Faculty Opinions recommendation of In vivo imaging of inflammasome activation reveals a subcapsular macrophage burst response that mobilizes innate and adaptive immunity.

Author(s):  
Michael McDermott
2015 ◽  
Vol 22 (1) ◽  
pp. 64-71 ◽  
Author(s):  
Pervinder Sagoo ◽  
Zacarias Garcia ◽  
Beatrice Breart ◽  
Fabrice Lemaître ◽  
David Michonneau ◽  
...  

2020 ◽  
Vol 169 (4) ◽  
pp. 474-477
Author(s):  
S. V. Gein ◽  
O. A. Kochina ◽  
M. S. Kuyukina ◽  
D. P. Klimenko ◽  
I. B. Ivshina

2016 ◽  
Vol 11 (6) ◽  
pp. 1934578X1601100
Author(s):  
Anna K Gazha ◽  
Lyudmila A. Ivanushko ◽  
Eleonora V. Levina ◽  
Sergey N. Fedorov ◽  
Tatyana S. Zaporozets ◽  
...  

The action of seven polyhydroxylated sterol mono- and disulfates (1-7), isolated from ophiuroids, on innate and adaptive immunity was examined in in vitro and in vivo experiments. At least, three of them (1, 2 and 4) increased the functional activities of neutrophils, including levels of oxygen-dependent metabolism, adhesive and phagocytic properties, and induced the expression of pro-inflammatory cytokines TNF-α and IL-8. Compound 4 was the most active for enhancing the production of antibody forming cells in the mouse spleen.


2004 ◽  
Vol 172 (5) ◽  
pp. 3059-3069 ◽  
Author(s):  
Silvia Bellocchio ◽  
Claudia Montagnoli ◽  
Silvia Bozza ◽  
Roberta Gaziano ◽  
Giordano Rossi ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1949-1949
Author(s):  
Kavita Dhodapkar ◽  
Lin Zhang ◽  
Rakesh Verma ◽  
Kartik Sehgal ◽  
Mehmet H. Kocoglu ◽  
...  

Abstract Pomalidomide (POM) is a novel IMiD® immunomodulatory agent with clinical activity in several settings including in relapsed / resistant myeloma (RRMM). Several preclinical studies have documented the immunologic effects of IMiD® immunomodulatory drugs. IMiD® now form the backbone of several emerging combination therapies in hematologic malignancies. In prior studies, the clinical activity of POM in relapsed myeloma has been demonstrated using both continuous and intermittent dosing regimens with and without steroids. However the impact of the specific POM dosing regimen and the effect of concurrent steroids (as commonly utilized in most combination regimens) on POM-induced immune activation in vivo is unknown. In order to evaluate these issues more directly, we analyzed samples from patients enrolled in a randomized phase II clinical trial comparing two POM dosing schedules. Comparison of drug-induced immune activation between the two dosing schedules was one of the pre-specified endpoints in this study. Patients (n=39) with RRMM documented to be refractory to lenalidomide were randomized to therapy with POM 2 mg/day for 28/28 days (Arm A, n=19) or POM 4 mg/day for 21/28 days (Arm B, n=20) of a 28 day cycle. All patients (pts) received POM alone for cycle 1, followed by the addition of dexamethasone (DEX) at 40 mg weekly in subsequent cycles in both arms. In recent studies, we have shown that immune effects of lenalidomide are manifest early, within a week of initiation of therapy (Richter et al, Blood 121:423, 2013). Therefore each patient was analyzed 1 week after initiation of POM alone at 2 or 4 mg (cycle 1) or POM + DEX (cycle 2). POM therapy led to an increase in T cells at day 7 after initiation of therapy (mean percent increase compared to baseline 47%, p <0.01). This POM-induced T cell expansion consisted predominantly of CD8+ T cells (mean increase compared to baseline 40%, p<0.01). POM therapy also led to an increase in circulating natural killer (NK) cells (mean increase compared to baseline 92%, p<0.01) which peaked at 7 days post-initiation of therapy. POM-induced NK expansion was associated with an increase in the expression of NKG2D as well as CD16 and CD56, consistent with NK activation in vivo. Changes in T and NK cells following cycle 2 (POM+DEX) were compared with the data on cycle 1 (POM alone) in the same patient to evaluate the impact of DEX. Addition of DEX led to dampening of drug-induced T cell activation in vivo, but surprisingly did not dampen drug induced NK activation. Comparison of data from the two cohorts demonstrated that both 2 mg and 4 mg dose of POM led to comparable degree of immune activation in vivo, particularly for NK cells. In selected patients, post-treatment bone marrow samples were available after completion of at least 2 cycles of therapy. Comparison of the transcriptome of the CD138-depleted fraction from these marrows (representing bulk tumor microenvironment) identified nearly 300 genes upregulated in the bone marrow microenvironment post therapy compared to paired baseline samples. Interestingly, these genes were highly enriched in genes associated with T and NK activation. As POM led to activation of both innate and adaptive immunity, we also analyzed drug-induced changes in the myeloid compartment. Comparison of gene expression profiles (GEP) of purified CD14+ monocytes before and after POM indicated that POM therapy leads to a distinct GEP signature in myeloid cells in vivo. Finally, correlation of drug-induced changes in immune cells with clinical response demonstrated that an increase in post-therapy T cells correlated with clinical response, but post-treatment increase in NK cells did not. Together these data demonstrate that both 2 mg and 4 mg dose of POM can mediate broad activation of both innate and adaptive immunity in vivo, even in the setting of immune paresis associated with advanced, heavily pre-treated disease. Both continuous and intermittent dosing schedules of POM have comparable pharmacodynamic effects on immune activation. POM-induced immune activation is detected even with concurrent steroids and immune-related genes constitute a dominant component of drug induced changes in tumor microenvironment. POM-mediated immune activation may thus be exploited in the context of combination with immune therapies as well as monoclonal antibodies. Disclosures: No relevant conflicts of interest to declare.


2016 ◽  
Vol 113 (23) ◽  
pp. E3240-E3249 ◽  
Author(s):  
Derek W. Gilroy ◽  
Matthew L. Edin ◽  
Roel P. H. De Maeyer ◽  
Jonas Bystrom ◽  
Justine Newson ◽  
...  

Resolution of inflammation has emerged as an active process in immunobiology, with cells of the mononuclear phagocyte system being critical in mediating efferocytosis and wound debridement and bridging the gap between innate and adaptive immunity. Here we investigated the roles of cytochrome P450 (CYP)-derived epoxy-oxylipins in a well-characterized model of sterile resolving peritonitis in the mouse. Epoxy-oxylipins were produced in a biphasic manner during the peaks of acute (4 h) and resolution phases (24–48 h) of the response. The epoxygenase inhibitor SKF525A (epoxI) given at 24 h selectively inhibited arachidonic acid- and linoleic acid-derived CYP450-epoxy-oxlipins and resulted in a dramatic influx in monocytes. The epoxI-recruited monocytes were strongly GR1+, Ly6chi, CCR2hi, CCL2hi, and CX3CR1lo. In addition, expression of F4/80 and the recruitment of T cells, B cells, and dendritic cells were suppressed. sEH (Ephx2)−/− mice, which have elevated epoxy-oxylipins, demonstrated opposing effects to epoxI-treated mice: reduced Ly6chi monocytes and elevated F4/80hi macrophages and B, T, and dendritic cells. Ly6chi and Ly6clo monocytes, resident macrophages, and recruited dendritic cells all showed a dramatic change in their resolution signature following in vivo epoxI treatment. Markers of macrophage differentiation CD11b, MerTK, and CD103 were reduced, and monocyte-derived macrophages and resident macrophages ex vivo showed greatly impaired phagocytosis of zymosan and efferocytosis of apoptotic thymocytes following epoxI treatment. These findings demonstrate that epoxy-oxylipins have a critical role in monocyte lineage recruitment and activity to promote inflammatory resolution and represent a previously unidentified internal regulatory system governing the establishment of adaptive immunity.


2021 ◽  
Author(s):  
Nell G. Bond ◽  
Marissa Fahlberg ◽  
Shan Yu ◽  
Namita Rout ◽  
Dollnovan Tran ◽  
...  

AbstractInvariant natural killer T-lymphocytes (iNKT) are a unique subset of immunomodulatory innate T-cells with an invariant TCRα chain recognizing glycolipids presented on the MHC class-I-like CD1d molecule. Activated iNKT rapidly secrete pro-and anti-inflammatory cytokines, potentiate innate and adaptive immunity, and modulate inflammation. While iNKT activation by glycolipid agonists are being explored as an adjuvant, their use depends on CD1d-restricted antigen presentation. Here, we report the effects of iNKT activation by a novel humanized monoclonal antibody, NKTT320, that binds to the invariant region of the iNKT TCR. A single dose of NKTT320 led to rapid iNKT activation, increased polyfunctionality, and elevation of multiple pro-inflammatory and chemotactic plasma analytes within 24 hours in cynomolgus macaques. Flow cytometry and RNA-Seq confirmed downstream effects of NKTT320 on multiple immune cell subsets. Inflammatory response, JAK/STAT and PI3K/AKT pathway genes were enriched along with upregulation of the inflammation-modulating genes CMKLR1, ARG2 and NLRP12. Finally, NKTT320 induced iNKT trafficking to adipose tissue and did not cause iNKT anergy. Our data indicate that NKTT320 has a sustained effect on in vivo iNKT activation, potentiation of innate and adaptive immunity, and resolution of inflammation, properties that support its future application as an immunotherapeutic and vaccine adjuvant.


Nutrients ◽  
2021 ◽  
Vol 13 (11) ◽  
pp. 3738
Author(s):  
Pirjo Pärnänen ◽  
Hanna Lähteenmäki ◽  
Taina Tervahartiala ◽  
Ismo T. Räisänen ◽  
Timo Sorsa

Lingonberry (Vaccinium vitis ideae L.) is a low-bush wild plant found in the northern hemisphere. The berries are used in traditional medicine in Finland to treat oral yeast infections. General and oral effects of lingonberries on the microbiome and inflammation are reviewed. A brief introduction to oral microbiome symbiosis and dysbiosis, innate and adaptive immunity and inflammation are included, and special features in microbe/host interactions in the oral environment are considered. In vitro anticancer, antimicrobial, antioxidant, anti-inflammatory, and in vivo mouse and human studies are included, focusing on the symbiotic effect of lingonberries on oral and general health.


2018 ◽  
Vol 165 (3) ◽  
pp. 368-372 ◽  
Author(s):  
S. V. Gein ◽  
O. A. Kochina ◽  
M. S. Kuyukina ◽  
I. B. Ivshina

2015 ◽  
Vol 98 (6) ◽  
pp. 995-1001 ◽  
Author(s):  
Rob J. W. Arts ◽  
Bastiaan A. Blok ◽  
Peter Aaby ◽  
Leo A. B. Joosten ◽  
Dirk de Jong ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document