into Hepatocytes in Vitro during Development. J. Cell Sci. 2013, 126, 5239–5246. [Google Scholar] [CrossRef] [PubMed]
  57. Wittmer, C.R.; Phelps, J.A.; Lepus, C.M.; Saltzman, W.M.; Harding, M.J.; Tassel, P.R.V. Multilayer Nanofilms as Substrates for Hepatocellular Applications. Biomaterials 2008, 29, 4082–4090. [Google Scholar] [CrossRef]
  58. Takehara, T.; Matsumoto, K.; Nakamura, T. Cell Density-Dependent Regulation of Albumin Synthesis and DNA Synthesis in Rat Hepatocytes by Hepatocyte Growth Factor. J. Biochem. 1992, 112, 330–334. [Google Scholar] [CrossRef]
  59. Ishii, K.; Yoshida, Y.; Akechi, Y.; Sakabe, T.; Nishio, R.; Ikeda, R.; Terabayashi, K.; Matsumi, Y.; Gonda, K.; Okamoto, H.; et al. Hepatic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells by Tetracycline-regulated Hepatocyte Nuclear Factor 3β. Hepatology 2008, 48, 597–606. [Google Scholar] [CrossRef]
  60. Huang, J.; Glauber, M.; Qiu, Z.; Gazit, V.; Dietzen, D.J.; Rudnick, D.A. The Influence of Skeletal Muscle on the Regulation of Liver: Body Mass and Liver Regeneration. Am. J. Pathol. 2012, 180, 575–582. [Google Scholar] [CrossRef]
  61. Kamiya, A.; Kinoshita, T.; Miyajima, A. Oncostatin M and Hepatocyte Growth Factor Induce Hepatic Maturation via Distinct Signaling Pathways. FEBS Lett. 2001, 492, 90–94. [Google Scholar] [CrossRef] [PubMed]
  62. Hadas, K.; Randriamboavonjy, V.; Elgheznawy, A.; Mann, A.; Fleming, I. Methylglyoxal Induces Platelet Hyperaggregation and Reduces Thrombus Stability by Activating PKC and Inhibiting PI3K/Akt Pathway. PLoS ONE 2013, 8, e74401. [Google Scholar] [CrossRef] [PubMed]
  63. Soboloff, J.; Sasaki, H.; Tsang, B.K. Follicular Stage-Dependent Tumor Necrosis Factor α-Induced Hen Granulosa Cell Integrin Production and Survival in the Presence of Transforming Growth Factor α In Vitro1. Biol. Reprod. 2001, 65, 477–487. [Google Scholar] [CrossRef]
  64. Zhao, Y.; Garcia, J.; Kolp, L.; Cheadle, C.; Rodriguez, A.; Vlahos, N.F. The Impact of Luteal Phase Support on Gene Expression of Extracellular Matrix Protein and Adhesion Molecules in the Human Endometrium during the Window of Implantation Following Controlled Ovarian Stimulation with a GnRH Antagonist Protocol. Fertil. Steril. 2010, 94, 2264–2271. [Google Scholar] [CrossRef]
  65. Jones, F.S.; Meech, R.; Edelman, D.B.; Oakey, R.J.; Jones, P.L. Prx1 Controls Vascular Smooth Muscle Cell Proliferation and Tenascin-C Expression and Is Upregulated with Prx2 in Pulmonary Vascular Disease. Circ. Res. 2001, 89, 131–138. [Google Scholar] [CrossRef]
  66. Desai, V.D.; Hsia, H.C.; Schwarzbauer, J.E. Reversible Modulation of Myofibroblast Differentiation in Adipose-Derived Mesenchymal Stem Cells. PLoS ONE 2014, 9, e86865. [Google Scholar] [CrossRef]
  67. Maier, S.; Lutz, R.; Gelman, L.; Sarasa-Renedo, A.; Schenk, S.; Grashoff, C.; Chiquet, M. Tenascin-C Induction by Cyclic Strain Requires Integrin-Linked Kinase. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2008, 1783, 1150–1162. [Google Scholar] [CrossRef] [PubMed]
  68. Sarközi, R.; Hauser, C.; Noppert, S.-J.; Kronbichler, A.; Pirklbauer, M.; Haller, V.M.; Grillari, J.; Grillari-Voglauer, R.; Mayer, G.; Schramek, H. Oncostatin M Is a Novel Inhibitor of TGF-Β1-Induced Matricellular Protein Expression. Am. J. Physiol.-Ren. Physiol. 2011, 301, F1014–F1025. [Google Scholar] [CrossRef] [PubMed]
  69. Santiago, J.-J.; McNaughton, L.J.; Koleini, N.; Ma, X.; Bestvater, B.; Nickel, B.E.; Fandrich, R.R.; Wigle, J.T.; Freed, D.H.; Arora, R.C.; et al. High Molecular Weight Fibroblast Growth Factor-2 in the Human Heart Is a Potential Target for Prevention of Cardiac Remodeling. PLoS ONE 2014, 9, e97281. [Google Scholar] [CrossRef] [PubMed]
  70. Wu, J.; Zhang, Y.; Yang, P.; Enkhjargal, B.; Manaenko, A.; Tang, J.; Pearce, W.J.; Hartman, R.; Obenaus, A.; Chen, G.; et al. Recombinant Osteopontin Stabilizes Smooth Muscle Cell Phenotype via Integrin Receptor/Integrin-Linked Kinase/Rac-1 Pathway after Subarachnoid Hemorrhage in Rats. Stroke 2018, 47, 1319–1327. [Google Scholar] [CrossRef]
  71. Horiguchi, K.; Sakamoto, K.; Koinuma, D.; Semba, K.; Inoue, A.; Inoue, S.; Fujii, H.; Yamaguchi, A.; Miyazawa, K.; Miyazono, K.; et al. TGF-β Drives Epithelial-Mesenchymal Transition through ΔEF1-Mediated Downregulation of ESRP. Oncogene 2012, 31, 3190–3201. [Google Scholar] [CrossRef] [PubMed]
  72. Delrieu, I. The High Molecular Weight Isoforms of Basic Fibroblast Growth Factor (FGF-2): An Insight into an Intracrine Mechanism. FEBS Lett. 2000, 468, 6–10. [Google Scholar] [CrossRef] [PubMed]
  73. Sárközy, M.; Szűcs, G.; Pipicz, M.; Zvara, Á.; Éder, K.; Fekete, V.; Szűcs, C.; Bárkányi, J.; Csonka, C.; Puskás, L.G.; et al. The Effect of a Preparation of Minerals, Vitamins and Trace Elements on the Cardiac Gene Expression Pattern in Male Diabetic Rats. Cardiovasc. Diabetol. 2015, 14, 85. [Google Scholar] [CrossRef] [PubMed]
  74. Jamaluddin, M.S.; Yan, S.; Lü, J.; Liang, Z.; Yao, Q.; Chen, C. Resistin Increases Monolayer Permeability of Human Coronary Artery Endothelial Cells. PLoS ONE 2013, 8, e84576. [Google Scholar] [CrossRef]
  75. Qiu, W.; Chen, N.; Zhang, Q.; Zhuo, L.; Wang, X.; Wang, D.; Jin, H. Resistin Increases Platelet P-Selectin Levels via P38 MAPK Signal Pathway. Diabetes Vasc. Dis. Res. 2014, 11, 121–124. [Google Scholar] [CrossRef]
  76. Kuipers, M.T.; Vogl, T.; Aslami, H.; Jongsma, G.; van den Berg, E.; Vlaar, A.P.J.; Roelofs, J.J.T.H.; Juffermans, N.P.; Schultz, M.J.; van der Poll, T.; et al. High Levels of S100A8/A9 Proteins Aggravate Ventilator-Induced Lung Injury via TLR4 Signaling. PLoS ONE 2013, 8, e68694. [Google Scholar] [CrossRef]
  77. Lukanidin, E.; Sleeman, J.P. Building the Niche: The Role of the S100 Proteins in Metastatic Growth. Semin. Cancer Biol. 2012, 22, 216–225. [Google Scholar] [CrossRef]
  78. Lu, S.-M.; Yu, C.-J.; Liu, Y.-H.; Dong, H.-Q.; Zhang, X.; Zhang, S.-S.; Hu, L.-Q.; Zhang, F.; Qian, Y.-N.; Gui, B. S100A8 Contributes to Postoperative Cognitive Dysfunction in Mice Undergoing Tibial Fracture Surgery by Activating the TLR4/MyD88 Pathway. Brain Behav. Immun. 2015, 44, 221–234. [Google Scholar] [CrossRef] [PubMed]
  79. Kostakis, I.D.; Cholidou, K.G.; Kallianidis, K.; Perrea, D.; Antsaklis, A. The Role of Calprotectin in Obstetrics and Gynecology. Eur. J. Obstet. Gynecol. Reprod. Biol. 2010, 151, 3–9. [Google Scholar] [CrossRef]
  80. Iyalomhe, O.; Chen, Y.; Allard, J.; Ntekim, O.; Johnson, S.; Bond, V.; Goerlitz, D.; Li, J.; Obisesan, T.O. A Standardized Randomized 6-Month Aerobic Exercise-Training down-Regulated pro-Inflammatory Genes, but up-Regulated Anti-Inflammatory, Neuron Survival and Axon Growth-Related Genes. Exp. Gerontol. 2015, 69, 159–169. [Google Scholar] [CrossRef]
  81. Ehrchen, J.M.; Sunderkötter, C.; Foell, D.; Vogl, T.; Roth, J. The Endogenous Toll–like Receptor 4 Agonist S100A8/S100A9 (Calprotectin) as Innate Amplifier of Infection, Autoimmunity, and Cancer. J. Leukoc. Biol. 2009, 86, 557–566. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, Y.; Fang, C.; Gao, H.; Bilodeau, M.L.; Zhang, Z.; Croce, K.; Liu, S.; Morooka, T.; Sakuma, M.; Nakajima, K.; et al. Platelet-Derived S100 Family Member Myeloid-Related Protein-14 Regulates Thrombosis. J. Clin. Investig. 2014, 124, 2160–2171. [Google Scholar] [CrossRef] [PubMed]
  83. Liu, X.; Huang, Y.; Pokreisz, P.; Vermeersch, P.; Marsboom, G.; Swinnen, M.; Verbeken, E.; Santos, J.; Pellens, M.; Gillijns, H.; et al. Nitric Oxide Inhalation Improves Microvascular Flow and Decreases Infarction Size after Myocardial Ischemia and Reperfusion. J. Am. Coll. Cardiol. 2007, 50, 808–817. [Google Scholar] [CrossRef]
  84. Ali, H.; Weigmann, B.; Neurath, M.F.; Collnot, E.M.; Windbergs, M.; Lehr, C.-M. Budesonide Loaded Nanoparticles with PH-Sensitive Coating for Improved Mucosal Targeting in Mouse Models of Inflammatory Bowel Diseases. J. Control. Release 2014, 183, 167–177. [Google Scholar] [CrossRef]
  85. Chen, L.Q.; Rohatgi, A.; Ayers, C.R.; Das, S.R.; Khera, A.; Berry, J.D.; McGuire, D.K.; de Lemos, J.A. Race-Specific Associations of Myeloperoxidase with Atherosclerosis in a Population-Based Sample: The Dallas Heart Study. Atherosclerosis 2011, 219, 833–838. [Google Scholar] [CrossRef]
  86. Purushothaman, M.; Krishnan, P.; Purushothaman, K.-R.; Baber, U.; Tarricone, A.; Perez, J.S.; Wiley, J.; Kini, A.; Sharma, S.K.; Fuster, V.; et al. Genotype-Dependent Impairment of Hemoglobin Clearance Increases Oxidative and Inflammatory Response in Human Diabetic Atherosclerosis. Arter. Thromb. Vasc. Biol. 2012, 32, 2769–2775. [Google Scholar] [CrossRef] [PubMed]
  87. Sunil, V.R.; Patel, K.J.; Nilsen-Hamilton, M.; Heck, D.E.; Laskin, J.D.; Laskin, D.L. Acute Endotoxemia Is Associated with Upregulation of Lipocalin 24p3/Lcn2 in Lung and Liver. Exp. Mol. Pathol. 2007, 83, 177–187. [Google Scholar] [CrossRef] [PubMed]
  88. Gouweleeuw, L.; Naudé, P.J.W.; Rots, M.; DeJongste, M.J.L.; Eisel, U.L.M.; Schoemaker, R.G. The Role of Neutrophil Gelatinase Associated Lipocalin (NGAL) as Biological Constituent Linking Depression and Cardiovascular Disease. Brain Behav. Immun. 2015, 46, 23–32. [Google Scholar] [CrossRef]
  89. Lindberg, S.; Jensen, J.S.; Mogelvang, R.; Pedersen, S.H.; Galatius, S.; Flyvbjerg, A.; Magnusson, N.E. Plasma Neutrophil Gelatinase-Associated Lipocalinin in the General Population. Arter. Thromb. Vasc. Biol. 2014, 34, 2135–2142. [Google Scholar] [CrossRef]
  90. Rajnics, P.; Kellner, Á.; Karádi, É.; Moizs, M.; Bödör, C.; Király, P.A.; Marosvári, D.; Andrikovics, H.; Egyed, M. Increased Lipocalin 2 Level May Have Important Role in Thrombotic Events in Patients with Polycythemia Vera and Essential Thrombocythemia. Leuk. Res. 2016, 48, 101–106. [Google Scholar] [CrossRef]
  91. Honda, K.; Littman, D.R. The Microbiome in Infectious Disease and Inflammation. Annu. Rev. Immunol. 2012, 30, 759–795. [Google Scholar] [CrossRef] [PubMed]
  92. Münzer, P.; Walker-Allgaier, B.; Geue, S.; Geuss, E.; Hron, G.; Rath, D.; Eißler, D.; Winter, S.; Schaeffeler, E.; Meinert, M.; et al. PDK1 Determines Collagen-Dependent Platelet Ca2+ Signaling and Is Critical to Development of Ischemic Stroke In Vivo. Arter. Thromb. Vasc. Biol. 2016, 36, 1507–1516. [Google Scholar] [CrossRef] [PubMed]
  93. Maryanoff, B.E.; de Garavilla, L.; Greco, M.N.; Haertlein, B.J.; Wells, G.I.; Andrade-Gordon, P.; Abraham, W.M. Dual Inhibition of Cathepsin G and Chymase Is Effective in Animal Models of Pulmonary Inflammation. Am. J. Respir. Crit. Care Med. 2010, 181, 247–253. [Google Scholar] [CrossRef]
  94. Baldini, M.; Manfredi, A.; Maugeri, N. Targeting Platelet-Neutrophil Interactions in Giant-Cell Arteritis. Curr. Pharm. Des. 2014, 20, 567–574. [Google Scholar] [CrossRef]
  95. Ma, Y.; Yabluchanskiy, A.; Lindsey, M.L. Neutrophil Roles in Left Ventricular Remodeling Following Myocardial Infarction. Fibrogenesis Tissue Repair 2013, 6, 11. [Google Scholar] [CrossRef]
  96. Ando, K.; Hasegawa, K.; Shindo, K.; Furusawa, T.; Fujino, T.; Kikugawa, K.; Nakano, H.; Takeuchi, O.; Akira, S.; Akiyama, T.; et al. Human Lactoferrin Activates NF-κB through the Toll-like Receptor 4 Pathway While It Interferes with the Lipopolysaccharide-Stimulated TLR4 Signaling. FEBS J. 2010, 277, 2051–2066. [Google Scholar] [CrossRef] [PubMed]
  97. Raqib, R.; Mia, S.M.S.; Qadri, F.; Alam, T.I.; Alam, N.H.; Chowdhury, A.K.; Mathan, M.M.; Andersson, J. Innate Immune Responses in Children and Adults with Shigellosis. Infect. Immun. 2000, 68, 3620–3629. [Google Scholar] [CrossRef]
  98. Özsan, H.G.; Pehlıvan, M.; Demırkan, F.; Ündar, B.; Sayan, M.; Çehrelı, C. High Lactoferrin Levels in Disseminated Intravascular Coagulation and Its Possible Negative Role in Coagulation. Thromb. Res. 2000, 98, 111–114. [Google Scholar] [CrossRef]
  99. Grant, L.; Shearer, K.D.; Czopek, A.; Lees, E.K.; Owen, C.; Agouni, A.; Workman, J.; Martin-Granados, C.; Forrester, J.V.; Wilson, H.M.; et al. Myeloid-Cell Protein Tyrosine Phosphatase-1B Deficiency in Mice Protects Against High-Fat Diet and Lipopolysaccharide-Induced Inflammation, Hyperinsulinemia, and Endotoxemia through an IL-10 STAT3-Dependent Mechanism. Diabetes 2014, 63, 456–470. [Google Scholar] [CrossRef]
  100. Coquerel, D.; Neviere, R.; Delile, E.; Mulder, P.; Marechal, X.; Montaigne, D.; Renet, S.; Remy-Jouet, I.; Gomez, E.; Henry, J.-P.; et al. Gene Deletion of Protein Tyrosine Phosphatase 1B Protects Against Sepsis-Induced Cardiovascular Dysfunction and Mortality. Arter. Thromb. Vasc. Biol. 2018, 34, 1032–1044. [Google Scholar] [CrossRef]
  101. Avlan, D.; Taşkinlar, H.; Ünlü, A.; Öztürk, C.; Cinel, L.; Nayci, A.; Cinel, İ.; Aksöyek, S. The Role of Poly(ADP-Ribose) Synthetase Inhibition on the Intestinal Mucosal Barrier after Thermal Injury. Burns 2004, 30, 785–792. [Google Scholar] [CrossRef] [PubMed]
  102. Szabó, C.; Cuzzocrea, S.; Zingarelli, B.; O’Connor, M.; Salzman, A.L. Endothelial Dysfunction in a Rat Model of Endotoxic Shock. Importance of the Activation of Poly (ADP-Ribose) Synthetase by Peroxynitrite. J. Clin. Investig. 1997, 100, 723–735. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, Y.; Ning, B. Signaling Pathways and Intervention Therapies in Sepsis. Signal Transduct. Target. Ther. 2021, 6, 407. [Google Scholar] [CrossRef]
  104. Cao, M.; Wang, G.; Xie, J. Immune Dysregulation in Sepsis: Experiences, Lessons and Perspectives. Cell Death Discov. 2023, 9, 465. [Google Scholar] [CrossRef]
  105. Nedeva, C. Inflammation and Cell Death of the Innate and Adaptive Immune System during Sepsis. Biomolecules 2021, 11, 1011. [Google Scholar] [CrossRef]
  106. Narasimhan, P.B.; Marcovecchio, P.; Hamers, A.A.J.; Hedrick, C.C. Nonclassical Monocytes in Health and Disease. Annu. Rev. Immunol. 2019, 37, 439–456. [Google Scholar] [CrossRef] [PubMed]
  107. Hortová-Kohoutková, M.; Lázničková, P.; Bendíčková, K.; Zuani, M.D.; Andrejčinová, I.; Tomášková, V.; Suk, P.; Šrámek, V.; Helán, M.; Frič, J. Differences in Monocyte Subsets Are Associated with Short-Term Survival in Patients with Septic Shock. J. Cell. Mol. Med. 2020, 24, 12504–12512. [Google Scholar] [CrossRef]
  108. Wang, P.; Yang, S.; Li, C.; Ma, B.; Yi, M.; Chen, X.; Yu, M. Monocyte Subsets along with Their Inflammatory Cytokines in Septic Lung Injury: Anti-Inflammatory Effect of Sulfotransferase Homolog 2 Receptors Blockade. Res. Sq. 2022. [Google Scholar] [CrossRef]
  109. Park, J.; Dean, L.S.; Jiyarom, B.; Gangcuangco, L.M.; Shah, P.; Awamura, T.; Ching, L.L.; Nerurkar, V.R.; Chow, D.C.; Igno, F.; et al. Elevated Circulating Monocytes and Monocyte Activation in COVID-19 Convalescent Individuals. Front. Immunol. 2023, 14, 1151780. [Google Scholar] [CrossRef]
  110. Chen, G.; Wen, D.; Chong, H.; Zhang, P.; Du, J.; Peng, G.; He, Y.; Zhang, K.; Zhang, A.; Deng, J. Value of Peripheral Blood Monocyte Subsets and CD64 Expression in the Diagnosis and Prognosis of Sepsis. Chin. Crit. Care Med. 2022, 34, 921–926. [Google Scholar] [CrossRef]
  111. Greco, M.; Mazzei, A.; Palumbo, C.; Verri, T.; Lobreglio, G. Flow Cytometric Analysis of Monocytes Polarization and Reprogramming from Inflammatory to Immunosuppressive Phase during Sepsis. EJIFCC 2019, 30, 371–384. [Google Scholar] [PubMed]
  112. Skrzeczyñska, J.; Kobylarz, K.; Hartwich, Z.; Zembala, M.; Pryjma, J. CD14+CD16+ Monocytes in the Course of Sepsis in Neonates and Small Children: Monitoring and Functional Studies. Scand. J. Immunol. 2002, 55, 629–638. [Google Scholar] [CrossRef] [PubMed]
  113. Poehlmann, H.; Schefold, J.C.; Zuckermann-Becker, H.; Volk, H.-D.; Meisel, C. Phenotype Changes and Impaired Function of Dendritic Cell Subsets in Patients with Sepsis: A Prospective Observational Analysis. Crit. Care 2009, 13, R119. [Google Scholar] [CrossRef]
  114. Urbanski, K.; Ludew, D.; Filip, G.; Filip, M.; Sagan, A.; Szczepaniak, P.; Grudzien, G.; Sadowski, J.; Jasiewicz-Honkisz, B.; Sliwa, T.; et al. CD14+CD16++ “Nonclassical” Monocytes Are Associated with Endothelial Dysfunction in Patients with Coronary Artery Disease. Thromb. Haemost. 2017, 117, 971–980. [Google Scholar] [CrossRef]
  115. Buscher, K.; Marcovecchio, P.; Hedrick, C.C.; Ley, K. Patrolling Mechanics of Non-Classical Monocytes in Vascular Inflammation. Front. Cardiovasc. Med. 2017, 4, 80. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, C.; Zhang, H.; Wong, W.-C.; Sem, X.; Han, H.; Ong, S.-M.; Tan, Y.-C.; Yeap, W.-H.; Gan, C.-S.; Ng, K.-Q.; et al. Identification of Novel Functional Differences in Monocyte Subsets Using Proteomic and Transcriptomic Methods. J. Proteome Res. 2009, 8, 4028–4038. [Google Scholar] [CrossRef]
  117. Monneret, G. Advancing Our Understanding of Monocyte HLA-DR, S100A9, and the Potential for Individualized Therapies in Sepsis. Mil. Med. Res. 2023, 10, 28. [Google Scholar] [CrossRef]
  118. da Mota, N.V.F.; Brunialti, M.K.C.; Santos, S.S.; Machado, F.R.; Assuncao, M.; Azevedo, L.C.P.; Salomao, R. Immunophenotyping of Monocytes during Human Sepsis Shows Impairment in Antigen Presentation: A Shift Toward Nonclassical Differentiation and Upregulation of FCγRi-Receptor. SHOCK 2018, 50, 293–300. [Google Scholar] [CrossRef]
  119. Teh, Y.C.; Chooi, M.Y.; Chong, S.Z. Behind the Monocyte’s Mystique: Uncovering Their Developmental Trajectories and Fates. Discov. Immunol. 2023, 2, kyad008. [Google Scholar] [CrossRef]
  120. Tamura, A.; Hirai, H.; Yokota, A.; Kamio, N.; Sato, A.; Shoji, T.; Kashiwagi, T.; Torikoshi, Y.; Miura, Y.; Tenen, D.G.; et al. C/EBPβ Is Required for Survival of Ly6C−Monocytes. Blood 2017, 130, 1809–1818. [Google Scholar] [CrossRef]
  121. Mildner, A.; Schönheit, J.; Giladi, A.; David, E.; Lara-Astiaso, D.; Lorenzo-Vivas, E.; Paul, F.; Chappell-Maor, L.; Priller, J.; Leutz, A.; et al. Genomic Characterization of Murine Monocytes Reveals C/EBPβ Transcription Factor Dependence of Ly6C−Cells. Immunity 2017, 46, 849–862.e7. [Google Scholar] [CrossRef] [PubMed]
  122. Ruan, H.; Zhang, Q.; Zhang, Y.; Li, S.; Ran, X. Unraveling the Role of HIF-1α in Sepsis: From Pathophysiology to Potential Therapeutics—A Narrative Review. Crit. Care 2024, 28, 100. [Google Scholar] [CrossRef] [PubMed]
  123. Codo, A.C.; Davanzo, G.G.; Monteiro, L.d.B.; de Souza, G.F.; Muraro, S.P.; Virgilio-da-Silva, J.V.; Prodonoff, J.S.; Carregari, V.C.; Junior, C.A.O.d.B.; Crunfli, F.; et al. Elevated Glucose Levels Favor SARS-CoV-2 Infection and Monocyte Response through a HIF-1α/Glycolysis-Dependent Axis. Cell Metab. 2020, 32, 437–446.e5. [Google Scholar] [CrossRef] [PubMed]
  124. Iske, J.; Fatimy, R.E.; Nian, Y.; Ghouzlani, A.; Eskandari, S.K.; Biefer, H.R.C.; Vasudevan, A.; Elkhal, A. NAD+ Prevents Septic Shock-Induced Death by Non-Canonical Inflammasome Blockade and IL-10 Cytokine Production in Macrophages. eLife 2024, 12, RP88686. [Google Scholar] [CrossRef]
  125. Chen, Y.; Ye, X.; Escames, G.; Lei, W.; Zhang, X.; Li, M.; Jing, T.; Yao, Y.; Qiu, Z.; Wang, Z.; et al. The NLRP3 Inflammasome: Contributions to Inflammation-Related Diseases. Cell. Mol. Biol. Lett. 2023, 28, 51. [Google Scholar] [CrossRef]
  126. Busani, S.; Biasi, S.D.; Nasi, M.; Paolini, A.; Venturelli, S.; Tosi, M.; Girardis, M.; Cossarizza, A. Increased Plasma Levels of Mitochondrial DNA and Normal Inflammasome Gene Expression in Monocytes Characterize Patients with Septic Shock Due to Multidrug Resistant Bacteria. Front. Immunol. 2020, 11, 768. [Google Scholar] [CrossRef]
  127. Xue, Z.; Xi, Q.; Liu, H.; Guo, X.; Zhang, J.; Zhang, Z.; Li, Y.; Yang, G.; Zhou, D.; Yang, H.; et al. MiR-21 Promotes NLRP3 Inflammasome Activation to Mediate Pyroptosis and Endotoxic Shock. Cell Death Dis. 2019, 10, 461. [Google Scholar] [CrossRef]
  128. Yang, L.; Gao, Q.; Li, Q.; Guo, S. PD-L1 Blockade Improves Survival in Sepsis by Reversing Monocyte Dysfunction and Immune Disorder. Inflammation 2024, 47, 114–128. [Google Scholar] [CrossRef]
  129. Santos, S.S.; Carmo, A.M.; Brunialti, M.K.C.; Machado, F.R.; Azevedo, L.C.; Assunção, M.; Trevelin, S.C.; Cunha, F.Q.; Salomao, R. Modulation of Monocytes in Septic Patients: Preserved Phagocytic Activity, Increased ROS and NO Generation, and Decreased Production of Inflammatory Cytokines. Intensive Care Med. Exp. 2016, 4, 5. [Google Scholar] [CrossRef]
  130. Iba, T.; Helms, J.; Connors, J.M.; Levy, J.H. The Pathophysiology, Diagnosis, and Management of Sepsis-Associated Disseminated Intravascular Coagulation. J. Intensiv. Care 2023, 11, 24. [Google Scholar] [CrossRef]
  131. Bode, C.; Weis, S.; Sauer, A.; Wendel-Garcia, P.; David, S. Targeting the Host Response in Sepsis: Current Approaches and Future Evidence. Crit. Care 2023, 27, 478. [Google Scholar] [CrossRef] [PubMed]
  132. Kellum, J.A.; Ronco, C. The Role of Endotoxin in Septic Shock. Crit. Care 2023, 27, 400. [Google Scholar] [CrossRef] [PubMed]
  133. Sirivongrangson, P.; Kulvichit, W.; Payungporn, S.; Pisitkun, T.; Chindamporn, A.; Peerapornratana, S.; Pisitkun, P.; Chitcharoen, S.; Sawaswong, V.; Worasilchai, N.; et al. Endotoxemia and Circulating Bacteriome in Severe COVID-19 Patients. Intensive Care Med. Exp. 2020, 8, 72. [Google Scholar] [CrossRef] [PubMed]
  134. Foster, D.M.; Kellum, J.A. Endotoxic Septic Shock: Diagnosis and Treatment. Int. J. Mol. Sci. 2023, 24, 16185. [Google Scholar] [CrossRef]
  135. Ishijima, T.; Nakajima, K. Inflammatory Cytokines TNFα, IL-1β, and IL-6 Are Induced in Endotoxin-Stimulated Microglia through Different Signaling Cascades. Sci. Prog. 2021, 104, 00368504211054985. [Google Scholar] [CrossRef]
  136. Turner, M.D.; Nedjai, B.; Hurst, T.; Pennington, D.J. Cytokines and Chemokines: At the Crossroads of Cell Signalling and Inflammatory Disease. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2014, 1843, 2563–2582. [Google Scholar] [CrossRef]
  137. Brown, G.C. The Endotoxin Hypothesis of Neurodegeneration. J. Neuroinflamm. 2019, 16, 180. [Google Scholar] [CrossRef]
  138. Bhan, C.; Dipankar, P.; Chakraborty, P.; Sarangi, P.P. Role of Cellular Events in the Pathophysiology of Sepsis. Inflamm. Res. 2016, 65, 853–868. [Google Scholar] [CrossRef]
  139. Bouck, E.G.; Denorme, F.; Holle, L.A.; Middelton, E.A.; Blair, A.M.; de Laat, B.; Schiffman, J.D.; Yost, C.C.; Rondina, M.T.; Wolberg, A.S.; et al. COVID-19 and Sepsis Are Associated with Different Abnormalities in Plasma Procoagulant and Fibrinolytic Activity. Arter. Thromb. Vasc. Biol. 2020, 41, 401–414. [Google Scholar] [CrossRef]
  140. Immanuel, J.; Yun, S. Vascular Inflammatory Diseases and Endothelial Phenotypes. Cells 2023, 12, 1640. [Google Scholar] [CrossRef]
  141. Silva, J.C.; Gorenstein, M.V.; Li, G.-Z.; Vissers, J.P.C.; Geromanos, S.J. Absolute Quantification of Proteins by LCMSE A Virtue of Parallel ms Acquisition * S. Mol. Cell. Proteom. 2006, 5, 144–156. [Google Scholar] [CrossRef] [PubMed]
  142. Cassoli, J.S.; Brandão-Teles, C.; Santana, A.G.; Souza, G.H.M.F.; Martins-de-Souza, D. Ion Mobility-Enhanced Data-Independent Acquisitions Enable a Deep Proteomic Landscape of Oligodendrocytes. Proteomics 2017, 17, 1700209. [Google Scholar] [CrossRef] [PubMed]
  143. Distler, U.; Kuharev, J.; Navarro, P.; Levin, Y.; Schild, H.; Tenzer, S. Drift Time-Specific Collision Energies Enable Deep-Coverage Data-Independent Acquisition Proteomics. Nat. Methods 2014, 11, 167–170. [Google Scholar] [CrossRef]
  144. Geromanos, S.J.; Hughes, C.; Ciavarini, S.; Vissers, J.P.C.; Langridge, J.I. Using Ion Purity Scores for Enhancing Quantitative Accuracy and Precision in Complex Proteomics Samples. Anal. Bioanal. Chem. 2012, 404, 1127–1139. [Google Scholar] [CrossRef]
  145. Silva, J.C.; Denny, R.; Dorschel, C.A.; Gorenstein, M.; Kass, I.J.; Li, G.-Z.; McKenna, T.; Nold, M.J.; Richardson, K.; Young, P.; et al. Quantitative Proteomic Analysis by Accurate Mass Retention Time Pairs. Anal. Chem. 2005, 77, 2187–2200. [Google Scholar] [CrossRef]
  146. Giles, K.; Pringle, S.D.; Worthington, K.R.; Little, D.; Wildgoose, J.L.; Bateman, R.H. Applications of a Travelling Wave-Based Radio-Frequency-only Stacked Ring Ion Guide. Rapid Commun. Mass Spectrom. 2004, 18, 2401–2414. [Google Scholar] [CrossRef]
  147. Souza, G.H.M.F.; Guest, P.C.; Martins-de-Souza, D. LC-MSE, Multiplex MS/MS, Ion Mobility, and Label-Free Quantitation in Clinical Proteomics. Methods Mol. Biol. 2016, 1546, 57–73. [Google Scholar] [CrossRef]
  148. Houel, S.; Abernathy, R.; Renganathan, K.; Meyer-Arendt, K.; Ahn, N.G.; Old, W.M. Quantifying the Impact of Chimera MS/MS Spectra on Peptide Identification in Large-Scale Proteomics Studies. J. Proteome Res. 2010, 9, 4152–4160. [Google Scholar] [CrossRef]
  149. Silva, W.M.; Folador, E.L.; Soares, S.C.; Souza, G.H.; Santos, A.V.; Sousa, C.S.; Figueiredo, H.; Miyoshi, A.; Le Loir, Y.; Silva, A.; et al. Label-free quantitative proteomics of Corynebacterium pseudotuberculosis isolates reveals differences between Biovars ovis and equi strains. BMC Genom. 2017, 18, 451. [Google Scholar] [CrossRef]
Figure 1. The biological components and interactions of differentially expressed proteins. The major cellular components involved are downregulated (A) and upregulated (B). The main biological processes participating in differentially expressed proteins are (C) down- and (D) upregulated. The interactions between differentially expressed proteins are not direct. However, they influence several molecules that communicate directly. Blue represents downregulated genes, red indicates upregulated genes, and green indicates the interaction genes of the FunRich database (E).
Figure 1. The biological components and interactions of differentially expressed proteins. The major cellular components involved are downregulated (A) and upregulated (B). The main biological processes participating in differentially expressed proteins are (C) down- and (D) upregulated. The interactions between differentially expressed proteins are not direct. However, they influence several molecules that communicate directly. Blue represents downregulated genes, red indicates upregulated genes, and green indicates the interaction genes of the FunRich database (E).
Ijms 25 10641 g001
Table 1. Differentially expressed proteins identified in the monocytes of patients with septic shock compared with healthy controls.
Table 1. Differentially expressed proteins identified in the monocytes of patients with septic shock compared with healthy controls.
GeneAccessionDescriptionScoreANOVA (p)RatioN 1C 1M 1S 1R 2
ANXA3P12429Annexin A3171.366.04 × 10−63.046 +
HIST1H2ACQ93077Histone H2A type 1-C44.507.00 × 10−63.087 +
FLNBO75369Filamin-B155.763.045 × 10−83.121 +
GXYLT2A0PJZ3Glucoside xylosyltransferase 29.126.82 × 10−53.122 +
IGSF10Q6WRI0Immunoglobulin superfamily member 1063.372.88 × 10−63.125 +
WDR90Q96KV7WD repeat-containing protein 9046.320.00023.146 +
ZEB2O60315Zinc finger E-box-binding homeobox 27.800.00013.167 +
EPRSP07814Bifunctional glutamate/proline--tRNA ligase19.409.10 × 10−53.168 +
HYDINQ4G0P3Hydrocephalus-inducing protein homolog91.085.43 × 10−63.187 +
ADGRD1Q6QNK2Adhesion G-protein coupled receptor D113.295.89 × 10−63.239 +
RETNQ9HD89Resistin24.980.00013.251 +
LCN2P80188Neutrophil gelatinase-associated lipocalin53.161.11 × 10−63.255 +
PDPK1O155303-phosphoinositide-dependent protein kinase 115.670.00013.284 +
NNMTP40261Nicotinamide N-methyltransferase13.641.56 × 10−53.286 +
HIST1H2BKO60814Histone H2B type 1-K68.545.53 × 10−53.356 +
RNASE3P12724Eosinophil cationic protein35.480.00023.359 +
ATP6V1B1C9JL73V-type proton ATPase subunit B, kidney isoform45.812.28 × 10−63.436 +
DLC1Q96QB1Rho GTPase-activating protein 710.113.05 × 10−63.452 +
HIST1H2BBP33778Histone H2B type 1-B75.936.93 × 10−63.540 +
MLH3Q9UHC1DNA mismatch repair protein Mlh336.534.81 × 10−73.599 +
ZNF133P52736Zinc finger protein 1337.840.00013.603 +
SACM1LQ9NTJ5Phosphatidylinositide phosphatase SAC125.196.30 × 10−63.625 +
TLN2Q9Y4G6Talin-2139.601.56 × 10−53.833 +
LTFP02788Lactotransferrin207.355.46 × 10−64.066 +
PPP1CBP62140Serine/threonine–protein phosphatase PP1-beta catalytic subunit36.261.12 × 10−54.134 +
S100A8P05109Protein S100-A868.053.52 × 10−54.458 +
PTPN1B4DSN5Tyrosine–protein phosphatase non-receptor type25.300.00014.506 +
KLBQ86Z14Beta-klotho15.356.19 × 10−64.515 +
ANAPC5F5H0F9Anaphase-promoting complex subunit 514.877.71 × 10−54.525 +
CTSGP08311Cathepsin G137.542.36 × 10−74.539 +
CHTF8P0CG12Chromosome transmission fidelity protein 8 homolog isoform 223.381.11 × 10−55.026 +
C8BP07358Complement component C8 beta 13.871.36 × 10−65.059 +
MPOP05164Myeloperoxidase350.671.19 × 10−65.399 +
EFTUD2Q15029116 kDa U5 small nuclear ribonucleoprotein component51.233.93 × 10−65.491 +
K7ERQ8Uncharacterized protein (Fragment)18.181.15 × 10−55.579 +
TUBGCP5Q96RT8Gamma-tubulin complex component9.594.57 × 10−55.580 +
FAM167AQ96KS9Protein FAM167A15.102.78 × 10−66.212 +
NRCAMC9JYY6Neuronal cell adhesion molecule8.182.43 × 10−56.232 +
S100A9P06702Protein S100-A9141.733.65 × 10−67.217 +
MATR3P43243Matrin-331.476.77 × 10−67.618 +
HIST1H4AP62805Histone H4132.302.00 × 10−610.050 +
CEACAM6P40199Carcinoembryonic antigen-related cell adhesion molecule 628.451.75 × 10−511.350 +
KIR2DL2Q8N742KIR2DL214.601.18 × 10−511.410 +
NUDCQ9Y266Nuclear migration protein nudC15.403.04 × 10−7611.518 +
SH3TC2E9PDF1SH3 domain and tetratricopeptide repeat-containing protein 228.982.27 × 10−618.110
KRT9P35527Keratin, type I cytoskeletal 910.621.42 × 10−79.427
ACP1P24666Low-molecular-weight phosphotyrosine protein phosphatase9.941.08 × 10−55.852
KRT1P04264Keratin, type II cytoskeletal 157.806.76 × 10−75.782
KRT10P13645Keratin, type I cytoskeletal 10137.216.77 × 10−65.695
LZTR1Q8N653Leucine-zipper-like transcriptional regulator 111.914.68 × 10−64.618
ATP13A3Q9H7F0Probable cation-transporting ATPase 13A328.151.61 × 10−54.614
ZNF677Q86XU0Zinc finger protein 6777.784.14 × 10−54.543
ITGB3P05106Integrin beta-3174.142.37 × 10−64.476
TNCP24821Tenascin36.378.73 × 10−64.307
ZFHX2Q9C0A1Zinc finger homeobox protein 214.110.00014.104
ESRP2Q9H6T0Epithelial splicing regulatory protein 216.191.7 × 10−54.011
MYH10P35580Myosin-1076.793.25 × 10−53.879
COG4Q9H9E3Conserved oligomeric Golgi complex subunit 414.500.00013.870
PSMC4P4368626S protease regulatory subunit 6B10.908.76 × 10−53.762
ATAD2BQ9ULI0ATPase family AAA domain-containing protein 2B13.668.72 × 10−53.703
PGK2P07205Phosphoglycerate kinase 294.212.49 × 10−63.701
ALBP02768Serum albumin164.054.34 × 10−53.670
ACAD9Q9H845Acyl-CoA dehydrogenase family member 9, mitochondrial14.604.86 × 10−63.262
CCT8P50990T-complex protein 1 subunit theta76.198.92 × 10−63.255
HMGB2P26583High-mobility group protein B263.462.43 × 10−53.059
CLCN4P51793H(+)/Cl(−) exchange transporter 48.300.00032.927
MYO5G3V394Unconventional myosin-Va128.701.25 × 10−62.855
1 Black squares indicate cellular location: N (nucleus); M (membrane); C (cytoplasm); S (secreted); 2 protein regulation: + (upregulation); − (downregulation).
Table 2. The primary regulation of proteins with increased expression in septic shock.
Table 2. The primary regulation of proteins with increased expression in septic shock.
ProteinEffectorEffectRef.
LTFCEBPECEBPE is involved in the positive regulation of lactoferrin gene expression[16]
CEBPAC/EBPα binds to the C/EBP site in the lactoferrin promoter in induced myeloid cells[17]
MPOCEBPEUpregulation of CBPE induces expression of myeloperoxidase[18]
LTFLTF decreased the serum C-reactive protein level and the inducible nitric oxide synthase (iNOS) and myeloperoxidase (MPO) gene expression levels[19]
HIF-1Activation of HIF-1 by P4HA2 gene silencing attenuated myeloperoxidase expression in myocardium following ischemia–reperfusion[20]
IL1-RAInterleukin-1ra and anti-TNF-α also significantly lowered MPO levels[21]
CEBPAC/EBPα inhibits monocyte/macrophage differentiation and initiates granulocyte differentiation by inducing myeloperoxidase gene expressions[22]
RETNCEBPEHuman leukocyte resistin expression depends on the myeloid-specific nuclear transcription factor CEBPE[23]
CEBPBResistin stimulates the expression of chemokine genes in chondrocytes via the combinatorial regulation of C/EBPβ and NF-κB.[24]
HIF-1Hypoxia-inducible factor-1 results in the increased production of leptin, resistin, TNF, and IL-6[25]
IL1-RAIL-1Ra treatment for reduction in leptin and resistin levels[26]
CEBPAResistin gene promoter carries the C/EBP-α binding site, which is necessary and sufficient for transcription from the resistin gene promoter[27]
LCN2CEBPEStable inducible expression of CEBPE in the murine fibroblast cell line NIH 3T-activated expression of mRNA LCN2[28]
INFLAMMASOMEInflammasome-mediated production of antimicrobial peptides, including Reg3β, Reg3γ, S100A8, S100A9, and LCN2[29]
CEBPBLCN2 promoter region contains the binding sites of several transcription factors such as NF-κB, STAT1, STAT3, and CEBPB[30]
IL17IL17 positively regulates LCN2 expression. [31]
CEBPAIL6 and LCN2 promoters require both NF-κB and C/EBP elements[32]
S100A8S100A9S100A3S100A3 co-expression inhibits AP-1 and NF-κB-dependent transcription upon S100A8 and S100A9 over-expression[33]
INFLAMMASOMEInflammasome-mediated production of antimicrobial peptides, including Reg3β, Reg3γ, S100A8, S100A9, and LCN4[29]
LTFLactoferrin induces the production of chemokines (MIP-1a, MCP-1, and S100A9)[34]
CEBPBIL-1α-induced S100A9 expression is signaled through the IL-1 receptor and p38 MAPK pathways, resulting in the binding of CEBPB to the upstream S100A9 promoter[35,36]
HIF-1Hypoxia and HIF-1 increase S100A8 and S100A9 expression[37]
IL1-RAIL-1Ra inhibited interleukin-1 α-induced S100A8 and S100A9 gene expression[38]
IL17FIL-17 stimulates the expression of human beta-defensin- 2, S100A9 and enhances the expression of S100A7 and S100A9[39]
CEBPAIncrease in the presence of reactive oxygen species and the expression levels of the transcription factors Klf-5 and CEBPA in neutrophils, both of which promote S100A8/S100A9 expression [40]
PDPK1LTFLactotransferrin downregulates the level of 3-phosphoinositide-dependent protein kinase 1 (PDK1) transcription and subsequently inhibits other proteins[41]
HIF-1Repression of hypoxia-inducible factor-1 activity, attenuate PDK-1 expression [42]
HIST4H4 CEBPBCEBPB can significantly transactivate the expression of HIST4H4[43]
NNMTCEBPBNNMT promoter region also contains the consensus sequences for signal transducers and activators of transcription binding elements[44]
CEACAM6HIF-1Hypoxia-inducible factor-1 transcription factor increases CEACAM6 expression in intestinal epithelial cells[45]
Table 3. The main regulation of proteins with low expression in septic shock.
Table 3. The main regulation of proteins with low expression in septic shock.
ProteinEffectorFunction Protein EffectorEffectRef.
KRT1GRHL2Transcription factor Downregulate KRT1 and KRT10[46]
NFX1Nuclear transcription factor Upregulate KRT1 and KRT10[47]
BCRBreakpoint cluster region proteinLoss of BCR reduces expression of KRT1 and KRT10[48]
PLATPlasminogen activator, tissuePlays a role in the expression of KRT1 and KRT10[49]
OSMOncostatin MDownregulate mRNA expression [50]
KRT10GRHL2Transcription factorDownregulate KRT1 and KRT10[46]
NFX1Nuclear transcription factorUpregulate KRT1 and KRT10[47]
COLLAGENMatrix proteinDownregulate the mRNA expression KRT10[51]
BCRBreakpoint cluster region proteinLoss of BCR reduces expression of KRT1 and KRT10[48]
TGFATransforming grown factorSuppress KRT10 expression, promoted late terminal differentiation[52]
FGF2Fibroblast growth factor 2Downregulate the KRT10 expression[53]
PLATPlasminogen activator, tissuePlays a role in the expression of KRT1 and KRT10[49]
ITGA3Integrin alpha 3Inhibition KRT10 production[54]
OSMOncostatin MDecrease expression[55]
ABLGRHL2Transcription factorInhibits expression ABL[56]
COLLAGENMatrix proteinInhibit albumin production at short times, but enhances albumin production at longer times[57]
TGFATransforming grown factorStimulate albumin synthesis[58]
FGF2Fibroblast growth factor 2Induce expression[59]
ILKIntegrin like kinaseALB mRNA expression is downregulated by ILK[60]
OSMOncostatin MUpregulate production[61]
ITGB3COLLAGENMatrix proteinEnhance β3 integrin tyrosine phosphorylation by adhesion platelets to collagen[62]
TGFATransforming grown factorStimulate ITGB3 expression[63]
ITGA3Integrin alpha 3Enhance the expression[64]
TNCCOLLAGENMatrix proteinTenascin-C mRNA expression is reduced by native collagen and is upregulated by denatured collagen[65]
FGF2Fibroblast growth factor 2Upregulator of tenascin expression and activation[66]
ILKIntegrin like kinaseInduce expression TNC[67]
OSMOncostatin MInhibit mRNA expression[68]
MYH10FGF2Fibroblast growth factor 2Decrease MYH10 expression [69]
ILKIntegrin like kinaseRegulate MYH10 expression[70]
ESRP2FGF2Fibroblast growth factor 2Repress the levels of ESRP2 mRNA [71]
PGK2FGF2Fibroblast growth factor 2Modulate transcription of PGK-2 genes[72]
Table 4. Systemic effect of secreted proteins upregulation on septic shock.
Table 4. Systemic effect of secreted proteins upregulation on septic shock.
ProteinEffectActionRef.
RETNInflammationMediated chronic inflammation can lead to atherosclerosis, and other cardiometabolic diseases[73]
Vascular damageMajor inducer of endothelial damage through the induction of permeability[74]
ThrombosisResistin an adipokine associated with the metabolic syndrome is believed to have a role in thrombotic conditions.[75]
S100A8EndotoxemiaS100A8 administration attenuated inflammation and injury in a mouse model of endotoxemia[76]
InflammationS100A8 important mediators of various processes during chronic inflammation[77]
Vascular damageHigh S100A8 expression leads to endothelial damage by inducing the apoptosis and death of endothelial cells [78]
ThrombosisS100A8 which is secreted by trophoblast cells probably regulates the level of macrophage activation and procoagulant factors[79]
S100A9InflammationS100A9, an important pro-inflammatory mediator in acute and chronic inflammation[80]
Vascular damageS100A8/S100A9 is released in high amounts at sites of inflammation, S100A8/S100A9-induced endothelial damage [81]
ThrombosisPlatelet-derived S100 family member myeloid-related protein-14 regulates thrombosis[82]
MPOEndotoxemiaMPO can aggravate this inflammatory response in rodent models of endotoxemia[83]
InflammationMPO plays an important role in the initiation and progression of acute and chronic inflammation.[84]
Vascular damageMPO consumes nitric oxide, leading to vasoconstriction and promoting endothelial damage[85]
ThrombosisHemoglobin-Hp2-2 complexes may promote a pro-inflammatory macrophage phenotype via oxidative mechanisms (MPO) leading to plaque destabilization and atherothrombosis[86]
LCN2EndotoxemiaAcute endotoxemia is associated with upregulation of lipocalin 24p3/Lcn2 in lung and liver[87]
InflammationLCN2 is involved in chronic inflammation [88]
Vascular damageLCN2 supposedly mediates vascular damage and plaque rupture.[89]
ThrombosisLCN2 could have an important role in thrombotic events associated with polycythemia vera and essential thrombocythemia[90]
PDPK1InflammationDeletion of PDPK1 induces chronic inflammation of the intestine[91]
ThrombosisPDK1 is required for Ca(2+)-dependent platelet activation on stimulation of collagen receptor glycoprotein VI, arterial thrombotic occlusion, and ischemic stroke in vivo[92]
CTSGInflammationCTSG is thought to contribute to self-propagating, chronic inflammation.[93]
Vascular damageCTSG causes the activation of bystander platelets, enhances vascular damage and inhibits fibrinolysis [94]
ThrombosisCTSG is a potent platelet activator and promotes intravascular thrombosis, thus contributing to the formation of a thrombus clot[95]
LTFEndotoxemiaLactoferrin suppresses endotoxemia by interfering with lipopolysaccharide dependent TLR4 activation[96]
InflammationPersistent production of lactoferrin in pediatric patients may contribute to chronic inflammation in the rectum[97]
ThrombosisLactoferrin may play a role in the pathogenesis of disseminated intravascular coagulation and thrombotic complications[98]
PTPN1EndotoxemiaPTP1 not protect lipopolysaccharide-induced inflammation, hyperinsulinemia, and endotoxemia through an IL-10 STAT3-dependent mechanism.[99]
Vascular damageCytokines, ROS, and COX trigger an acute inflammatory response and induce vascular damage that may be reduced by PTP1B deletion[100]
EPRSEndotoxemiaEPRS inhibition has beneficial effects against organ dysfunction due to reperfusion injury and endotoxemia[101]
Vascular damagePARS activation plays a role in the pathogenesis of endothelial injury in endotoxin shock.[102]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alves, P.T.; de Souza, A.G.; Bastos, V.A.F.; Miguel, E.L.; Ramos, A.C.S.; Cameron, L.C.; Goulart, L.R.; Cunha, T.M. The Modulation of Septic Shock: A Proteomic Approach. Int. J. Mol. Sci. 2024, 25, 10641. https://doi.org/10.3390/ijms251910641

AMA Style

Alves PT, de Souza AG, Bastos VAF, Miguel EL, Ramos ACS, Cameron LC, Goulart LR, Cunha TM. The Modulation of Septic Shock: A Proteomic Approach. International Journal of Molecular Sciences. 2024; 25(19):10641. https://doi.org/10.3390/ijms251910641

Chicago/Turabian Style

Alves, Patrícia Terra, Aline Gomes de Souza, Victor Alexandre F. Bastos, Eduarda L. Miguel, Augusto César S. Ramos, L. C. Cameron, Luiz Ricardo Goulart, and Thúlio M. Cunha. 2024. "The Modulation of Septic Shock: A Proteomic Approach" International Journal of Molecular Sciences 25, no. 19: 10641. https://doi.org/10.3390/ijms251910641

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop