Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Immunobiology ; 229(4): 152822, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852289

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy that represents a significant challenge in cancer research and clinical management. In this study, we reanalyzed a published single-cell RNA sequencing (scRNA-seq) dataset from PDAC and adjacent tissues to investigate the heterogeneity of tumor and normal tissue, specifically focusing on the regulatory T cells (Tregs) and their interactions with other cells in the tumor microenvironment (TME). Treg cells were identified and clustered into natural Tregs (nTreg) and induced Tregs (iTreg) based on the expression of specific genes. It was found that the number of iTregs was higher in the tumor than in healthy tissues, while the number of n Tregs was higher in healthy tissues. Differential gene expression analysis was performed, and biological process analysis revealed that the Tregs in PDAC were mostly involved in protein targeting and translation pathways. In addition, ligand-receptor pairs between Tregs and other cell types were identified, and the critical communication pathways between Tregs and endothelial and ductal cells were revealed, which could potentially contribute to the immunosuppressive TME of PDAC. These findings provide insights into the role of Tregs in PDAC and their interactions with other cell types in the TME, highlighting potential targets for immunotherapy, such as the inhibitory immune checkpoint receptors CTLA4 and TIGIT, which are known to be expressed on Tregs and have been shown to play a role in suppressing anti-tumor immune responses.


Subject(s)
CTLA-4 Antigen , Carcinoma, Pancreatic Ductal , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms , Receptors, Immunologic , Single-Cell Analysis , T-Lymphocytes, Regulatory , Tumor Microenvironment , Humans , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/genetics , T-Lymphocytes, Regulatory/immunology , CTLA-4 Antigen/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Single-Cell Analysis/methods , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/genetics , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Sequence Analysis, RNA , Gene Expression Profiling , Transcriptome
2.
Int Immunopharmacol ; 137: 112407, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38875996

ABSTRACT

AIM: Sepsis is a life-threatening condition caused by a dysregulated immune response to infection. Broad-spectrum antibiotics are used to treat it. However, due to antibiotic resistance, alternative treatments are needed. Mesenchymal stem cells (MSCs) have become a promising therapeutic tool for sepsis due to their immunomodulatory properties. The limitations of MSC therapy have led to increased attention to cell derivatives such as conditioned medium (CM). This study investigates the immunomodulatory effects of young and old MSC-CM during the inflammatory phase of sepsis. MAIN METHODS: The cecal ligation and puncture (CLP) model was used to induce sepsis in mice. The mice were divided into four groups: sham, CLP, CLP treated with young MSC-CM, and CLP treated with old MSC-CM. The CM was injected intraperitoneally at 2-, 12-, and 24-hours post-surgery. After 72 h, blood was collected and white blood cells (WBCs) were counted. In addition, serum and tissue were isolated, and the levels of alanine transaminase (ALT) and aspartate transaminase (AST) in serum, bacterial load in the spleen, concentration of pro- and anti-inflammatory cytokines, and histopathology of liver and lung were investigated. KEY FINDINGS: MSC-CM decreased serum AST and ALT levels, bacterial load in the spleen, and pro-inflammatory cytokines in serum. In addition, tissue damage was reduced, and the survival rate and WBC count increased. There was no significant difference between the young and old MSC-CM. SIGNIFICANCE: MSC-CM effectively reduced inflammation-induced tissue damage in the liver and lungs during sepsis. Although young MSC-CM had better immunomodulatory effects than old MSC-CM, the difference was not significant.

3.
Hum Immunol ; 85(3): 110798, 2024 May.
Article in English | MEDLINE | ID: mdl-38569354

ABSTRACT

BACKGROUND: Antigen-specific T-cell immunity is provided by dendritic cells (DCs), which are specialized antigen-presenting cells. Furthermore, they establish a link between innate and adaptive immune responses. Currently, DC modification is a new approach for the therapy of several disorders. During solid organ transplantation, Everolimus, which is a mammalian target of rapamycin (mTOR) inhibitor, was initially utilized to suppress the immune system's functionality. Due to the intervention of Everolimus in various signaling pathways in cells and its modulatory properties on the immune system, this study aims to investigate the effect of treatment with Everolimus on the maturation and expression of immune checkpoint genes in monocyte-derived DCs. METHODS: To isolate monocytes from PBMCs, the CD14 marker was used via the MACS method. Monocytes were cultured and induced to differentiate into monocyte-derived DCs by utilizing GM-CSF and IL-4 cytokines. On the fifth day, immature DCs were treated with Everolimus and incubated for 24 h. On the sixth day, the flow cytometry technique was used to investigate the effect of Everolimus on the phenotypic characteristics of DCs. In the end, the expression of immune checkpoint genes in both the Everolimus-treated and untreated DCs groups was assessed using the real-time PCR method. RESULTS: The findings of this research demonstrated that the administration of Everolimus to DCs led to a notable rise in human leukocyte antigen (HLA)-DR expression and a decrease in CD11c expression. Furthermore, there was a significant increase in the expression of immune checkpoint molecules, namely CTLA-4, VISTA, PD-L1, and BTLA, in DCs treated with Everolimus. CONCLUSION: The findings of this study show that Everolimus can target DCs and affect their phenotype and function in order to shift them toward a partially tolerogenic state. However, additional research is required to gain a comprehensive understanding of the precise impact of Everolimus on the activation status of DCs.


Subject(s)
Cell Differentiation , Dendritic Cells , Everolimus , Monocytes , Humans , Dendritic Cells/immunology , Dendritic Cells/drug effects , Everolimus/pharmacology , Monocytes/immunology , Monocytes/drug effects , Cells, Cultured , Cell Differentiation/drug effects , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology
4.
PLoS One ; 18(12): e0295863, 2023.
Article in English | MEDLINE | ID: mdl-38096229

ABSTRACT

Immunotherapy is changing the Head and Neck Squamous Cell Carcinoma (HNSCC) landscape and improving outcomes for patients with recurrent or metastatic HNSCC. A deeper understanding of the tumor microenvironment (TME) is required in light of the limitations of patients' responses to immunotherapy. Here, we aimed to examine how Nivolumab affects infiltrating Tregs in the HNSCC TME. We used single-cell RNA sequencing data from eight tissues isolated from four HNSCC donors before and after Nivolumab treatment. Interestingly, the study found that Treg counts and suppressive activity increased following Nivolumab therapy. We also discovered that changes in the CD44-SSP1 axis, NKG2C/D-HLA-E axis, and KRAS signaling may have contributed to the increase in Treg numbers. Furthermore, our study suggests that decreasing the activity of the KRAS and Notch signaling pathways, and increasing FOXP3, CTLA-4, LAG-3, and GZMA expression, may be mechanisms that enhance the killing and suppressive capacity of Tregs. Additionally, the result of pseudo-temporal analysis of the HNSCC TME indicated that after Nivolumab therapy, the expression of certain inhibitory immune checkpoints including TIGIT, ENTPD1, and CD276 and LY9, were decreased in Tregs, while LAG-3 showed an increased expression level. The study also found that Tregs had a dense communication network with cluster two, and that certain ligand-receptor pairs, including SPP1/CD44, HLA-E/KLRC2, HLA-E/KLRK1, ANXA1/FPR3, and CXCL9/FCGR2A, had notable changes after the therapy. These changes in gene expression and cell interactions may have implications for the role of Tregs in the TME and in response to Nivolumab therapy.


Subject(s)
Head and Neck Neoplasms , T-Lymphocytes, Regulatory , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Programmed Cell Death 1 Receptor/metabolism , Nivolumab/pharmacology , Nivolumab/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Gene Expression Profiling , Tumor Microenvironment , B7 Antigens , NK Cell Lectin-Like Receptor Subfamily C/metabolism
5.
Regen Ther ; 24: 630-641, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38034858

ABSTRACT

Recently, the demand for organ transplantation has promptly increased due to the enhanced incidence of body organ failure, the increasing efficiency of transplantation, and the improvement in post-transplant outcomes. However, due to a lack of suitable organs for transplantation to fulfill current demand, significant organ shortage problems have emerged. Developing efficient technologies in combination with tissue engineering (TE) has opened new ways of producing engineered tissue substitutes. The use of natural nanoparticles (NPs) such as nanocellulose (NC) and nano-lignin should be used as suitable candidates in TE due to their desirable properties. Many studies have used these components to form scaffolds and three-dimensional (3D) cultures of cells derived from different tissues for tissue repair. Interestingly, these natural NPs can afford scaffolds a degree of control over their characteristics, such as modifying their mechanical strength and distributing bioactive compounds in a controlled manner. These bionanomaterials are produced from various sources and are highly compatible with human-derived cells as they are derived from natural components. In this review, we discuss some new studies in this field. This review summarizes the scaffolds based on NC, counting nanocrystalline cellulose and nanofibrillated cellulose. Also, the efficient approaches that can extract cellulose with high purity and increased safety are discussed. We concentrate on the most recent research on the use of NC-based scaffolds for the restoration, enhancement, or replacement of injured organs and tissues, such as cartilage, skin, arteries, brain, and bone. Finally, we suggest the experiments and promises of NC-based TE scaffolds.

6.
J Health Popul Nutr ; 42(1): 128, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-37986111

ABSTRACT

BACKGROUND: Trauma has a significant impact on the overall health of individuals worldwide, being a leading cause of morbidity and mortality with long-lasting effects. The identification of suitable biomarkers is crucial to predict patient outcomes, providing information about the severity of a condition or the probability of a specific outcome. Hence, in this study, we addressed a new biomarker, copeptin, and discussed its prognostic roles in various trauma researches. MAIN BODY: Copeptin is a peptide derived from the precursor of the hormone vasopressin, which is released in response to stress. Copeptin can serve as a valuable biomarker for determining the severity, prognosis, and outcome of trauma patients. Elevated levels of copeptin are associated with increased mortality and poor clinical outcomes in patients with severe injuries or bleeding. Implementing copeptin measurements in clinical practice can enable healthcare providers to more accurately gauge the degree of trauma and predict patient mortality and morbidity outcomes facilitating prompt interventions and personalized treatment. CONCLUSION: The measurement of novel biomarker copeptin can serve as a prognostic molecule for further outcomes in trauma patients. Nevertheless, supplementary research is needed to fully comprehend its role in the development and progression of traumatic injuries.


Subject(s)
Glycopeptides , Humans , Prognosis , Biomarkers
7.
Front Immunol ; 14: 1280601, 2023.
Article in English | MEDLINE | ID: mdl-38022534

ABSTRACT

Malignancies contain a relatively small number of Mesenchymal stem/stromal cells (MSCs), constituting a crucial tumor microenvironment (TME) component. These cells comprise approximately 0.01-5% of the total TME cell population. MSC differentiation potential and their interaction with the tumor environment enable these cells to affect tumor cells' growth, immune evasion, metastasis, drug resistance, and angiogenesis. This type of MSC, known as cancer-associated mesenchymal stem/stromal cells (CA-MSCs (interacts with tumor/non-tumor cells in the TME and affects their function by producing cytokines, chemokines, and various growth factors to facilitate tumor cell migration, survival, proliferation, and tumor progression. Considering that the effect of different cells on each other in the TME is a multi-faceted relationship, it is essential to discover the role of these relationships for targeting in tumor therapy. Due to the immunomodulatory role and the tissue repair characteristic of MSCs, these cells can help tumor growth from different aspects. CA-MSCs indirectly suppress antitumor immune response through several mechanisms, including decreasing dendritic cells (DCs) antigen presentation potential, disrupting natural killer (NK) cell differentiation, inducing immunoinhibitory subsets like tumor-associated macrophages (TAMs) and Treg cells, and immune checkpoint expression to reduce effector T cell antitumor responses. Therefore, if these cells can be targeted for treatment so that their population decreases, we can hope for the treatment and improvement of the tumor conditions. Also, various studies show that CA-MSCs in the TME can affect other vital aspects of a tumor, including cell proliferation, drug resistance, angiogenesis, and tumor cell invasion and metastasis. In this review article, we will discuss in detail some of the mechanisms by which CA-MSCs suppress the innate and adaptive immune systems and other mechanisms related to tumor progression.


Subject(s)
Mesenchymal Stem Cells , Neoplasms , Humans , Cytokines/metabolism , Cell Differentiation , Immunity , Mesenchymal Stem Cells/metabolism , Tumor Microenvironment
8.
Life Sci ; 326: 121813, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37257578

ABSTRACT

Aim Sepsis is a medical emergency with no definitive treatment. Animal experiments have confirmed the therapeutic characteristics of exosomes in reducing inflammation and tissue damage. The study investigates the effect of MSC and hepatocyte-derived exosomes along with imipenem in controlling systemic and local (liver) inflammation in a mouse model of sepsis. MAIN METHODS: To induce sepsis in C57BL/6 mice, the Cecal Ligation and Puncture (CLP) model was used. The mice were given various treatments, including imipenem, MSC-derived exosomes, hepatocyte-derived exosomes, and a mixture of exosomes. Blood and liver samples were collected and analyzed for cell blood count, liver enzymes, NO levels, cytokine concentrations, and bacterial presence. The percentages of TCD3 + CD4+/CD8+ and Treg in the spleen and mesenteric lymph nodes were also assessed using flow cytometry. The pathological changes were assessed in the liver, lung, and heart tissues. In addition, the cytokine content of exosomes was measured by ELISA. KEY FINDINGS: Our results demonstrated that MSC-derived exosomes+imipenem could control systemic and local inflammation and increase the TCD4+ and Treg populations. Hepatocyte-derived exosomes+imipenem reduced inflammation in the liver and increased the TCD8+ and Treg populations. The mixture of exosomes+imipenem had the best function in reducing inflammation, maintaining all T lymphocyte populations, reducing liver damage, and ultimately increasing the survival rate. SIGNIFICANCE: The mixture of exosomes derived from MSCs and hepatocytes, along with imipenem, in the inflammatory phase of sepsis could be a promising therapeutic strategy in sepsis treatment.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Sepsis , Mice , Animals , Imipenem/pharmacology , Exosomes/pathology , Mice, Inbred C57BL , Hepatocytes/pathology , Cytokines , Liver/pathology , Inflammation/drug therapy , Sepsis/pathology , Mesenchymal Stem Cells/pathology
9.
Heliyon ; 9(5): e15694, 2023 May.
Article in English | MEDLINE | ID: mdl-37144199

ABSTRACT

Prostate cancer (PCa) is one of the two solid malignancies in which a higher T cell infiltration in the tumor microenvironment (TME) corresponds with a worse prognosis for the tumor. The inability of T cells to eliminate tumor cells despite an increase in their number reinforces the possibility of impaired antigen presentation. In this study, we investigated the TME at single-cell resolution to understand the molecular function and communication of dendritic cells (DCs) (as professional antigen-presenting cells). According to our data, tumor cells stimulate the migration of immature DCs to the tumor site by inducing inflammatory chemokines. Many signaling pathways such as TNF-α/NF-κB, IL2/STAT5, and E2F up-regulated after DCs enter the tumor location. In addition, some molecules such as GPR34 and SLCO2B1 decreased on the surface of DCs. The analysis of molecular and signaling alterations in DCs revealed some suppression mechanisms of tumors, such as removing mature DCs, reducing the DC's survival, inducing anergy or exhaustion in the effector T cells, and enhancing the differentiation of T cells to Th2 and Tregs. In addition, we investigated the cellular and molecular communication between DCs and macrophages in the tumor site and found three molecular pairs including CCR5/CCL5, CD52/SIGLEC10, and HLA-DPB1/TNFSF13B. These molecular pairs are involved in the migration of immature DCs to the TME and disrupt the antigen-presenting function of DCs. Furthermore, we presented new therapeutic targets by the construction of a gene co-expression network. These data increase our knowledge of the heterogeneity and the role of DCs in PCa TME.

10.
Curr Med Chem ; 2023 05 03.
Article in English | MEDLINE | ID: mdl-37138420

ABSTRACT

Psoriasis is defined as a chronic autoimmune disorder of the skin in which abnormal proliferation and differentiation of keratinocytes are blamed as the central culprit of disease etiopathogenesis. A complex interplay between environmental factors and genetic risk factors has been suggested to trigger the disease. However, epigenetic regulation appears to connect external stimuli and genetic abnormalities in the development of psoriasis. The discordance in the prevalence of psoriasis between monozygotic twins and environmental factors that contribute to its onset have caused a paradigm shift regarding the mechanisms underlying the pathogenesis of this disease. Epigenetic dysregulation may be involved in aberrancies of keratinocyte differentiation, T-cell activation, and other plausible cells, leading to the initiation and perpetuation of psoriasis. Epigenetics is characterized by heritable alterations in the transcription of genes without nucleotide change and is commonly considered at three levels, i.e., DNA methylation, histone modifications, and microRNAs. To date, scientific evidence has indicated abnormal DNA methylation, histone modifications, and non-coding RNA transcription in psoriatic patients. In order to reverse aberrant epigenetic changes in psoriasis patients, several compounds and drugs (epi-drugs) have been developed to affect the major enzymes involved in the methylation of DNA, or the acetylation of histones, which aim to correct the aberrant methylation and acetylation patterns. A number of clinical trials have suggested the therapeutic potential of such drugs in the treatment of psoriasis. In the present review, we attempt to clarify recent findings with respect to epigenetic irregularities in psoriasis and discuss future challenges.

11.
Iran J Allergy Asthma Immunol ; 22(6): 588-599, 2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38477955

ABSTRACT

Pathogen recognition receptors (PRRs), which play a crucial role in responding to pathogens, affect the function of mesenchymal stem cells (MSCs). One important group of PRRs is the toll-like receptors (TLRs). When PRRs are activated, they can alter the expression of specific surface markers, the ability of MSCs to differentiate, and the types of substances they secrete. These modifications in MSC function may have unexpected consequences for patients. In this study, we examined how Leishmania major (L. major) promastigotes affect the properties of MSCs. MSCs were isolated from adipose tissue and categorized into two groups: one group left untreated and the other group exposed to L. major. Giemsa staining was employed to accurately quantify the number of parasites that entered the cells. After 72 hours, real-time polymerase chain reaction was utilized to assess the expression of TLRs. Additionally, the flow cytometry technique was used to evaluate the expression of surface markers on the MSCs. Our results showed that MSCs can engulf parasites and increase the expression of TLR4 and TLR6. The pro-inflammatory cytokine increased, and the transforming growth factor-ß decreased significantly. The parasite exposure increased reactive oxygen species production. Additionally, the percentage of cluster differentiation (CD) 73 decreased, and the mean fluorescent index of CD29 and CD73 was down-regulated by L. major. Exposure to parasites diminishes the immunomodulatory capacity of MSCs. This discovery holds significance for the application of MSCs in addressing parasite infections and underscores the need for additional research to enhance their therapeutic effectiveness.


Subject(s)
Leishmania major , Mesenchymal Stem Cells , Humans , Cell Differentiation , Transforming Growth Factor beta , Adipose Tissue
12.
Life Sci ; 310: 120938, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36150466

ABSTRACT

BACKGROUND: Sepsis is a debilitating systemic inflammation that resulted from infection or injury. Despite many advances in treatment, the resulting mortality rate has remained high due to increasing antibiotic resistance and aging communities. The present study investigated the effects of stem cell-derived exosomes in a mouse model of LPS-induced systemic inflammation. MATERIALS AND METHODS: To induce sepsis, the LPS model was used. Mice were divided into three groups: normal, patient group (LPS + PBS), and treatment group (LPS + exosome). The treatment group received an intravenous exosome 1 h after induction of the model. Patient and treatment groups were sacrificed at 4, 6, 24, and 48 h after induction of the model, and their tissues were isolated. Blood samples were taken from animal hearts to perform biochemical and immunological tests. The study results were analyzed using Graph Pad Prism software version 9. RESULTS: Mesenchymal stem cell-derived exosomes decreased serum levels of ALT and AST liver enzymes, decreased neutrophil to lymphocyte ratio (NLR), and improved kidney, liver, and lung tissue damage at 4, 6, and 24 h after model induction. At 24 h, the exosomes were able to reduce serum urea levels. This study revealed decreased levels of inflammatory cytokines such as IL-6, IL-1ß, and TNF-α after exosome injection. CONCLUSION: Our findings suggest that treating mice with stem cell-derived exosomes can ameliorate the destructive effects of inflammation caused by sepsis by reducing inflammatory factors and tissue damage.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Sepsis , Mice , Animals , Lipopolysaccharides/toxicity , Mice, Inbred C57BL , Inflammation , Disease Models, Animal , Sepsis/chemically induced , Sepsis/therapy
13.
Cancers (Basel) ; 14(15)2022 Jul 29.
Article in English | MEDLINE | ID: mdl-35954362

ABSTRACT

Glioblastoma (GBM) therapy has seen little change over the past two decades. Surgical excision followed by radiation and chemotherapy is the current gold standard treatment. Immunotherapy techniques have recently transformed many cancer treatments, and GBM is now at the forefront of immunotherapy research. GBM immunotherapy prospects are reviewed here, with an emphasis on immune checkpoint inhibitors and oncolytic viruses. Various forms of nanomaterials to enhance immunotherapy effectiveness are also discussed. For GBM treatment and immunotherapy, we outline the specific properties of nanomaterials. In addition, we provide a short overview of several 3D (bio)printing techniques and their applications in stimulating the GBM microenvironment. Lastly, the susceptibility of GBM cancer cells to the various immunotherapy methods will be addressed.

14.
Trends Cardiovasc Med ; 32(8): 525-533, 2022 11.
Article in English | MEDLINE | ID: mdl-34492295

ABSTRACT

Mounting scientific evidence over decades has established that atherosclerosis is a chronic inflammatory disorder. Among the potentially critical sources of vascular inflammation during atherosclerosis are the components of pathogenic bacteria, especially lipopolysaccharide (LPS). Toll-like receptor (TLR)-4, expressed on different inflammatory cells involved with the recognition of bacterial LPS, has been recognized to have mutations that are prevalent in a number of ethnic groups. Such mutations have been associated with a decreased risk of atherosclerosis. In addition, epidemiological investigations have proposed that LPS confers a risk factor for the development of atherosclerosis. Gram-negative bacteria are the major source of LPS in an individual's serum, which may be generated during subclinical infections. The major cell receptors on inflammatory cells involved in the pathogenesis of atherosclerosis, like macrophages, monocytes, and dendritic cells (DCs), are CD14, MD-2, and LPS binding protein (LBP). These receptors have been blamed for the development of atherosclerosis through dysregulated activation following LPS recognition. Lipoproteins may also play a role in modulating the LPS-induced inflammatory events during atherosclerosis development. In this review article, we attempt to clarify the role of LPS in the initiation and progression of atherosclerotic lesion development.


Subject(s)
Atherosclerosis , Lipopolysaccharides , Humans , Lipopolysaccharides/pharmacology , Monocytes , Atherosclerosis/genetics , Macrophages
15.
Curr Genomics ; 23(3): 163-174, 2022 Jul 05.
Article in English | MEDLINE | ID: mdl-36777004

ABSTRACT

Psoriasis is an organ-specific autoimmune disease characterized by the aberrant proliferation and differentiation of keratinocytes, leading to skin lesions. Abnormal immune responses mediated by T cells and dendritic cells and increased production of inflammatory cytokines have been suggested as underlying mechanisms in the pathogenesis of psoriasis. Emerging evidence suggests that there is a heritable basis for psoriatic disorders. Moreover, numerous gene variations have been associated with the disease risk, particularly those in innate and adaptive immune responses and antigen presentation pathways. Herein, this article discusses the genetic implications of psoriatic diseases' etiopathogenesis to develop novel investigative and management options.

16.
Vaccines (Basel) ; 9(9)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34579244

ABSTRACT

Among the vaccines have been developed thus far against SARS-CoV-2, the mRNA-based ones have demonstrated more promising results regarding both safety and efficacy. Two remarkable features of the mRNA vaccines introduced by the Pfizer/BioNTech and Moderna companies are the use of (N1-methyl-pseudouridine-) modified mRNA and the microfluidics-based production of lipid nanoparticles (LNPs) as the carrier. In the present study, except Anti-Reverse Cap Analog (ARCA), no other nucleoside analogs were employed to synthesize Spike-encoding mRNA using the in vitro transcription (IVT) method. Furthermore, LNPs were prepared via the ethanol injection method commonly used for liposome formation as an alternative for microfluidics-based approaches. The produced mRNA-LNP vaccine was evaluated for nanoparticles characteristics, encapsulation and transfection efficiencies, in vitro cytotoxicity as well as stability and storability. The safety of vaccine was assessed in Balb/c mice injected with mRNA-LNPs containing 10 µg of spike-encoding mRNA. Eventually, the vaccine efficacy in inducing an immune response against SARS-CoV-2 was studied in Balb/c and C57BL/6 mice (received either 1 or 10 µg of mRNA) as well as in rhesus macaque monkeys (infused with mRNA-LNPs containing 100 µg of mRNA). The ELISA and virus neutralizing test (VNT) results showed a significant augmentation in the level of neutralizing antibodies against SARS-CoV-2. Moreover, the ELISA assay showed virus-specific IFN-γ secretion in immunized mice as a marker of TH1 cell-based immune response, whereas favorably no change in the production of IL-4 was detected.

17.
Int Immunopharmacol ; 100: 108076, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34450402

ABSTRACT

BACKGROUND: Evidence show that Matrix metalloproteinases (MMPs) have been associated with neurological complications in the viral infections. Here in the current investigation, we intended to reveal if MMPs are potentially involved in the development of neurological symptoms in the patients with Coronavirus disease 2019 (COVID-19). METHODS: The levels of MMPs, inflammatory cytokines, chemokines, and adhesion molecules were evaluated in the serum and cerebrospinal fluid (CSF) samples from 10 COVID-19 patients with neurological syndrome (NS) and 10 COVID-19 patients lacking NS. Monocytes from the CSF samples were treated with TNF-α and the secreted levels of MMPs were determined. RESULTS: The frequency of monocytes were increased in the CSF samples of COVID-19 patients with NS compared to patients without NS. Levels of inflammatory cytokines IL-1ß, IL-6, and TNF-α, chemokines CCL2, CCL3, CCL4, CCL7, CCL12, CXCL8, and CX3CL1, MMPs MMP-2, MMP-3, MMP-9, and MMP-12, and adhesion molecules ICAM-1, VCAM-1, and E-selectin were significantly increased in the CSF samples of COVID-19 patients with NS compared with patients without NS. Treatment of CSF-derived monocytes obtained from COVID-19 patients with NS caused increased production of MMP-2, MMP-3, MMP-9, and MMP-12. CONCLUSIONS: Higher levels of inflammatory cytokines might promote the expression of adhesion molecules on blood-CSF barrier (BCSFB), resulting in facilitation of monocyte recruitment. Increased levels of CSF chemokines might also help to the trafficking of monocytes to CSF. Inflammatory cytokines might enhance production of MMPs from monocytes, leading to disruption of BCSFB (and therefore further infiltration of inflammatory cells to CSF) in COVID-19 patients with NS.


Subject(s)
COVID-19/complications , Matrix Metalloproteinases/physiology , Nervous System Diseases/etiology , SARS-CoV-2 , Aged , Chemokines/analysis , Cytokines/analysis , Female , Humans , Intercellular Adhesion Molecule-1/analysis , Male , Middle Aged
18.
Microb Pathog ; 158: 105066, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34174356

ABSTRACT

Neutrophil extracellular traps (NETs) are networks of extracellular chromosomal DNA fibers, histones, and cytoplasmic granule proteins. The release of NET components from neutrophils is involved in the suppression of pathogen diffusion. Development of NETs around target microbes leads to disruption of the cell membrane, eventuating in kind of cell death that is called as NETosis. The very first step in the process of NETosis is activation of Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase upon signaling by innate immune receptors. Afterwards, produced Reactive oxygen species (ROS) trigger protein-arginine deiminase type 4, neutrophil elastase, and myeloperoxidase to generate decondensed chromatin and disrupted integrity of nuclear membrane. Subsequently, decondensed chromatin is mixed with several enzymes in the cytoplasm released from granules, leading to release of DNA and histones, and finally formation of NET. Several reports have indicated that NETosis might contribute to the immune responses through limiting the dissemination of microbial organisms. In this review, we discuss recent advances on the role of neutrophils, NETs, and their implications in the pathogenesis of microbial infections. Additionally, the prospective of the NET modulation as a therapeutic strategy to treat infectious diseases are clarified.


Subject(s)
Communicable Diseases , Extracellular Traps , Humans , NADPH Oxidases , Neutrophils , Prospective Studies , Reactive Oxygen Species
19.
Int Immunopharmacol ; 93: 107421, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33548581

ABSTRACT

BACKGROUND: Considering the role of inflammation in the outcome of sepsis and the widespread use of imipenem in the disease, this study was designed to assess the effect of imipenem on the dynamics of inflammatory responses in the sepsis mouse model. METHODS: Cecal Ligation and Puncture (CLP) model was used to induce sepsis in mice. C57BL/6 mice were divided into sham, CLP-induced sepsis mice, CLP-induced sepsis mice receiving 25 mg/kg, and 125 mg/kg imipenem. Blood and liver samples were obtained and bacterial load, endotoxin level, and liver enzymes were evaluated. The concentration and mRNA expression of cytokines were also determined. RESULTS: Sepsis mice treated with a high dose (125 mg/kg) of imipenem showed a significant reduction in bacterial load, while increased liver enzymes, endotoxin level, and inflammatory cytokine production in plasma and liver. In contrast, significant reduction in the liver enzymes, bacterial load, endotoxin levels, and inflammatory cytokine levels was observed in the mice treated with a low dose (25 mg/kg) of imipenem compared with other mice groups. Liver tissue pathology of mice indicated little tissue destruction in the sepsis mice treated with 25 mg/kg of imipenem compared to other groups. Mice receiving 25 mg/kg of imipenem had better survival rate. CONCLUSIONS: Our results demonstrated the dose-dependent effect of subcutaneous administration of imipenem on the inflammatory responses in sepsis mice. A dose of 25 mg/kg imipenem resulted in better pathology, lower inflammatory mediators, and increased survival rate in sepsis mice.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Imipenem/therapeutic use , Liver/metabolism , Sepsis/drug therapy , Animals , Bacterial Load , Cecum/surgery , Cytokines/metabolism , Disease Models, Animal , Endotoxins/metabolism , Female , Humans , Injections, Subcutaneous , Liver/pathology , Mice , Mice, Inbred C57BL
20.
Article in English | MEDLINE | ID: mdl-33404796

ABSTRACT

Rheumatoid arthritis (RA) is a chronic immune-mediated inflammatory disorder characterized by the destruction of the joint and bone resorption. The production of pro-inflammatory cytokines and chemokines, dysregulated functions of three important subtypes of T helper (TH) cells including TH1, TH17, and regulator T (Treg) cells are major causes of the initiation and development of RA. Moreover, B cells as a source of the production of several autoantibodies play key roles in the pathogenesis of RA. The last decades have seen increasingly rapid advances in the field of immunopharmacology using natural origin compounds for the management of various inflammatory diseases. Curcumin, a main active polyphenol compound isolated from turmeric, curcuma longa, possesses a wide range of pharmacologic properties for the treatment of several diseases. This review comprehensively will assess beneficial immunomodulatory effects of curcumin on the production of pro-inflammatory cytokines and also dysregulated functions of immune cells including TH1, TH17, Treg, and B cells in RA. We also seek the clinical efficacy of curcumin for the treatment of RA in several recent clinical trials. In conclusion, curcumin has been found to ameliorate RA complications through modulating inflammatory and autoreactive responses in immune cells and synovial fibroblast cells via inhibiting the expression or function of pro-inflammatory mediators, such as nuclear factor-κB (NF-κB), activated protein-1 (AP-1), and mitogen-activated protein kinases (MAPKs). Of note, curcumin treatment without any adverse effects can attenuate the clinical symptoms of RA patients and, therefore, has therapeutic potential for the treatment of the diseases.


Subject(s)
Arthritis, Rheumatoid , Curcumin , Synoviocytes , Arthritis, Rheumatoid/drug therapy , Curcumin/pharmacology , Curcumin/therapeutic use , Humans , Inflammation Mediators , NF-kappa B
SELECTION OF CITATIONS
SEARCH DETAIL
...