immune barriers
Recently Published Documents


TOTAL DOCUMENTS

29
(FIVE YEARS 13)

H-INDEX

9
(FIVE YEARS 4)

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Dobrin D. Draganov ◽  
Antonio F. Santidrian ◽  
Ivelina Minev ◽  
Duong Nguyen ◽  
Mehmet Okyay Kilinc ◽  
...  

An amendment to this paper has been published and can be accessed via the original article.


2021 ◽  
Vol In Press (In Press) ◽  
Author(s):  
Fahimeh Ghotbizadeh ◽  
Farzaneh Nazari ◽  
Sedigheh Hantoushzadeh ◽  
Zahra Panahi ◽  
Zahra Naeiji

Context: Recently, a new coronavirus (SARS-CoV-2) as the cause of COVID-19 has made a global health crisis and a great challenge. Pregnant women and fetuses are among the high-risk groups for COVID-19. In this review, we summarize studies regarding SARS-CoV-2 virus-placenta interactions at the maternal-fetal interface by demonstrating the pathogenicity of the virus and defense methods of the placenta. Evidence Acquisition: In the present study, a search was done in domestic and international databases including Google Scholar, Web of Science, PubMed, and Scopus using specific keywords (“Coronavirus” OR “COVID-19” OR “SARS-CoV-2”) AND (“Fetus” OR “Placenta”) AND (“Pregnancy”), limited until August 2020. Finally, we reviewed 250 articles. Results: Generally, the pathogenicity and the life cycle of SARS-CoV-2 and virus entry and replication methods allow the virus to pass through the placenta, although there are hormonal and immune barriers in the placenta against SARS-CoV-2, such as placental type interferons. Conclusions: The SARS-CoV-2 can pass through the placenta, but there are defense methods against it.


2021 ◽  
Vol 131 (1) ◽  
Author(s):  
Manish Muhuri ◽  
Yukiko Maeda ◽  
Hong Ma ◽  
Sanjay Ram ◽  
Katherine A. Fitzgerald ◽  
...  

Background: MicroRNAs (miRNAs) are a subset of small regulatory RNAs that are expressed by almost all metazoans and protozoans. They express genes either by direct cleavage or by suppressing the translation of target mRNAs by partial complementary base pairing. The active and functional unit of miRNA is a complex of Argonaute proteins known as microRNA-induced silencing complex (miRISC). They are known to regulate various growth and physiological processes. Irregular expression of miRNA in human cells is associated with a variety of disorders such as cancer, cardiovascular dysfunction, liver damage, immune dysfunction, metabolic syndromes, and pathogenic infections. A growing number of studies have shown that miRNAs are in fact a major component of host interactions and pathogens and play an important role in host immune responses to microorganisms. Emerging miRNAs are recognized as important tools for the genetic study, therapeutic development, and diagnosis of human pathogenic infections caused by various pathogenic organisms such as viruses, bacteria, parasites, and fungi. Many pathogens use the host miRNA system for their own benefits, including pathogenesis, survival within the host cell, and crossing some host immune barriers. Other pathogens express their miRNA within the host and contribute to their replication, survival, or delay. This article aims to review the role and importance of miRNA in relation to some important parasitic diseases.


2020 ◽  
Author(s):  
Lisa M Godsel ◽  
Quinn R Roth-Carter ◽  
Jennifer L Koetsier ◽  
Lam C Tsoi ◽  
Joshua A Broussard ◽  
...  

AbstractDesmoglein 1 (Dsg1) is a cadherin restricted to stratified tissues of terrestrial vertebrates, which serve as essential physical and immune barriers. Dsg1’s importance in epidermal integrity is underscored by genetic, autoimmune and bacterial toxin-mediated disorders interfering with Dsg1 function. Dsg1 loss-of-function mutations in humans result not only in skin lesions, but also multiple allergies, and isolated patient keratinocytes exhibit increased pro-allergic cytokine expression. However, the mechanism by which genetic deficiency of Dsg1 causes chronic inflammation is unknown. To determine the systemic response to Dsg1 loss, we deleted the three tandem Dsg1 genes in mice using CRISPR/Cas9. Whole transcriptome analysis of E18.5 Dsg1−/− skin showed changes consistent with the observed aberrant differentiation and barrier impairment. Comparing epidermal transcriptomes from E18.5 Dsg1-deficient mice and humans with Dsg1 mutations revealed a shared psoriatic-like IL-17-skewed inflammatory signature and less so a pro-allergic IL-4/13 signature. Although the impaired intercellular adhesion observed in Dsg1−/− mice resembles that resulting from autoimmune anti-Dsg1 pemphigus foliaceus antibodies, transcriptomic analysis of pemphigus skin lesions lacks a prominent IL-17 signature. Thus, beyond impairing the physical barrier, chronic loss of Dsg1 function through gene mutation results in a psoriatic-like inflammatory signature before birth, possibly predisposing to skin inflammation.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A758-A758
Author(s):  
Duong Nguyen ◽  
Alberto Gomez ◽  
Forrest Neuharth ◽  
Ashley Alamillo ◽  
Thomas Herrmann ◽  
...  

BackgroundOncolytic virotherapy has been recognized as a promising new therapy for cancer for decades but only few viruses have been approved worldwide. The therapeutic potential of oncolytic viruses can be severely restricted by innate and adaptive immune barriers making oncolytic virus clinically inefficient. To overcome this obstacle, we utilized adipose-derived stem cells (AD-MSC) loaded with tumor selective CAL1 oncolytic vaccinia virus to generate a new therapeutic agent called SNV1 (SuperNova-1).MethodsCAL1 vaccinia virus was tested for its ability to replicate and selectively kill various human cancer cell lines in vitro and in vivo. Additionally, CAL1 was loaded into adipose-derived mesenchymal stem cells to generate SuperNova1 (SNV1). Both CAL1 and SNV1 were tested for their ability to kill cancer cells in the presence of active complement and neutralizing antibodies in cell culture as well as in mice. Immune cell infiltration of the treated and untreated tumors was analyzed by flow cytometry.ResultsCAL1 showed preferential amplification and killed various tested human (PC3, FaDu, MDA-MB-231, RPMI) and mouse cancer cells (CT26, EMT6, TRAMP-C2, RM1). In animals, CAL1 caused tumor regression in PC3 and CT26 mouse models without signs of toxicity. SNV1 significantly enhanced protection of CAL1 virus from clearance by the immune system as compared to naked CAL1 virus, leading to higher therapeutic efficacy in animals. Five days after SNV1 administration, tumor infiltrating lymphocytes (TILs) from both treated and untreated tumors showed increased CD4 and CD8 T-cell infiltrations. Importantly, we documented a decreased frequency of Tregs, and improved effector to Treg ratios, which was associated with inhibition of tumor growth at the treated tumor site and also at distant untreated sites.ConclusionsCAL1 is potentially used as an oncolytic agent. In addition, SNV1 cell-based platform protects and potentiates oncolytic vaccinia virus by circumventing humoral innate and adaptive immune barriers, resulting in enhanced oncolytic virotherapy. Particularly, SNV1 provided instantly active viral particles for immediate infection and simultaneous release of therapeutic proteins in the injected tumors.


2020 ◽  
Author(s):  
Hak Myong Choe ◽  
Zhao-Bo Luo ◽  
Mei-Fu Xuan ◽  
Biao-Hu Quan ◽  
Jin-Dan Kang ◽  
...  

AbstractThe recent GGTA1 and CMAH DKO pigs have made it possible to resolve the immune barriers which are duo to xenoantigens on RBC such as αGal and Neu5Gc. Nevertheless, it still requires the detection of glycosylation alternation on the pig RBCs because even the minor changes would be unexpected xenoantigens.DKO RBC immune reactivity with human serum was assessed by hemagglutination assay. Glycosylation alteration of RBC membranes was characterized by NanoLC-Q-TOF-MS system and lectin blotting assay.Twelve GGTA1/CMAH DKO piglets were successfully produced. The immunoreactivity with human serum was remarkably reduced in DKO (less than 1:2 dilution), whereas wild type(WT) pigs showed agglutination (the least 1:256 dilution). The MS results showed that DKO increased neutral N-glycans as well as decreased total sialylated N-glycans, especially suggesting significant decrease of di-sialylated N-glycans (P < 0.05). Moreover, lectin blotting assay revealed that DKO pigs reduced the binding signals with AAL, AOL, LCA and SNA and increased the binding signal with MAL.DKO pigs decreased the expression of total fucosylation and sialylated N-glycans on the erythrocyte membrane. Our findings will support further investigation into DKO pig RBC glycosylation and contribute to uncover the roles of glycan changes for xenotransfusion.Summary statementTo detect glycosylation changes in red blood cells(RBC) of GGTA1/CMAH double knockout(DKO) pigs, comparative analysis of the glycan profiling was done.


2020 ◽  
Vol 38 (5_suppl) ◽  
pp. 21-21
Author(s):  
Duong Nguyen ◽  
Alberto Gomez ◽  
Ashley Alamillo ◽  
Forrest Neuharth ◽  
Ivelina Minev ◽  
...  

21 Background: Oncolytic virotherapy has been pursued by multiple companies and institutions with few candidates reaching the clinic and demonstrating limited efficacy. The therapeutic potential of oncolytic viruses can be severely restricted by innate and adaptive immune barriers. To overcome this obstacle, we load and protect tumor selective CAL1 oncolytic vaccinia virus into adipose-derived stem cells (AD-MSC) to generate a new therapeutic agent called SNV1(SuperNova1). Methods: SNV1s were generated by incubating AD-MSC with CAL1 virus. SNV1 was analyzed for its ability to kill cancer cell lines and protect virus in the presence of active neutralizing antibodies and complement. In animals, SNV1 was intratumorally injected in various xenograft and syngeneic models. Viral biodistribution was also evaluated by PCR. Immune infiltration were analyzed using flow cytometry. Results: Compared to the naked virus, SNV1 showed improved protection against the humoral barriers and efficient eradication of various human cancer cell lines in vitro. Intratumoral SNV1 treatment showed statistically significant and potentiated tumor growth inhibition compared to control or CAL1 naked virus treatment in all tested models (prostate, breast, melanoma, colon, and prostate cancers). Importantly, local administration of SNV1 induced systemic therapeutic effects. Five days after SNV1 administration, tumor infiltrating lymphocytes (TILs) from both treated and untreated tumors showed increased CD4 and CD8 T-cell populations. As well as decreased frequency of Tregs, and improved effector to Treg ratios, which was associated with inhibition of tumor growth at the treated tumor site and also at distant untreated sites. Ongoing and persistent virus infection could be detected in the treated tumor as late as 15 days after administration. Conclusions: This study demonstrates the ability of our cell-based platform to protect and potentiate oncolytic vaccinia virus by circumventing the innate and adaptive immune barriers, resulting in enhanced oncolytic virotherapy. These findings provide fundamental rationale for the development of cell-based platforms to maximize the therapeutic potential of various oncolytic viruses.


Author(s):  
Jun Wei ◽  
Peiwen Chen ◽  
Pravesh Gupta ◽  
Martina Ott ◽  
Daniel Zamler ◽  
...  

Abstract CNS immune defenses are marshalled and dominated by brain resident macrophages and microglia, which are the innate immune sentinels and frontline host immune barriers against various pathogenic insults. These myeloid lineage cells are the predominant immune population in gliomas, and can constitute up to 30–50% of the total cellular composition. Parenchymal microglial cells and recruited monocyte-derived macrophages from the periphery exhibit disease specific phenotypic characteristics with spatial and temporal distinctions and are heterogeneous subpopulations based on their molecular signatures. A preponderance of myeloid over lymphoid lineage cells during CNS inflammation, including gliomas, is a contrasting feature of brain immunity relative to peripheral immunity. Herein we discuss glioma associated macrophage and microglia immune biology in the context of their identity, molecular drivers of recruitment, nomenclature and functional paradoxes, therapeutic reprogramming and polarization strategies, relevant challenges, and our perspectives on therapeutic modulation.


2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Patricia I. S. Pinto ◽  
Cláudia C. Guerreiro ◽  
Rita A. Costa ◽  
Juan F. Martinez-Blanch ◽  
Carlos Carballo ◽  
...  

Abstract Pseudo-albinism is a pigmentation disorder observed in flatfish aquaculture with a complex, multi-factor aetiology. We tested the hypothesis that pigmentation abnormalities are an overt signal of more generalised modifications in tissue structure and function, using as a model the Senegalese sole and two important innate immune barriers, the skin and intestine, and their microbiomes. Stereological analyses in pseudo-albino sole revealed a significantly increased mucous cell number in skin (P < 0.001) and a significantly thicker muscle layer and lamina propria in gut (P < 0.001). RNA-seq transcriptome analysis of the skin and gut identified 573 differentially expressed transcripts (DETs, FDR < 0.05) between pseudo-albino and pigmented soles (one pool/tissue from 4 individuals/phenotype). DETs were mainly linked to pigment production, skin structure and regeneration and smooth muscle contraction. The microbiome (16 S rRNA analysis) was highly diverse in pigmented and pseudo-albino skin but in gut had low complexity and diverged between the two pigmentation phenotypes. Quantitative PCR revealed significantly lower loads of Mycoplasma (P < 0.05) and Vibrio bacteria (P < 0.01) in pseudo-albino compared to the control. The study revealed that pseudo-albinism in addition to pigmentation changes was associated with generalised changes in the skin and gut structure and a modification in the gut microbiome.


Sign in / Sign up

Export Citation Format

Share Document