36 research outputs found
์ ๋ฐฉ์์ธํฌ์ ๋ ์์์ข์ ๋์์ธํฌ ๋ฐ ์์ง์์ธํฌ ๋ฉด์ญ์กฐ์ ํจ๊ณผ์ ๊ดํ ์ฐ๊ตฌ
ํ์๋
ผ๋ฌธ(๋ฐ์ฌ)--์์ธ๋ํ๊ต ๋ํ์ :์๊ณผ๋ํ ์๊ณผํ๊ณผ,2020. 2. ๋ฌธ์ฐ๊ฒฝ.์๋ก : ์์ธํฌ ์ ๋ ์์์ข์ ๋ฉด์ญ์ธํฌ์์ ์ ๋ณด๊ตํ์ ํตํด ์ข
์๋ฉด์ญ์ ์กฐ์ ํ์ฌ ์์ ์งํ๊ณผ ์ ์ด์ ๊ด์ฌํ๋ค๊ณ ์๋ ค์ ธ ์์ง๋ง ์ ๋ฐฉ์์์ ์์์ข์ ์๊ทน์ ๋ฐ์ ๋์์ธํฌ ๋๋ ์์ง์์ธํฌ์ ๋ฉด์ญ๊ธฐ๋ฅ ๋ณํ๊ฐ ์ ์งํ๊ณผ ์ ์ด์ ์ฃผ๋ ์ํฅ์ ๊ดํ ์์ฒด ์ฐ๊ตฌ๋ ๋ถ์กฑํ๋ค. ๋ฐ๋ผ์ ๋ณธ ์ฐ๊ตฌ์์๋ ์ธํฌ์ถ์ ์์์ผ๋ก ์ ๋ฐฉ์์ธํฌ์ ๋ ์์์ข๊ณผ ๋์์ธํฌ ๋๋ ์์ง์์ธํฌ์์ ์ํธ์์ฉ์ ๊ด์ฐฐํ๊ณ ์์์ข์ ์ํ ๋์์ธํฌ์ ์์ง์์ธํฌ์ ์ข
์๋ฉด์ญ ๊ธฐ๋ฅ ๋ณํ๋ฅผ ๋ถ์ํ๊ณ ์ ํ๋ค. ๋ํ ์์ฒด๊ดํ์์๊ธฐ๋ฒ๊ณผ ์ผ์ฒดํ ๊ด์ํฅ ์ด์ํ ์์๊ธฐ๋ฒ์ ์ด์ฉํ์ฌ ์์์ข์ ์ํ ์ ๋ฐฉ์ ์งํ๊ณผ ์ ์ด ๊ธฐ์ ์ ๊ท๋ช
ํ๊ณ ์ ํ๋ค.
์คํ๋ฐฉ๋ฒ: ๋ณธ ์ฐ๊ตฌ์์๋ ์ฌ๋๊ณผ ๋ง์ฐ์ค ์ผ์ค์์ฑ์ ๋ฐฉ์์ธํฌ(MDA-MB-231๊ณผ 4T1), ๋ง์ฐ์ค ๋์์ธํฌ(Raw264.7) ๊ทธ๋ฆฌ๊ณ ๋ง์ฐ์ค ์์ง์์ธํฌ(DC2.4)๋ฅผ ์ฌ์ฉํ์๋ค. ๋ ํฐ๋ฐ์ด๋ฌ์ค ์์คํ
์ ์ด์ฉํด ์์์ข ํน์ด ๋ง๋จ๋ฐฑ์ง(CD63)๊ณผ ์ ์ํ๊ด๋จ๋ฐฑ์ง(RFP)๋ฅผ ์ฌ์กฐํฉํ CD63/RFP๋ฅผ ๋ฐํํ๋ MDA-MB-231๏ผCD63/RFP, 4T1๏ผCD63/RFP, ๋ฃจ์ํผ๋ผ์ (firefly luciferase)์ ๋
น์ํ๊ด๋จ๋ฐฑ์ง(GFP)์ ๋์์ ๋ฐํํ๋ MDA-MB-231๏ผLuc/GFP, GFP๋ฅผ ๋ฐํํ๋ RAW264.7/GFP์ DC2.4/GFP๋ฅผ ์๋ฆฝํ์๋ค. MDA-MB-231๏ผCD63/RFP ์ 4T1๏ผCD63/RFP์์ ๋ถ๋น๋๋ ์ ์ํ๊ด๋จ๋ฐฑ์ง์ ๋ฐํํ๋ ์์์ข์ ์ถ์ถํ์ฌ, ๋๋
ธ์
์๋ถ์ ๊ธฐ๊ธฐ(NanoSight)๋ฅผ ์ฌ์ฉํ์ฌ ํฌ๊ธฐ๋ฅผ ์ธก์ ํ๊ณ , ์จ์คํด ๋ธ๋ฟ์ผ๋ก ์์์ข ํน์ด ๋จ๋ฐฑ์ง์ ๋ฐํ์ ํ์ธํ์๋ค. ์ถ์ถํ ์์์ข์ ์ธํฌ๊ฐ ์ด๋์ ๊ณต์ด์ ํ๋ฏธ๊ฒฝ์ผ๋ก ์ถ์ ๊ด์ฐฐํ์์ผ๋ฉฐ, ์ธํฌ์ฆ์ ํ๊ฐ(MTT)์ ํธ๋ ์ค์ฐ ์ด๋ ๋ถ์๋ฒ์ผ๋ก ์์์ข์ ์ํ ์ธํฌ๋ค์ ์ฆ์, ์ด๋ ๋ฐ ์นจ์ค ๋ฅ๋ ฅ์ ๋ถ์ํ์๊ณ , ์ค์๊ฐ ์ญ์ ์ฌ์คํฉํจ์์ฐ์๋ฐ์๋ฒ, ์จ์คํด ๋ธ๋ฟ๊ณผ ์ ์ธํฌ ๋ถ์์ผ๋ก ๋์์ธํฌ์ ์์ง์์ธํฌ์ ๋ฉด์ญ๊ธฐ๋ฅ์ ๋ถ์ํ์๋ค. ๋ฉด์ญ๊ฒฐํ๋ง์ฐ์ค(BALB/c nude)์ ์ ์ ์ง๋ฐฉ์กฐ์ง์ MDA-MB-231-Luc/GFP๋ฅผ ์ด์ํ์ฌ ์ ๋ฐฉ์๋ชจ๋ธ์ ๋ง๋ค์๋ค. ์์ฒด๋ฐ๊ด์์์ผ๋ก ์์์ข์ ์ํ ์ข
์์ ์ฑ์ฅ๊ณผ ์ ์ด ๊ณผ์ ์ ์ถ์ ๊ด์ฐฐํ์๊ณ ์ผ์ฒดํ ๊ด์ํฅ ์ด์ํ ์์์ผ๋ก EGFRํญ์ฒด๊ฐ ํ์ฌ๋ ๊ณจ๋๋๋
ธ๋ก๋(anti-EGFR-GNs)๋ฅผ ์ผ์ฐจ ์ข
์์ ์ฃผ์
ํ์ฌ ์ ๋ฐฉ์ ๋ฆผํ์ ์ ์ด๋ฅผ ๋ถ์ํ์๋ค. ๋ํ ๋ง์ฐ์ค ํผํ์ ์์์ข์ผ๋ก ํ์ฑ์ํจ ์์ง์์ธํฌ๋ฅผ ๊ณจ๋๋๋
ธ๋ก๋(GNs)๋ก ํ์งํ์ฌ ์ฃผ์ฌํ๊ณ ํผํ๋ถํฐ ์ก์๋ฆผํ์ ๋ก์ ์ด๋์ ์ผ์ฒดํ ๊ด์ํฅ ์ด์ํ ์์๊ธฐ๋ฒ์ผ๋ก ์ถ์ ๊ด์ฐฐ ํ์๋ค. ์์ ๋ถ์ ํ ์ ์ถํ ์ข
์ ๋ฐ ๋ฆผํ์ ์กฐ์ง์์ H&E์ผ์๊ณผ ๋ฉด์ญ์ผ์์ ์ํํ์๋ค.
๊ฒฐ๊ณผ: ์ ๋ฐฉ์์ธํฌ, ๋์์ธํฌ ๊ทธ๋ฆฌ๊ณ ์์ง์์ธํฌ๋ด์์ ์ ๋ฐฉ์์ธํฌ ์ ๋ ์์์ข์ ํฌ์์ ํ์ธํ์๊ณ ๊ทธ ์ค ๋์์ธํฌ์ ์์ง์์ธํฌ๋ ์์์ข์ ์ํด ํ์ฑํ ๋๋ ๊ฒ์ด ๊ด์ฐฐ๋์๋ค. ์์์ข์ ์ํด ๊ฐ ์ธํฌ๋ค์ ์ฑ์ฅ, ์ด๋ ๋ฐ ์นจ์ค ๋ฅ๋ ฅ์ด ์ฆ๊ฐ๋์๊ณ , ๋์์ธํฌ์์๋ M1, M2 ๋ถ๊ทนํ ๋ฐํ ๋ง์ปค์ธ NOS2์ CD206, Arginase-1์ ๋ฐํ์ด ์ ๋๋์๊ณ , ์์ง์์ธํฌ์์๋ ๋ณด์กฐ์๊ทน ๋ถ์์ธ CD40, CD80, CD86์ ๋ฐํ๊ณผ ์ด๋์ฑ ๊ด๋ จ ์ผ๋ชจ์นด์ธ ์์ฉ์ฒด์ธ CCR7์ ๋ฐํ๋ฟ๋ง ์๋๋ผ ๋ถํ์ ์ฑ์์ ์๊ทนํ๋ TNF-ฮฑ์ ๋ฐํ์ด ์ฆ๊ฐ๋์๋ค. ์์ฒด๋ฐ๊ด์์๊ณผ ์ผ์ฒดํ ๊ด์ํฅ ์ด์ํ ์์์ผ๋ก ์ ๋ฐฉ์ ๋ชจ๋ธ์์ ์์์ข์ ์ํด ์ข
์์ ์ฑ์ฅ๊ณผ ์ ์ด๊ฐ ์ด์ง๋๋ ๊ฒ์ ํ์ธํ์๊ณ ๋ง์ฐ์ค์ ํผํ์ ์ด์ํ ์์์ข์ผ๋ก ํ์ฑ ๋ ์์ง์์ธํฌ๋ ๋ฆผํ์ ๋ก์ ์ด๋ ๋ฅ๋ ฅ์ด ์ฆ๊ฐ๋์๋ค.
๊ฒฐ๋ก : ํ๊ด๋จ๋ฐฑ์ง์ ๋ฐํํ๋ ์ ๋ฐฉ์์ธํฌ ์ ๋์ ์์์ข์ด ์ ๋ฐฉ์์ธํฌ, ๋์์ธํฌ, ์์ง์์ธํฌ๋ก ์ด๋ํ๋ ๊ณผ์ ์ ์ค์๊ฐ์ผ๋ก ์ถ์ ๊ด์ฐฐํ ๊ฒฐ๊ณผ, ์์์ข์ ๋์์ธํฌ์ ๋ถ๊ทนํ์ ์์ง์์ธํฌ์ ๋ถํ๋ฅผ ์ ๋ํ์ฌ ์ ๋ฐฉ์์ ์ฑ์ฅ๊ณผ ์ ์ด๋ฅผ ์กฐ์ ํ ๊ฒ์ด๋ผ๋ ๊ฐ๋ฅ์ฑ์ ์ ์ํ์๋ค. ๋ํ ์์ฒด๊ดํ์์๊ธฐ๋ฒ๊ณผ ์ผ์ฒดํ ๊ด์ํฅ ์ด์ํ ์์๊ธฐ๋ฒ์ ์์์ข์ ์ํ ๊ตญ์ ๋ฆผํ์ ์ ์ด ์ฌ๋ถ์ ์์ง์์ธํฌ์ ์ด๋๊ณผ ๋ถํฌ๋ฅผ ํจ์จ์ ์ผ๋ก ์ถ์ ๊ด์ฐฐํ๊ณ , ๋ถ์ํ ์ ์๋ ์์ฒด ์์๋ฒ์ผ๋ก ๋ค์ํ ์์ข
์ ํ์ฉ์ ํ์ฉ๋ ์ ์์ ๊ฒ์ด๋ค.Introduction: Cancer cell-derived exosomes known as the mediators of intercellular communications, are involved in tumor progress and metastasis by modulating tumor immunity. Nevertheless, the role of macrophages or dendritic cells stimulated by breast cancer cell-derived exosome in breast tumor progression and metastasis is not fully elucidated. In this study, we investigated the interaction of breast cancer cell-derived exosomes with macrophages or dendritic cells, and analyzed the tumor immunity of macrophage and dendritic cells which stimulated by exosomes. We also analyzed the molecular mechanisms of exosome mediated breast cancer growth and metastasis using bioluminescent imaging and ultrasound-guided photoacoustic imaging.
Methods: The human and mouse triple negative breast cancer cell lines MDA-MB-231 and 4T1, mouse macrophage cell line Raw264.7 and mouse dendritic cell line DC2.4 were used. MDA-MB-231๏ผCD63/RFP, 4T1๏ผCD63/RFP cells were established using lentivirus expressing RFP-tagged CD63, which is a specific marker of exosomes. MDA-MB-231๏ผLuc/GFP cells expressing both green fluorescence protein (GFP) and the firefly luciferase (Luc), RAW264.7/GFP cells and DC2.4/GFP cells expressing GFP were established using a lentiviral system. RFP-tagged exosomes secreted from MDA-MB-231๏ผCD63/RFP or 4T1๏ผCD63/RFP cells were isolated. The size of isolated exosomes was measured by NanoSight and specific exosomal marker proteins were assessed by Western blot. The intercellular transfer of RFP tagged exosomes between cells was monitored by confocal microscopy. The effects of exosomes on cell proliferation, migration and invasive abilities were evaluated by MTT and Trans-well migration assays. The expression of genes associated with the anti-or pro-tumor immunity in the exosome-stimulated macrophage and dendritic cells were evaluated using RT-PCR, Western blot and flow cytometry. The xenograft tumor models were produced by injection with MDA-MB-231-Luc/GFP cells into the mammary gland of female Balb/c nude mice. Tumor progression and metastasis affected by exosomes were noninvasively monitored by bioluminescence imaging and ultrasound-guided photoacoustic imaging after intratumoral injection of anti-EGFR-GNs. Histologic examination of tissue was performed by H&E staining and immunostaining.
Result: RFP tagged exosomes taken up by breast cancer cells, macrophages and dendritic cells can be tracked by live-cell microscopy, macrophage and dendritic cells when took up exosomes were exhibited slightly elongated morphology. Exosomes significantly increased cell growth, migration and invasion abilities. Exosomes activated macrophages to M1 and M2 phenotypes, which exhibited high expression of M1 marker (NOS2) and M2 markers (CD206, Arginase-1). Exosomes led to dendritic cell activation by inducing upregulation of co-stimulatory molecules CD40, CD80, and CD86, the chemokine receptor CCR7, and TNF-ฮฑ. Primary tumor growth and metastasis promoted by exosomes and exosome-stimulated dendritic cell migration into lymph node were observed by bioluminescent imaging and ultrasound-guided photoacoustic imaging.
Conclusions: We monitored the interaction of RFP-tagged exosomes with macrophages and dendritic cells in real-time. Our data demonstrated breast cancer cell-derived exosomes mediate breast cancer progress by modulating the immune function of macrophage and dendritic cells. The bioluminescent imaging and ultrasound-guided photoacoustic imaging combined with GNs allowed for sensitive and longitudinal monitoring of tumor growth, metastasis and dendritic cell migration in vivo.Chapter 1 1
Introduction 2
Materials and methods 5
Results 15
Discussion 25
Chapter 2 31
Introduction 32
Materials and methods 35
Results 48
Discussion 54
References 71
Abstract in korean 84Docto
CarboxyAmido-Triazole Orotate inhibits the growth of Imatinib-resistant chronic myeloid leukaemia cells and modulates exosomes-stimulated angiogenesis
Chronic myelogenous leukemia is a myeloproliferative disorder characterized by the t(9:22) (q34:q11) reciprocal chromosomal translocation, resulting in the expression of the chimeric BcrโAbl oncoprotein with constitutive tyrosine kinase activity. Deregulated BcrโAbl induces the hyperactivation of various signalling pathways that promote cell growth, suppress apoptosis and alter cell adhesion. Bcr-Abl has also been involved in VEGF-mediated angiogenesis in CML and evidence indicates that the formation of new vessels plays an important role in the development and progression of CML. Imatinib mesylate (IM) is a selective well tolerated inhibitor of the BcrโAbl tyrosine kinase that has significantly improved the prognosis of patients with chronic phase CML. Despite this remarkable progress, a major problem associated with the administration of imatinib is acquired resistance. Bcr-Abl gene amplification, increased expression of BcrโAbl protein, point mutations in the BcrโAbl tyrosine kinase domain have been reported as mechanisms of resistance to imatinib. Therefore, there is an urgent need for new anticancer agents and combinations that could improve responses and survival rates for CML. Recent studies from our laboratory have shown that addition of carboxyamidotriazole (CAI), an inhibitor of calcium-mediated signal transduction, to imatinib resistant human CML cells induces a marked decrease in cell viability and augmented apoptosis, events associated with downregulation of BcrโAbl protein and inhibition of tyrosine phosphorylation of BcrโAbl, STAT5, CrkL. Carboxyamidotriazole Orotate (CTO), is a derivate of CAI that has been developed at Tactical Therapeutics. CTO has a higher bioavailability and efficacy with respect to the parental compound. Exosomes are small vesicles of 40-100 nm diameter that are initially formed within the endosomal compartment and are secreted when a multivesicular body (MVB) fuses with the plasma membrane. These vesicles are released by many cell types including cancer cells and are considered messengers in intercellular communication. The exact function of exosomes in malignant cells has yet to be elucidated, but investigation has suggested roles in cell-to-cell communication, tumor-stroma interaction, and antigen presentation, thus potentially affecting cancer progression at different steps. Recent studies from our laboratory suggest that exosomes released from IM-sensitive CML cells directly affect endothelial cells modulating the process of neovascularization. Our data show that CTO is able to inhibit both in vitro and in vivo the growth of imatinib-resistant CML cells and to affect tumor microenvironment by modulating exosome-stimulated angiogenesis. CTO may be effective in targeting both cancer cell growth and the tumor microenvironment, thus suggesting a potential therapeutic utility in the treatment of leukemia patients
Role of exosomes released by chronic myelogenous leukemia cells in angiogenesis
The present study is designed to assess if exosomes released from Chronic Myelogenous Leukemia (CML) cells may modulate angiogenesis. We have isolated and characterized the exosomes generated from LAMA84 CML cells and demonstrated that addition of exosomes to
human vascular endothelial cells (HUVEC) induces an increase of both ICAM-1 and VCAM-1 cell adhesion molecules and interleukin-8 expression. The stimulation of cell-cell adhesion
molecules was paralleled by a dose-dependent increase of adhesion of CML cells to a HUVEC
monolayer. We further showed that the treatment with exosomes from CML cells caused an increase in endothelial cell motility accompanied by a loss of VE-cadherin and ฮฒ-catenin from the
endothelial cell surface. Functional characterization of exosomes isolated from CML patients
confirmed the data obtained with exosomes derived from CML cell line. CML exosomes caused reorganization into tubes of HUVEC cells cultured on Matrigel. When added to Matrigel plugs in vivo, exosomes induced ingrowth of murine endothelial cells and vascularization of the Matrigel
plugs. Our results suggest for the first time that exosomes released from CML cells directly affect endothelial cells modulating the process of neovascularization
Serum-derived extracellular vesicles from breast cancer patients contribute to differential regulation of T-cell-mediated immune-escape mechanisms in breast cancer subtypes
BackgroundIntracellular communication within the tumour is complex and extracellular vesicles (EVs) have been identified as major contributing factors for the cell-to-cell communication in the local and distant tumour environments. Here, we examine the differential effects of breast cancer (BC) subtype-specific patient serum and cell-line derived EVs in the regulation of T cell mediated immune responses. MethodsUltracentrifugation was used to isolate EVs from sera of 63 BC patients, 15 healthy volunteers and 4 human breast cancer cell lines. Longitudinal blood draws for EV isolation for patients on neoadjuvant chemotherapy was also performed. Characterization of EVs was performed by Nanoparticle Tracking Analysis (NTA), transmission electron microscopy (TEM) and immunoblotting. CD63 staining was performed on a tissue microarray of 218 BC patients. In-house bioinformatics algorithms were utilized for the computation of EV associated expression scores within The Cancer Genome Atlas (TCGA) and correlated with tumour infiltrating lymphocyte (TIL) scores. In vitro stimulation of PBMCs with EVs from serum and cell-line derived EVs was performed and changes in the immune phenotypes characterized by flow cytometry. Cytokine profiles were assessed using a 105-plex immunoassay or IL10 ELISA. ResultsPatients with triple negative breast cancers (TNBCs) exhibited the lowest number of EVs in the sera; whilst the highest was detected in ER+HER2+ cancers; reflected also in the higher level of CD63+ vesicles found within the ER+HER2+ local tumour microenvironment. Transcriptomic analysis of the TCGA data identified that samples assigned with lower EV scores had significantly higher abundance of CD4+ memory activated T cells, T follicular cells and CD8 T cells, plasma, and memory B cells; whilst samples with high EV scores were more enriched for anti-inflammatory M2 macrophages and mast cells. A negative correlation between EV expression scores and stromal TIL counts was also observed. In vitro experiments confirmed that circulating EVs within breast cancer subtypes have functionally differing immunomodulatory capabilities, with EVs from patients with the most aggressive breast cancer subtype (TNBCs) demonstrating the most immune-suppressive phenotype (decreased CD3+HLA-DR+ but increased CD3+PD-L1 T cells, increased CD4+CD127-CD25hi T regulatory cells with associated increase in IL10 cytokine production). In depth assessment of the cytokine modulation triggered by the serum/cell line derived exosomes confirmed differential inflammatory cytokine profiles across differing breast cancer subtypes. Studies using the MDA-231 TNBC breast cancer cell-line derived EVs provided further support that TNBC EVs induced the most immunosuppressive response within PBMCs.DiscussionOur study supports further investigations into how tumour derived EVs are a mechanism that cancers can exploit to promote immune suppression; and breast cancer subtypes produce EVs with differing immunomodulatory capabilities. Understanding the intracellular/extracellular pathways implicated in alteration from active to suppressed immune may provide a promising way forward for restoring immune competence in specific breast cancer patient populations
Trypanosoma brucei effect on leukocyte differentiation and activation
Tripanossomรญase Africana ou doenรงa do sono รฉ uma doenรงa zoonรณtica causada por Trypanosoma brucei, um protozoรกrio parasita transmitido pela mosca tsetse ou Glossina. A introduรงรฃo do parasita no hospedeiro vertebrado provoca uma sucessรฃo de eventos que envolvem a imunidade inata e adaptativa. Os macrรณfagos (Mฮฆ) apresentam um papel fundamental na defesa inata por serem cรฉlulas apresentadoras profissionais de antigรฉnio (APC) e cรฉlulas fagocรญticas, importantes na eliminaรงรฃo dos tripanossomas. A imunidade adaptativa รฉ assegurada pelos linfรณcitos, nomeadamente pelos linfรณcitos T. Contudo, o papel desempenhado por estas cรฉlulas imunitรกrias ainda nรฃo se encontra completamente clarificado. Assim, este estudo teve como objetivo analisar a atividade dos Mฮฆ na infeรงรฃo por T. brucei, avaliando os nรญveis de ureia e รณxido nรญtrico (NO) e a expressรฃo membranar das molรฉculas de classe I (MHCI) e classe II (MHCII) do complexo principal de histocompatibilidade. A diferenciaรงรฃo das populaรงรตes linfocitรกrias, T helper (Th), T citotรณxicas (Tc) e T reguladoras foi tambรฉm avaliada. Mฮฆ de murganho foram expostos a tripomastigotas de T. brucei e estimulados com antigรฉnio total e exossomas de T. brucei. A caracterizaรงรฃo da ativaรงรฃo macrofรกgica atravรฉs de ensaios colorimรฉtricos demonstrou que ambos os fenรณtipos M1 e M2 foram expressos e foi evidenciada correlaรงรฃo positiva entre a produรงรฃo de ureia e de NO. Adicionalmente, os resultados da citometria de fluxo indicaram que o parasita prejudica a diferenciaรงรฃo das subpopulaรงรตes de MฮฆMHCI+ e MฮฆMHCII+ mas induz o aumento de molรฉculas MHCI. Contrariamente, os exossomas demonstraram estimular as funรงรตes APC atravรฉs das molรฉculas MHCI e MHCII. Os rรกcios MHCI/MHCII indicaram que o contacto com o parasita favorece a apresentaรงรฃo antigรฉnica ร s cรฉlulas TCD4+, enquanto que os exossomas direcionam a apresentaรงรฃo antigรฉnica para as subpopulaรงรตes de linfรณcitos TCD4+ e TCD8+. A caraterizaรงรฃo das populaรงรตes linfocitรกrias atravรฉs da citometria de fluxo demonstrou que T. brucei causa a diminuiรงรฃo das populaรงรตes de linfรณcitos Th e Tc. Contrariamente, a estimulaรงรฃo com o antigรฉnio total ou com os exossomas induziram a expansรฃo de ambas as subpopulaรงรตes linfocitรกrias. T. brucei parece promover a expansรฃo das subpopulaรงรตes linfocitรกrias CD8-CD25+FoxP3- e CD8+CD25+FoxP3-. A expansรฃo da subpopulaรงรฃo celular CD25-FoxP3+ foi observada na fraรงรฃo celular CD8- apรณs estimulaรงรฃo do antigรฉnio e na fraรงรฃo celular CD8+ estimulada por exossomas. Alรฉm disso, os exossomas tambรฉm promoveram a expansรฃo da subpopulaรงรฃo de linfรณcitos T CD8+CD25+FoxP3+. Curiosamente, apesar da contraรงรฃo celular T. brucei induziu o aumento das molรฉculas FoxP3. Em conjunto, os resultados obtidos indicam que o parasita e os exossomas parecem exercer efeitos opostos nas cรฉlulas analisadas. Os parasitas parecem minimizar a apresentaรงรฃo antigรฉnica e evitam induzir a expansรฃo das subpopulaรงรตes de linfรณcitos T, facilitando a sua permanรชncia no hospedeiro. Por outro lado, os exossomas segregados por T. brucei parecem estimular a apresentaรงรฃo antigรฉnica e mediar a expansรฃo dos linfรณcitos Th, possivelmente deslocando o foco da atividade do sistema imunitรกrio dos parasitas para os exossomas. Estes resultados permitiram clarificar alguns dos princรญpios subjacentes ร resposta imunitรกria inata e adaptativa na fase inicial da infeรงรฃo. A compreensรฃo destes mecanismos pode vir a contribuir para o desenvolvimento de novas estratรฉgias de controlo e eliminaรงรฃo da Tripanossomรญase Africana.African trypanosomiasis or sleeping sickness is a zoonotic disease caused by Trypanosoma brucei, a protozoan parasite transmitted by tsetse fly or Glossina. Parasite introduction into mammal hosts, triggers a succession of events, involving both innate and adaptive immunity. Macrophages (Mฮฆ) have a key role in innate defense, since they are antigen-presenting cells (APC) and have a phagocytosis function essential for trypanosomes clearance. Adaptive immune defense is carried out by lymphocytes, in particular by T lymphocytes. However, the exact role of these immune cells remains not completely understood. Thus, this study aimed to assess the role of Mฮฆ in T. brucei infection by measuring the urea and nitric oxide (NO) levels, and by evaluating membrane expression of class I (MHCI) and class II (MHCII) molecules of major histocompatibility complex. The differentiation of T helper (Th), T cytotoxic (Tc) and T regulatory cell subsets was assessed. Mouse Mฮฆ were exposed to T. brucei trypomastigotes and stimulated by T. brucei extract and T. brucei exosomes. Characterization of Mฮฆ activation with colorimetric assays have indicated that both M1 and M2 phenotypes were expressed, evidencing a positive correlation between urea and NO levels produced. Additionally, results of flow cytometry indicated that T. brucei impairs the expansion of both MHCI+ and MHCII+ Mฮฆ subsets, but enhanced MHCI molecules. On the contrary, T. brucei exosomes stimulated APC functions through MHCI and MHCII molecules. MHCI/MHCII rates indicated that T. brucei shift the antigen presentation to CD4+ T cells, while exosomes directed the antigen presentation to both CD4+ and CD8+ T cells. Characterization of lymphocyte subsets by flow cytometry demonstrated that T. brucei impairs both Th and Tc lymphocytes. On the contrary, cell stimulation by extract and exosomes promote the expansion of both T cell subpopulations. T. brucei seem to promote the expansion of CD8-CD25+FoxP3- and CD8+CD25+FoxP3- T cell subsets. The expansion of CD25-FoxP3+ T cell subset was observed in CD8- cell fraction antigen stimulated and in the CD8+ cell fraction exosome stimulated. Moreover, exosomes also induced the expansion of CD8+ CD25+FoxP3+ T cell subset. Interestingly, despite cell decrease T. brucei seemed to increase FoxP3 molecules. Taken together, these findings indicate that parasite and parasite exosomes seem to have opposite effects on the evaluated cells. Parasites seem to minimize antigen presentation and avoid inducing the expansion of T cell subsets, facilitating its permanence in the host. On the other hand, T. brucei secreted exosomes seems to induce APC functions and mediate the expansion of Th lymphocytes, probably focusing the immune activity on the exosomes and not on the parasites. These findings allowed to understand some underlying principles of the innate and adaptive immune response in the early-stage of infection. Comprehension of these mechanisms can endorse the development of new strategies for control and elimination of African Trypanosomiasis
Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through NF-Kฮฒ dependent glycolytic metabolic reprogramming.
The formation of a pre-metastatic niche is a fundamental requirement for primary tumor metastasis. One of the defining characteristics of a pre-metastatic niche is infiltration of immunosuppressive macrophages. However, how these macrophages acquire their immunosuppressive phenotype remains largely unexplored. Here, we demonstrate that tumor-derived exosomes (TDE) polarize macrophages towards an immunosuppressive phenotype characterized by increased PD-L1 expression through NF-kB-dependent metabolic reprogramming in mice and humans. While NF-ฮบB has previously been shown to act as a direct transcription factor for PD-L1, we report a novel mechanism where TDE-induced NF-ฮบB activation drives PD-L1 expression by augmenting the glycolytic capacity of macrophages through two separate pathways. First, NF-ฮบB increases glucose uptake into macrophages via a HIF-1ฮฑ/GLUT-1-dependent mechanism. Secondly, elevated NOS2-dependent nitric oxide inhibits mitochondrial oxidative phosphorylation resulting in an increased conversion of pyruvate to lactate. Lactate then feeds back on NF-ฮบB further increasing PD-L1 expression. Analysis of metastasis negative draining lymph nodes of non-small cell lung cancer patients revealed that macrophage PD-L1 expression positively correlates with expression levels of GLUT-1 and exosomal release genes YKT6 and TSG101 from primary tumors. Collectively, our study provides a novel mechanism by which macrophages within a pre-metastatic niche acquire their immunosuppressive phenotype and identifies an important link between exosomes, metabolism, and metastasis
Exosome-mediated crosstalk between chronic myelogenous leukemia cells and human bone marrow stromal cells triggers an interleukin 8-dependent survival of leukemia cells
Chronic myelogenous leukemia (CML) is a myeloproliferative disorder characterized by the BcrโAbl oncoprotein
with constitutive tyrosine kinase activity. Exosomes are nanovesicles released by cancer cells that
are involved in cell-to-cell communication thus potentially affecting cancer progression. It is well known
that bone marrow stromal microenvironment contributes to disease progression through the establishment
of a bi-directional crosstalk with cancer cells. Our hypothesis is that exosomes could have a functional
role in this crosstalk. Interleukin-8 (IL 8) is a proinflammatory chemokine that activates multiple
signalling pathways downstream of two receptors (CXCR1 and CXCR2). We demonstrated that exosomes
released from CML cells stimulate bone marrow stromal cells to produce IL 8 that, in turn, is able to modulate
both in vitro and in vivo the leukemia cell malignant phenotype
Therapeutic targeting of tumor associated macrophages.
Previous studies on the mechanistic induction of anti-tumor responses by IL-12 cytokine therapy have focused on the adaptive immune response, specifically the activation NK cells and T cells as the primary targets of IL-12 treatment. In contrast, little attention has been given to the potential role of macrophages in the initiation of anti-tumor responses by IL-12 therapy despite reports that macrophages play a major role in promoting tumor growth and metastasis and in suppressing anti-tumor immune responses. Based on the functional adaptivity hypothesis, which is the concept that macrophages functionally adapt, rather than differentiate into specific mature subsets, in response to environmental stimuli, we hypothesized that tumor infiltrating (TIMs) as well as tumor-associated macrophages (TAMs) could be converted from tumor supportive activities to pro-immunogenic activities by IL-12 therapy. We examined the functional activities displayed by TIMs and TAMs after treatment with IL-12. Our data demonstrate (1) that tumor cells and tumor exosomes activate TIMs and TAMs by cell-contact dependent mechanisms involving ligation of CD40 and/or NKG2D, (2) that IL-12 treatment both in vivo and in vitro induces a rapid reduction of tumor supportive activities and a concomitant increase in pro-inflammatory activities in TIMs as well as TAMs, (3) that IL-12 induces a rapid release of cytoplasmic IL-I5 from the in situ activated tumor associated macrophages and (4) the release of IL-15 in essential to the recruitment of lymphocytes to the tumor and the metastatic lung, and to the destruction of the tumor and clearance of metastasis. It is concluded that macrophages in the tumor environment are activated and functionally modulated by the tumor. TIMs and TAMs respond to IL-12 treatment by rapidly converting from suppressive, tumor supportive activities to inflammatory, pro-immunogenic activities. Tumor associated macrophages thus are a critical target of IL-12 therapy and may orchestrate the subsequent NK and T cell cytotoxic response against the tumor
Tumor-associated stromal cells as key contributors to the tumor microenvironment.
The tumor microenvironment is a heterogeneous population of cells consisting of the tumor bulk plus supporting cells. It is becoming increasingly evident that these supporting cells are recruited by cancer cells from nearby endogenous host stroma and promote events such as tumor angiogenesis, proliferation, invasion, and metastasis, as well as mediate mechanisms of therapeutic resistance. In addition, recruited stromal cells range in type and include vascular endothelial cells, pericytes, adipocytes, fibroblasts, and bone-marrow mesenchymal stromal cells. During normal wound healing and inflammatory processes, local stromal cells change their phenotype to become that of reactive stroma. Under certain conditions, however, tumor cells can co-opt these reactive stromal cells and further transition them into tumor-associated stromal cells (TASCs). These TASCs express higher levels of proteins, including alpha-smooth muscle actin, fibroblast activating protein, and matrix metalloproteinases, compared with their normal, non-reactive counterparts. TASCs are also known to secrete many pro-tumorigenic factors, including IL-6, IL-8, stromal-derived factor-1 alpha, vascular endothelial growth factor, tenascin-C, and matrix metalloproteinases, among others, which recruit additional tumor and pro-tumorigenic cells to the developing microenvironment. Here, we review the current literature pertaining to the origins of recruited host stroma, contributions toward tumor progression, tumor-associated stromal cells, and mechanisms of crosstalk between endogenous host stroma and tumor cells