[go: up one dir, main page]

 
 

Molecular and Cellular Mechanisms of Lymphomas

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Pathology".

Deadline for manuscript submissions: 31 December 2024 | Viewed by 4768

Special Issue Editor


E-Mail Website
Guest Editor
1. Biobank of Research, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
2. Department of Medical and Surgical Sciences (DIMEC), Bologna University, Bologna, Italy
Interests: leukemia; lymphoma; targeted therapy; precision medicine; digital pathology; hematopathology; molecular pathogenesis of haematopoietic neoplasms; the role of viruses in lymphomagenesis; the metabolic alterations associated with leukemias and lymphomas; molecular diagnostics
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Recent improvements in our knowledge of the molecular pathogenesis of malignant lymphoma offer new rational basis for innovative targeted therapies. However, there is still a consistent gap between the identification of pathways as potential targets and the development of effective therapeutic strategies in this field.

This Special Issue aims to provide new concepts concerning the molecular pathogenesis, genetics, molecular diagnosis and cell metabolism of human lymphomas. New experimental findings (molecular diagnosis, molecular and cellular mechanisms ) in addition to review articles will be considered.

The objective is to eventually favor the translation of genomic and cellular findings into concrete benefits for lymphoma patients.

Prof. Dr. Pier Paolo Piccaluga
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • lymphoma
  • pathogenes and lymphomas
  • molecular pathogenesis
  • molecular diagnosis
  • cellular or gene-targeted therapy
  • cell metabolism

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (4 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

16 pages, 5075 KiB  
Article
The Oncoprotein Fra-2 Drives the Activation of Human Endogenous Retrovirus Env Expression in Adult T-Cell Leukemia/Lymphoma (ATLL) Patients
by Julie Tram, Laetitia Marty, Célima Mourouvin, Magali Abrantes, Ilham Jaafari, Raymond Césaire, Philippe Hélias, Benoit Barbeau, Jean-Michel Mesnard, Véronique Baccini, Laurent Chaloin and Jean-Marie Jr. Peloponese
Cells 2024, 13(18), 1517; https://doi.org/10.3390/cells13181517 - 10 Sep 2024
Viewed by 539
Abstract
Human endogenous retroviruses (HERVs) are retroviral sequences integrated into 8% of the human genome resulting from ancient exogenous retroviral infections. Unlike endogenous retroviruses of other mammalian species, HERVs are mostly replication and retro-transposition defective, and their transcription is strictly regulated by epigenetic mechanisms [...] Read more.
Human endogenous retroviruses (HERVs) are retroviral sequences integrated into 8% of the human genome resulting from ancient exogenous retroviral infections. Unlike endogenous retroviruses of other mammalian species, HERVs are mostly replication and retro-transposition defective, and their transcription is strictly regulated by epigenetic mechanisms in normal cells. A significant addition to the growing body of research reveals that HERVs’ aberrant activation is often associated with offsetting diseases like autoimmunity, neurodegenerative diseases, cancers, and chemoresistance. Adult T-cell leukemia/lymphoma (ATLL) is a very aggressive and chemoresistant leukemia caused by the human T-cell leukemia virus type 1 (HTLV-1). The prognosis of ATLL remains poor despite several new agents being approved in the last few years. In the present study, we compare the expression of HERV genes in CD8+-depleted PBMCs from HTLV-1 asymptomatic carriers and patients with acute ATLL. Herein, we show that HERVs are highly upregulated in acute ATLL. Our results further demonstrate that the oncoprotein Fra-2 binds the LTR region and activates the transcription of several HERV families, including HERV-H and HERV-K families. This raises the exciting possibility that upregulated HERV expression could be a key factor in ATLL development and the observed chemoresistance, potentially leading to new therapeutic strategies and significantly impacting the field of oncology and virology. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Lymphomas)
Show Figures

Figure 1

Figure 1
<p>Prevalence of Abs against human HERV Env antigens in patients with acute ATL and the respective control groups. (<b>A</b>) HERV envelope (ENV) antigenemia in sera from non-infected patients NI (<span class="html-italic">n</span> = 7; black circle), HTLV-1 asymptomatic carriers (<span class="html-italic">n</span> = 7; green square), and acute ATLL patients ATLL (<span class="html-italic">n</span> = 7; red diamond). The dotted lines represent positivity thresholds calculated by ROC analysis. (<b>B</b>) The area under the curve (AUC) and its statistical significance are reported (ns <span class="html-italic">p</span> ≤ 0.05 and **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 2
<p>Detection of human HERV mRNA by qRT-PCR in ATL-derived cell lines. (<b>A</b>–<b>C</b>) Expression of HERV mRNA was assessed in one HTLV-1-negative cell line (Jurkat) and three HTLV-1-derived cell lines (HUT102, C81–66, and ATL-2). (<b>D</b>,<b>E</b>) HERV gag mRNA expression was compared in Jurkat cells and HTLV-1-derived cell lines. (<b>F</b>) HERV-R-Pol mRNA was compared in Jurkat cells and HTLV-1-derived cell lines (** <span class="html-italic">p</span> ≤ 0.001). (<b>G</b>–<b>I</b>) Relative Fra-2, Tax, and HBZ expression in Jurkat cells and HTLV-1-derived cell lines (statistical significance was determined using a one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001, **** <span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 3
<p>Detection of human HERV mRNA in CD8<sup>+</sup>-depleted PBMCs from HTLV-1-infected patients. (<b>A</b>–<b>D</b>) HERV mRNA was expressed in acute ATLL patients (red square) compared to HTLV-1 asymptomatic carriers (ACs) (black dot). (<b>E</b>,<b>F</b>) HERV gag mRNA expression was compared in AC (black dot) and ATLL patients (red square) (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.001; **** <span class="html-italic">p</span> ≤ 0.00001). (<b>G</b>) HERV-R-Pol was measured in AC and ATLL patients compared to ACs (** <span class="html-italic">p</span> ≤ 0.001). (<b>H</b>,<b>I</b>) The relative expression of Tax and HBZ in AC patients and ATL patients.</p>
Full article ">Figure 4
<p>HBZ does not activate HERV LTR. HEK293T was co-transfected with a plasmid carrying the luciferase reporter gene under the control of an NF-kB-dependent promoter (<b>A</b>), the collagenase promoter (<b>B</b>), or different HERV LTRs (<b>C</b>–<b>F</b>) and Tax-Flag, p65-Flag, or HBZ-Myc expression vectors, in addition to pRcActin-LacZ for the normalization of transfection efficiency. Cells were harvested 48 h. post-transfection and assayed for luciferase activity. The results show a fold increase compared to the mock control (set at a value of 1) and represent the mean values of three independently transfected cells. (<b>A</b>,<b>B</b>) Western blot analyses assessed the expression of Tax, p65, and HBZs. Actin is shown as a loading control (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, * <span class="html-italic">p</span> ≤ 0.01, ** <span class="html-italic">p</span> ≤ 0.001; *** <span class="html-italic">p</span> ≤ 0.00001).</p>
Full article ">Figure 5
<p>Fra-2 but not cFos activate the HERV LTR. HEK293T cells were co-transfected with a plasmid carrying the luciferase reporter gene under the control of the collagenase promoter in triplicate as shown (<b>A</b>) or different HERV 5′LTRs (<b>B</b>–<b>E</b>), different combinations of binding partners of AP-1-related transcription factors, and pRcActin-LacZ. The cells were harvested 48 h post-transfection and assayed for luciferase activity. The results show a fold increase in the mock control and represent the mean values of three independently transfected cell samples. (<b>F</b>) Western blot analyses were carried out to assess the expression of AP-1 transcription factors. Actin is shown as a loading control (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05,* <span class="html-italic">p</span> ≤ 0.01, ** <span class="html-italic">p</span> ≤ 0.001; *** <span class="html-italic">p</span> ≤ 0.0001 and **** <span class="html-italic">p</span> ≤ 0.00001).</p>
Full article ">Figure 6
<p>HBZ does not alter the activation of type I and type II HERV-H LTRs by Fra-2. (<b>A</b>) HEK293T cells were co-transfected with the different AP-1 in the absence or presence of HBZ and a plasmid carrying the luciferase reporter gene under the control of the collagenase promoter. (<b>B</b>) Western blot analyses were carried out to assess the expression of AP-1 transcription factors and HBZ. Actin is shown as a loading control. HEK293T cells were co-transfected with a plasmid carrying the luciferase reporter gene under the control of the collagenase promoter (<b>A</b>) or different HERV 5′LTRs (<b>C</b>–<b>E</b>), different AP-1 expression vectors, and pRcActin-LacZ in the presence or absence of HBZ. The cells were harvested 48 h post-transfection and assayed for luciferase activity. The results show a fold increase in the mock control and represent the mean values of three independently transfected cell samples (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, * <span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure 7
<p>HERV Env mRNA detected in HEK293T cells stably expressing Fra-2. (<b>A</b>) Western blot analyses were carried out on the lysate of two pools of HEK293-Fra2 to assess the expression of Fra-2. Beta actin is shown as a loading control. (<b>B</b>–<b>E</b>) HEK293T stably expressing Fra2 was harvested at different passages (from p3 to p8), and the expression of HERV-H Env, HERV-R Env, HERV-K Env, and HERV-E gag mRNAs was assessed by qRT-PCR (one-way ANOVA test with Dunn‘s multiple comparisons post-test ns <span class="html-italic">p</span> ≤ 0.05, * <span class="html-italic">p</span> ≤ 0.01; ** <span class="html-italic">p</span> ≤ 0.001).</p>
Full article ">Figure 8
<p>Kinetic analysis of Fra-2 and HERV Env mRNA in CD8<sup>+</sup>-depleted PBMCs from asymptomatic carriers and ATL patients. CD8<sup>+</sup>-depleted PBMCs from five ATL patients were cultivated ex vivo for five days, and Fra-2 (closed black square) (<b>A</b>), HERV-H Env (closed green triangle) (<b>B</b>), and HERV-K Env mRNA (closed red triangle) (<b>C</b>) were quantified at different time points using qRT-PCR (one-way ANOVA test with Dunn‘s multiple comparisons post-test; *** <span class="html-italic">p</span> ≤ 0.0001 and **** <span class="html-italic">p</span> ≤ 0.00001 (<b>D</b>,<b>E</b>) Relevance of Fra-2 and HERV Env mRNA expression in ATL patients, as analyzed with the Pearson correlation Test.</p>
Full article ">Figure 9
<p>Fra-2 binds to HERV-H-LTR in ATL-2 cells. ChIP assays were performed on chromatin prepared from the indicated ATL-2 cell lines using antibodies against Fra-2. Data are presented as fold enrichment relative to the IgG control. Data are an average of three independent experiments. Error bars represent the SEM (two-way ANOVA <span class="html-italic">t</span>-test, ns <span class="html-italic">p</span> ≤ 0.05 ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
14 pages, 3123 KiB  
Article
SATB1 and p16 Expression and Prognostic Value in Croatian Hodgkin Lymphoma Patients: A Unicentric Study
by Lučana Vicelić Čutura, Milan Vujčić, Davor Galušić, Viktor Blaslov, Marija Petrić, Antonija Miljak, Mirela Lozić, Benjamin Benzon, Katarina Vukojević, Toni Bubić, Nenad Kunac, Danijela Zjačić Puljiz, Ivana Kristina Delić Jukić, Marinela Križanac and Bernarda Lozić
Cells 2024, 13(16), 1323; https://doi.org/10.3390/cells13161323 - 8 Aug 2024
Viewed by 734
Abstract
Hodgkin lymphoma (HL) is a rare lymphoid neoplasm in which Hodgkin/Reed–Stenberg (HRS) cells are admixed with a population of non-neoplastic inflammatory cells and fibrosis. Dysregulated expressions of cell cycle regulators and transcription factors have been proven as one of the hallmarks of HL. [...] Read more.
Hodgkin lymphoma (HL) is a rare lymphoid neoplasm in which Hodgkin/Reed–Stenberg (HRS) cells are admixed with a population of non-neoplastic inflammatory cells and fibrosis. Dysregulated expressions of cell cycle regulators and transcription factors have been proven as one of the hallmarks of HL. In that context, SATB1 and p16 have been reported as potential regulators of HL progression and survival. However, to date, no studies have assessed the expression levels of SATB1 and p16 in HL in Croatian patients or their prognostic values. Therefore, we investigated the expression pattern of SATB1 and p16 in paraffin-embedded lymph node biopsies using standard immunohistochemistry. We found that 21% of the patients stained positive for SATB1, while 15% of the patients displayed positive staining for p16. Furthermore, we aimed to understand the prognostic value of each protein through the analysis of the overall survival (OS) and progression-free survival (PFS). SATB1 showed a significantly positive correlation with better OS and PFS, while p16 expression had no impact. Interestingly, when patients were stratified by a combination of the two studied markers, we found that patients in the SATB1+/p16- group tended to have the best prognosis in HL, according to statistical significance. In conclusion, SATB1 and p16 might be potentially useful as diagnostic and prognostic markers for HL. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Lymphomas)
Show Figures

Figure 1

Figure 1
<p>Immunohistochemical staining of (<b>A</b>) SATB1 and (<b>B</b>) p16 in lymph node biopsies of patients with Hodgkin lymphoma according to positivity score (1–3). The positive controls for SATB1 were surrounding lymphocytes; for p16, they were cervix adenocarcinoma cells. Pictures were taken at 40× magnification. Arrows indicate HRS cells. Abbreviations: +CTRL, positive control.</p>
Full article ">Figure 2
<p>Kaplan–Meier analysis of overall survival (OS) for SATB1 (<b>a</b>) and p16 (<b>b</b>). (<b>a</b>) The OS of SATB1-positive patients was characterized by a population half-life of 4.5 ± 1.2 months and a survival plateau of 87 ± 1%. The SATB1-negative group demonstrated a much longer half-life of 84 ± 13.2 months; however, a survival plateau was not observed during the follow-up; the last survival probability observed was 72% after 12 years of follow-up. (<b>b</b>) Patients positive for p16 staining showed a half-life of 1.4 ± 0.77 months and a survival probability plateau of 75 ± 1.3%. Patients who were negative for p16 exhibited a half-life of 58 ± 6.1 months and the same survival plateau probability of 71 ± 1.4%. Abbreviations: neg., negative; pos., positive, mo., month.</p>
Full article ">Figure 3
<p>Kaplan–Meier analysis of progression-free survival (PFS) for SATB1 (<b>a</b>) and p16 (<b>b</b>). (<b>a</b>) SATB1-positive patients relapsed at a rate characterized by a half-life of 14.5 ± 1 months and a probability of relapse settled at the plateau of 87 ± 1%. On the other hand, SATB1-negative patients relapsed at a slower rate with a half-life of 31.1 ± 1 months, eventually reaching a plateau of 62 ± 0.5% (<b>b</b>) p16-positive patients exhibited a faster rate of relapse (PFS half-life = 0.97 ± 0.57 months) than the p16-negative patients (PFS half-life = 13.6 ± 0.68 months). However, the probability of relapse settled to an almost identical plateau in both groups, that is, 68 ± 0.5% for p16- patients and 67 ± 2% for p16+ patients. Abbreviations: neg., negative; pos., positive, mo., month.</p>
Full article ">Figure 4
<p>Stratification of the patient cohort by SATB1 and p16. (<b>a</b>) The SATB1+/p16- group (<span class="html-italic">n</span> = 12) of patients seems to have the best prognosis. (<b>b</b>) Dividing the study population into three subpopulations based on SATB1 and p16 expression resulted in a more parsimonious and accurate model than the one that ignored such information regarding OS (<span class="html-italic">p</span> &lt; 0.0001, ΔAIC = 147.9, ER &gt; 100). SATB1+/p16- patients reached the survival plateau of survival probability at 90.7 ± 0.54%; on the other hand, SATB1-/p16- patients had the last observed survival probability of 74% and a predicted, but not observed, survival plateau at 60 ± 0.5%. (<b>c</b>) If PFS was analyzed in the same way, it turned out that SATB1-/p16- and p16+ subpopulations follow the same dynamic (<span class="html-italic">p</span> = 0.7, ΔAIC = −9.672, ER = 1/126), with a half-life of 14.7 ± 1 months and a plateau of 63.4 ± 0.7%. On the other hand, the PFS curve of the SATB1+/p16- population was almost identical to the OS curve of the same group. Abbreviations: mo., month; AIC, Akaike information criterion; OS, overall survival; PFS, progression-free survival; ER, evidence ratio.</p>
Full article ">Figure 5
<p>Correlation of genetic signatures with SATB1 and p16. The SATB1 transcript positively correlated with B cell differentiation, B cell apoptosis genetic signatures and inversely correlated with response to xenobiotic. P16 expression correlated inversely with pro B cell differentiation gene expression signature.</p>
Full article ">
21 pages, 7244 KiB  
Article
In Doxorubicin-Adapted Hodgkin Lymphoma Cells, Acquiring Multidrug Resistance and Improved Immunosuppressive Abilities, Doxorubicin Activity Was Enhanced by Chloroquine and GW4869
by Naike Casagrande, Cinzia Borghese, Michele Avanzo and Donatella Aldinucci
Cells 2023, 12(23), 2732; https://doi.org/10.3390/cells12232732 - 29 Nov 2023
Cited by 2 | Viewed by 1610
Abstract
Classical Hodgkin lymphoma (cHL) is a highly curable disease (70–80%), even though long-term toxicities, drug resistance, and predicting clinical responses to therapy are major challenges in cHL treatment. To solve these problems, we characterized two cHL cell lines with acquired resistance to doxorubicin, [...] Read more.
Classical Hodgkin lymphoma (cHL) is a highly curable disease (70–80%), even though long-term toxicities, drug resistance, and predicting clinical responses to therapy are major challenges in cHL treatment. To solve these problems, we characterized two cHL cell lines with acquired resistance to doxorubicin, KM-H2dx and HDLM-2dx (HRSdx), generated from KM-H2 and HDLM-2 cells, respectively. HRSdx cells developed cross-resistance to vinblastine, bendamustin, cisplatin, dacarbazine, gemcitabine, brentuximab vedotin (BV), and γ-radiation. Both HDLM-2 and HDLM-2dx cells had intrinsic resistance to BV but not to the drug MMAE. HDLM-2dx acquired cross-resistance to caelyx. HRSdx cells had in common decreased CD71, CD80, CD54, cyt-ROS, HLA-DR, DDR1, and CD44; increased Bcl-2, CD58, COX2, CD26, CCR5, and invasive capability; increased CCL5, TARC, PGE2, and TGF-β; and the capability of hijacking monocytes. In HRSdx cells less sensitive to DNA damage and oxidative stress, the efflux drug transporters MDR1 and MRP1 were not up-regulated, and doxorubicin accumulated in the cytoplasm rather than in the nucleus. Both the autophagy inhibitor chloroquine and extracellular vesicle (EV) release inhibitor GW4869 enhanced doxorubicin activity and counteracted doxorubicin resistance. In conclusion, this study identifies common modulated antigens in HRSdx cells, the associated cross-resistance patterns, and new potential therapeutic options to enhance doxorubicin activity and overcome resistance. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Lymphomas)
Show Figures

Figure 1

Figure 1
<p>Characteristics of HRS and HRSdx cells. (<b>A</b>) Phase-contrast photo-micrographs of HRS and HRSdx cells. (<b>B</b>) Morphological images of HRS (upper panels) and HRSdx cells (red, lower panels) obtained after May–Grünwald–Giemsa staining (MGG). (Magnification, ×20; scale bar, 10 µm.) (<b>C</b>) Growth curves of HRS and HRSdx cells. The number of viable cells was evaluated via trypan blue dye exclusion assays. The calculated doubling times (DTs, in days) for each cell line were reported in the figure. (<b>D</b>) Clonogenic growth. Cells were seeded in medium containing 0.8% methylcellulose. After 14 days, aggregates with ≥40 cells were scored as colonies. Values (total number of colonies) are the mean ± SD of eight replicates of three independent experiments. (<b>E</b>) HRS and HRSdx cells were lysed, and NF-kB p65 transcription factor activity was analyzed in nuclear extracts using the Transcription Factor Kit (p65). Results are represented as the percent of control (activity HRSdx respect to HRS parental cells) and are the mean ± SD of three independent experiments. Chemotaxis assays in Boyden chambers. (<b>F</b>) Migration of HRS and HRSdx cells through fibronectin-coated (20 µg/mL) chambers towards 20% complete medium. Data are the percentages of cells that migrated from the serum-free upper chamber to the lower complete medium chamber. (<b>G</b>) Invasion. Migration of HRS and HRSdx cells through Matrigel-coated (50 ug/mL) chambers towards 20% complete medium. Data are the percentages of cells that migrated from the Matrigel-coated upper chamber to the lower complete medium chamber. (<b>H</b>) Western blot analysis for ROCK, RhoA, and vinculin in HRS and HRSdx cells. Images were acquired using a ChemiDoc XRS system (Bio-Rad). (<b>I</b>) Flow cytometry expression of CCR5 and CXCR4 in HRS and HRSdx cells. Red arrows indicate up-regulated antigens. Mean fluorescence intensities are reported in the boxes. * <span class="html-italic">p</span> &lt; 0.05 HRSdx vs. HRS cells.</p>
Full article ">Figure 2
<p>Phenotypes of HRS and HRSdx cells. Flow cytometry assay of molecules expressed by HRS and HRSdx cells. Representative flow cytometry histograms showing the expression of (<b>A</b>) survival factors and antiapoptotic molecules, (<b>B</b>) markers of the putative cancer stem cells, and (<b>C</b>) molecules involved in the interactions with collagen and stromal cells (<b>D</b>) or with white blood cells (lymphocytes, monocytes, eosinophils, and mast cells). Mean fluorescence intensities are reported in the boxes. Red arrows indicate up-regulated antigens and black arrows down-modulated antigens in doxorubicin resistant HRSdx cells with respect to parental HRS cells. (<b>E</b>) Venn diagrams showing the molecules modulated in both HRSdx cells and those specifically * up-regulated (red) or * down-regulated (blue) in KM-H2dx and HDLM-2dx.</p>
Full article ">Figure 3
<p>Tumor cell expression and secretion of immunosuppressive molecules and monocyte immunosuppressive education by tumor cell conditioned medium (CM). (<b>A</b>) Flow cytometry assay of immunosuppressive molecules expressed by HRS and HRSdx cells. Red arrows indicate up-regulated antigens and black arrows down-modulated antigens in HRSdx cells with respect to HRS cells. (<b>B</b>) Cytokines secreted by HRS and HRSdx cells cultured for 72 h in complete medium. Their concentrations were evaluated by an ELISA assay and reported as pg × 10<sup>6</sup> cells, excluding L-lactate (*, ng/10<sup>6</sup> cells). Values for KM-H2 and HDLM-2 are shown in the respective insert. Bar charts report the fold-increase in the concentration of each chemokine secreted by HRSdx cells with respect to HRS cells. (<b>C</b>) Monocytic THP-1 cells were cultured with HRS-CM and HRSdx-CM, and then, CD206, PDL-1, and IDO expression was evaluated via flow cytometry. Mean fluorescence intensities are reported in the boxes. Red arrows indicate antigens up-regulated by HRSdx-CM with respect to HRS-CM. (<b>D</b>) Immunosuppression (IS). Schematic representation of common HRSdx modifications in cells leading to immunosuppression (antigens, cytokines, monocytes, tumor education). The red arrow indicates up-regulated antigens and the black arrow down-modulated antigens (HRSdx respect to HRS cells).</p>
Full article ">Figure 4
<p>Cross-resistance pattern and γ-radiation activity in HRS and HRSdx cells. (<b>A</b>) The resistance factor (RF) value is the ratio of the HRSdx IC<sub>50</sub> (KM-H2dx and HDLM-2dx) over the HRS IC<sub>50</sub> (KM-H2 and HDLM-2). RFs are reported in ascending order. RF &lt; 1 indicates cross-sensitivity (CS), and RF ≥1 indicates cross-resistance (CR). An RF ranging from 1 to 2 indicates low CR, an RF from 2 to 5 indicates moderate CR, and an RF ≥ 5 indicates high CR. (<b>B</b>–<b>D</b>) Tumor cells were treated with γ-radiation (0–12 Gy). Then, cell viability, clonogenic growth, and cell cycle distribution were evaluated. (<b>B</b>) Cells were double-stained with Annexin-V-FITC and 7AAD and analyzed via flow cytometry. Bar charts show the percentage of viable cells (Annexin-V and 7AAD negative cells). (<b>C</b>) Clonogenic growth assay. Untreated and γ-radiation-treated cells were seeded in medium containing 0.8% methylcellulose and cultured for 14 days; aggregates with ≥40 cells were scored as colonies. Values (total number of colonies) are the mean ± SD of eight replicates. (<b>D</b>) Bar charts show the percentage of cells in each cell cycle phase, evaluated after propidium iodide staining and flow cytometry analysis. (<b>E</b>) Representative cytofluorimetric histograms of the cell cycle progression after γ-radiation treatment. Results are the mean ± SD of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 HRSdx vs. parental HRS cells.</p>
Full article ">Figure 5
<p>Expression of drug transporters, uptake, distribution, and DNA damage via doxorubicin in HRS and HRSdx cells. (<b>A</b>) Relative mRNA expression of MDR1/ABCB1 and MRP1/ABCC1 in HRS and HRSdx cells using GAPDH gene expression as internal control. (<b>B</b>) Western blot for MDR1, MRP1, and α-tubulin expression. (<b>C</b>) Flow cytometry-based doxorubicin accumulation assay. HRS and HRSdx cells were incubated with doxorubicin (0–200 ng/mL) for 2 h. Then, the percentage of red fluorescence-positive cells was evaluated via flow cytometry. (<b>D</b>) Cells were incubated with doxorubicin (DOX, 1 µg/mL). After 2 h, doxorubicin internalization and distribution were evaluated via confocal microscopy. (<b>E</b>) HRS and HRSdx cells were incubated for 24 h with doxorubicin (KM-H2 IC<sub>90</sub> = 100 ng/mL and HDLM-2 IC<sub>90</sub> = 175 ng/mL). Then, γ-H2AX expression was evaluated via flow cytometry. (<b>F</b>) KM-H2dx and HDLM-2dx cells were incubated for 24 h with doxorubicin (KM-H2dx IC<sub>90</sub> = 300 ng/mL and HDLM-2dx IC<sub>90</sub> = 450 ng/mL). γ-H2AX expression was evaluated via flow cytometry. Results are the mean ± SD of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 HRSdx vs. parental HRS cells.</p>
Full article ">Figure 6
<p>Sensitivity of HRS and HRSdx cells to oxidative stress. HRS and HRSdx cells were treated with H<sub>2</sub>O<sub>2</sub> (0–0.5 mM). (<b>A</b>) After 1 h, cell viability was evaluated with a trypan blue dye exclusion assay. (<b>B</b>) Alternatively, cells were double-stained with Annexin-V-FITC and 7AAD and analyzed via flow cytometry. Bar charts show the percentage of viable cells (Annexin-V- and 7AAD-negative cells). (<b>C</b>) HRS and HRSdx cells were treated for 24 h with H<sub>2</sub>O<sub>2</sub> (0.25 mM). After 24 h, cell viability was evaluated with a trypan blue dye exclusion assay. (<b>D</b>) TrxR enzymatic activity was evaluated using a TrxR assay kit and expressed as U/mg of protein. Results are the mean ± SD of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 HRSdx vs. parental HRS cells.</p>
Full article ">Figure 7
<p>Chloroquine and GW4869 enhance cell death induced by doxorubicin. HRS and HRSdx cells were cultured with doxorubicin (DOX) in the presence or not in the presence of (<b>A</b>) a non-toxic concentration of chloroquine (CQ) (2.5 µM) or (<b>B</b>) GW4869 (2 µM). After 72 h, cell viability was evaluated via a trypan blue dye exclusion assay. Results are the mean ± SD of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05 DOX vs. DOX plus CQ or DOX plus GW4869. Arrows indicate the IC<sub>50</sub> of doxorubicin, in the presence or not in the presence of CQ or GW4869. The difference in the IC<sub>50</sub> is shown by the horizontal black double-headed arrow.</p>
Full article ">

Review

Jump to: Research

21 pages, 2462 KiB  
Review
Tumour Microenvironment: The General Principles of Pathogenesis and Implications in Diffuse Large B Cell Lymphoma
by Stanislavs Sinkarevs, Boriss Strumfs, Svetlana Volkova and Ilze Strumfa
Cells 2024, 13(12), 1057; https://doi.org/10.3390/cells13121057 - 18 Jun 2024
Viewed by 1384
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma worldwide, constituting around 30–40% of all cases. Almost 60% of patients develop relapse of refractory DLBCL. Among the reasons for the therapy failure, tumour microenvironment (TME) components could be [...] Read more.
Diffuse large B cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma worldwide, constituting around 30–40% of all cases. Almost 60% of patients develop relapse of refractory DLBCL. Among the reasons for the therapy failure, tumour microenvironment (TME) components could be involved, including tumour-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), tumour-associated neutrophils (TANs), cancer-associated fibroblasts (CAFs), and different subtypes of cytotoxic CD8+ cells and T regulatory cells, which show complex interactions with tumour cells. Understanding of the TME can provide new therapeutic options for patients with DLBCL and improve their prognosis and overall survival. This review provides essentials of the latest understanding of tumour microenvironment elements and discusses their role in tumour progression and immune suppression mechanisms which result in poor prognosis for patients with DLBCL. In addition, we point out important markers for the diagnostic purposes and highlight novel therapeutic targets. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Lymphomas)
Show Figures

Figure 1

Figure 1
<p>Role of tumour-associated macrophages (TAM) in tumour microenvironment (TME). Figure shows pro-tumourous effects of TAM-M2 cells as suppressing of cytotoxic and helper T cells, stimulating angiogenesis and dissemination of the tumour. The TAM-M1 type has an anti-tumourous function by cytotoxicity and phagocytosis.</p>
Full article ">Figure 2
<p>Role of myeloid-derived suppressor cells (MDSCs) in tumour microenvironment (TME). Figure shows negative effects mostly aimed at immune suppression in the TME.</p>
Full article ">Figure 3
<p>Role of tumour-associated neutrophils (TANs) in the tumour microenvironment. N1-TANs have shown cytotoxicity by producing toxic granules and have anti-tumourous effects. N2-TANs, conversely, are pro-tumourous, by increasing DNA damage to the tumour, which leads to the new mutations in the DNA, suppressing cytotoxic T cells and stimulating angiogenesis.</p>
Full article ">Figure 4
<p>Different types of CD8+ T cells (Tc): Tc1, Tc2, Tc9, and Tc17 and their potential role on the tumour microenvironment (TME). Also illustrated is the immune-suppressing function of the T regulatory (Treg) cells.</p>
Full article ">
Back to TopTop