1
views
0
recommends
+1 Recommend
2 collections
    0
    shares

      Call for Papers: Digital Diagnostic Techniques

      Submit here before August 31, 2024

      About Pathobiology: 3.5 Impact Factor I 8.5 CiteScore I 1.088 Scimago Journal & Country Rank (SJR)

      Call for Papers: Beyond Biology: The Crucial Role of Sex and Gender in Oncology

      Submit here before July 31, 2024

      About Oncology Research and Treatment: 2.0 Impact Factor I 3.2 CiteScore I 0.521 Scimago Journal & Country Rank (SJR)

      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Malaria as Potential Aetiology and Treatment Complicating Factor in DLBCL Patient: A Case Report

      case-report

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Malaria is known to be a significant risk factor and also a potential complicating factor during the treatment of lymphoid malignancy. There has not been a reported case of malaria reactivation that occurred weeks after cytotoxic chemotherapy completion, especially in non-endemic regions. Our patient was a 47-year-old man with a history of repeated falciparum malaria infection experiencing 2 months of progressive unilateral nasal blockage and recurrent anterior epistaxis, which was diagnosed as diffuse large B-cell lymphoma (DLBCL) through pathological examination. He was treated with six cycles of classical R-CHOP, resulting in complete remission. One month after remission, he experienced shivering, fever, sweating, and a return to normal temperature, which repeated irregularly for roughly 1 week. His laboratory result showed anaemia, leucopenia, and profound thrombocytopenia. Immunochromatographic testing (ICT) confirmed the diagnosis of falciparum malaria. This case was considered a relapse since our centre is not in the malaria-endemic region. He was cured with a combination of dihydroartemisinin-piperaquine and primaquine. Our case demonstrated the duality of malaria as potential aetiology and treatment complicating factor in DLBCL.

          Related collections

          Most cited references17

          • Record: found
          • Abstract: found
          • Article: not found

          Two levels of protection for the B cell genome during somatic hypermutation.

          Somatic hypermutation introduces point mutations into immunoglobulin genes in germinal centre B cells during an immune response. The reaction is initiated by cytosine deamination by the activation-induced deaminase (AID) and completed by error-prone processing of the resulting uracils by mismatch and base excision repair factors. Somatic hypermutation represents a threat to genome integrity and it is not known how the B cell genome is protected from the mutagenic effects of somatic hypermutation nor how often these protective mechanisms fail. Here we show, by extensive sequencing of murine B cell genes, that the genome is protected by two distinct mechanisms: selective targeting of AID and gene-specific, high-fidelity repair of AID-generated uracils. Numerous genes linked to B cell tumorigenesis, including Myc, Pim1, Pax5, Ocab (also called Pou2af1), H2afx, Rhoh and Ebf1, are deaminated by AID but escape acquisition of most mutations through the combined action of mismatch and base excision repair. However, approximately 25% of expressed genes analysed were not fully protected by either mechanism and accumulated mutations in germinal centre B cells. Our results demonstrate that AID acts broadly on the genome, with the ultimate distribution of mutations determined by a balance between high-fidelity and error-prone DNA repair.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Plasmodium Infection Promotes Genomic Instability and AID-Dependent B Cell Lymphoma.

            Chronic infection with Plasmodium falciparum was epidemiologically associated with endemic Burkitt's lymphoma, a mature B cell cancer characterized by chromosome translocation between the c-myc oncogene and Igh, over 50 years ago. Whether infection promotes B cell lymphoma, and if so by which mechanism, remains unknown. To investigate the relationship between parasitic disease and lymphomagenesis, we used Plasmodium chabaudi (Pc) to produce chronic malaria infection in mice. Pc induces prolonged expansion of germinal centers (GCs), unique compartments in which B cells undergo rapid clonal expansion and express activation-induced cytidine deaminase (AID), a DNA mutator. GC B cells elicited during Pc infection suffer widespread DNA damage, leading to chromosome translocations. Although infection does not change the overall rate, it modifies lymphomagenesis to favor mature B cell lymphomas that are AID dependent and show chromosome translocations. Thus, malaria infection favors mature B cell cancers by eliciting protracted AID expression in GC B cells. PAPERCLIP.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A Molecular Link between Malaria and Epstein–Barr Virus Reactivation

              Introduction Epstein–Barr virus (EBV) is a human γ-herpes virus that establishes a persistent infection in >90% of the world's population. Like other herpes viruses, EBV has two alternative lifestyles: latent (non-productive) infection, and lytic (productive) replication. Following primary infection, EBV persists within memory B lymphocytes in a latent state for the life of the host. A low level of reactivation into lytic replication allows viral shedding into the saliva and transmission of the virus in vivo [1]. The lifelong persistent infection established by EBV is harmless in almost every host and rarely causes disease, unless the host–virus equilibrium is upset. Thus, viral persistence represents a balance between viral latency, viral replication, and host immune responses. The lytic phase of viral replication can be triggered in vitro by a variety of reagents and stimuli, including halogenated pyrimidine [2], phorbol esters [3], calcium ionophores [4], transforming growth factor β [5], butyrate [6], and triggering of the B cell receptor (BCR) with anti-immunoglobulin (anti-Ig) antibody (Ab) [7]. Less is known about the physiological stimuli that control activation of the virus productive cycle in vivo, although replication seems to occur following plasma cell differentiation [8]. It has been well documented that EBV is causally associated with various malignancies, including endemic Burkitt lymphoma (BL), nasopharyngeal carcinoma, and B cell lymphoma, in immunocompromised hosts [9]. Both EBV infection and intense exposure to Plasmodium falciparum malaria (holoendemic malaria) are recognized cofactors in the pathogenesis of BL, which is the most common paediatric cancer in equatorial Africa, accounting for up to 74% of childhood malignant disorders [10]. Development of BL, a B cell malignancy, is heralded by high Ab titers to replicative antigens indicative of EBV reactivation [11]. Recent reports indicate that the impact of malaria infection on EBV persistence is reflected by an increased viral replication. Children living in malaria-endemic areas have an elevated EBV load [12,13], and acute malaria infection leads to increased levels of circulating EBV that are cleared following anti-malaria treatment [14]. The mechanisms that may lead to viral reactivation during P. falciparum malaria are not well understood. The identification of a polyclonal B cell activator and Ig-binding protein in P. falciparum is of particular relevance in this context. We demonstrated that the cystein-rich inter-domain region 1α (CIDR1α) of the P. falciparum erythrocyte membrane protein 1 (PfEMP1) induces proliferation and activation of B cells, preferentially of the memory subset, where EBV is known to reside [15,16]. To understand the relative contribution of malarial antigens on EBV reactivation, we used the prototype EBV-positive BL cell line Akata as a model to determine whether CIDR1α could induce reactivation of the EBV lytic cycle in latently infected B cells. Furthermore, we analyzed the effect of the CIDR1α on freshly isolated peripheral blood mononuclear cells (PBMCs) from EBV-positive healthy donors and from children with BL living in malaria-endemic areas. The results support the hypothesis that CIDR1α is one of the molecules involved in EBV reactivation during the course of malaria infection. Our data provide new insights into how malaria infection may contribute to BL development. Results P. falciparum–Infected Red Blood Cells and Purified CIDR1α Bind to EBV-Carrying B Cells During the blood stage of P. falciparum malaria, infected red blood cells (iRBCs) express high levels of PfEMP1, reaching their maximum at the trophozoite stage (28–32 h post-invasion). The CIDR1α domain of PfEMP1 (clone FCR3S1.2-var1) has a multi-adhesive phenotype and binds to different cell surface receptors, such as CD36, PECAM-1 (CD31), and immunoglobulins (Igs) [17]. CIDR1α also binds to isolated B cells via an interaction that involves surface Igs [15]. To establish whether iRBCs and the soluble form of CIDR1α interact with EBV-carrying B cells, we used the EBV-positive BL cell line Akata as a model. Akata cells stained with PKH67 (green) were co-incubated with PKH26 (red)-stained uninfected red blood cells (RBCs), or with enriched iRBCs at the trophozoite stage (28 h post-invasion, 75%–80% final parasitemia), at a ratio of 1:2, respectively. RBCs did not bind to Akata (Figure 1A), but co-incubation with iRBCs led to the formation of conjugates that varied in size but frequently involved two to five iRBCs/Akata cell (Figure 1B). A higher magnification of the conjugates showed a polarization of the iRBC, where the parasites were found at the proximity of the membrane's tight junction between the two cell types (Figure 1C). Figure 1 P. falciparum iRBCs and Soluble CIDR1α Bind to the EBV-Positive B Cell Line Akata (A–C) Akata cells stained with PKH67 (green) were co-incubated with RBCs stained with PK26 (red) (A) or with purified iRBCs (B, C) at a ratio of 1:2 (Akata:iRBC). Binding was evaluated by light microscopy (left panel) and immunofluorescence microscopy (right panel). (C) Magnification of the binding between iRBCs and Akatas. Localization of the parasite in the iRBC (indicated by white arrows) was revealed using phase contrast microscopy (left panel). (D–F) Akata cells were incubated with soluble GST (1 μM) (D) or CIDR1α (1 μM) (E) for 1 h at RT and stained with anti-GST Ab and anti-mouse Alexa-488-conjugated Ab. The binding was evaluated by light microscopy (left panel, [D, E]), immunofluorescence microscopy (right panel, [D, E]), and fluorescent-activated cell sorting (FACS) analysis (F). Being that CIDR1α is a domain expressed on iRBCs with multi-adhesive phenotypes [17], we investigated its ability to bind Akata cells. A soluble form of CIDR1α was produced as a glutathione-S-transferase (GST) fusion protein, and the GST protein alone was used as control. Immunofluorescence studies with anti-GST fluorescent Abs demonstrated that CIDR1α, but not the GST control, binds to the membrane of Akata cells (Figure 1D and 1E). Flow cytometry analysis showed a peak shift representing an increased mean fluorescence intensity (MFI) as compared to the MFI values obtained when Akata cells were incubated with GST control protein or with the isotype control Ab (Figure 1F). Thus, both iRBCs and the recombinant CIDR1α domain of PfEMP1 bind to the EBV-carrying B cell line Akata. CIDR1α and iRBC Stimulation Lead to Increased EBV Viral DNA Load in Akata Cells In contrast to the variety of reagents and signals able to induce EBV lytic production in vitro [18], the physiological signals involved in the activation of the virus productive cycle have not been well characterized yet, although plasma cell differentiation seems to represent one such trigger [8]. Anti-Ig treatment, which leads to BCR signalling [19], has served as a relevant in vitro model of reactivation for inducing virus replication in some EBV-carrying BL cell lines, including Akata [7,20]. Because CIDR1α is an Ig-binding protein [17], we analyzed its functional impact on the reactivation of lytic virus production and used the well-characterized Akata cell line model as a read-out system. First, we analyzed whether stimulation of Akata cells with CIDR1α would affect the number of viral DNA copies produced. Cells were cultured with increasing concentrations of CIDR1α, GST (range 0,5–2 μM, corresponding to 25–100 μg/mL), or in medium alone. After 48 h, we quantified the EBV viral DNA copy number in the cultures (cells + supernatant) by monitoring the EBV LMP1 gene, which is present as a single copy in the virus genome. As shown in Figure 2A and 2B, stimulation with CIDR1α increased the viral DNA load in a dose-dependent manner. Cells incubated with GST contained numbers of EBV genomes comparable to that of cells cultured in medium alone. In two out of four independent experiments, there was a statistical significance in relative increase of EBV load between the concentrations of 0,5 μM and 2 μM (p = 0,03). Figure 2 CIDR1α and iRBC Stimulation Lead to Increased Viral Load in Akata Cells (A) Akata cells were cultured in medium alone (referred to as Ag concentration 0) with GST (white circles) or with CIDR1α (black circles) at protein concentrations ranging from 0,5 to 2,5 μM in the presence or absence of zVAD. After 48 h of incubation, the viral genome copy number was determined by real-time PCR. Results from a representative experiment (out of four independent experiments) are expressed as numbers of EBV genome copies per μg of total DNA. (B) Akata cells were cultured with GST or CIDR1α (0–2,5 μM), and with anti-Ig (10 μg/mL). After 48 h of incubation, the viral genome copy number was determined by real-time PCR. Results are expressed as percentage of relative increase in EBV copy number in cultures stimulated with CIDR1α versus cultures stimulated with GST, and represent the mean of four independent experiments ± standard deviation. *, p 2vs0,5 μM = 0,03. (C) Akata cells were cultured with increasing concentrations of crude extracts from iRBCs and RBCs. After 48 h of incubation, the viral genome copy number was determined by real-time PCR. Results from one representative experiment (out of three performed) are expressed as percentage of relative increase in EBV copy numbers in cultures stimulated with iRBCs versus cultures stimulated with RBC extracts. We have recently demonstrated that CIDR1α induces B cell proliferation and protects B cells from apoptosis [16]. It could be then argued that the augmented viral load might simply result from a net increase in the number of cells in the culture. Cell cycle analysis performed by propidium iodide staining after 24 and 48 h of incubation did not reveal any significant change in the proportion of dead ( 95% as revealed by cytofluorimetric (FACS) analysis after staining with the pan–T cell Ab CD3 and the monocyte marker CD14. Parasites. The highly rosetting and auto-agglutinating P. falciparum parasite clone FCR3S1.2 was obtained through cloning by micromanipulation of the mother clone FCR3 [33] and cultured according to standard methods. Parasites were maintained and expanded in RBCs (O Rh+) at 5% haematocrit. iRBCs were cultured in RPMI 1640 medium supplemented with 10% B+ human serum, 0,6% HEPES (GIBCO-BRL), 25 μg/mL gentamycin (GIBCO-BRL), 0,25% sodium bicarbonate (GIBCO-BRL), and 2 mM L-glutamine (GIBCO-BRL). Trophozoites were enriched by magnetic-assisted cell sorting, using a Vario-MACS (Miltenyi Biotec, http://www.miltenyibiotec.com). Synchronous P. falciparum parasite cultures, grown 24–28 h post-invasion, were washed twice in RPMI 1640 and resuspended in 5–10 mL of phosphate-buffered saline (PBS) with 2% bovine serum albumin (BSA). Rosettes were disrupted mechanically by repeated passage through a 0,6-mm-thick injection needle. The material was slowly added to a MACS separation column mounted in the magnet and rinsed with 50 mL of 2% BSA in PBS. The iRBCs were eluted in 50 mL of 2% BSA in PBS after removing the column from the magnet, spun down at 500g, and resuspended in 1 mL of RPMI 1640. Following enrichment, the parasitemia was evaluated by fluorescence microscopy after addition of one drop of acridine orange (10 μg/mL). Parasite extracts. Parasite extracts were prepared as previously described [34]. Briefly, parasites at trophozoite stage (iRBCs, 28 h post-invasion) or RBCs were washed, resuspended in PBS, and sonicated (25w) on ice at short intervals for 2 min. The extracts were then centrifuged at 500g for 10 min at 4 °C and filter-sterilized. After determination of protein concentration by Bradford Assay (Bio-Rad, http://www.bio-rad.com), the extracts were diluted with PBS. Recombinant CIDR1α. The sequence of the CIDR1α domain of the PfEMP1 from clone FCR3S1.2-var1 was optimized for codon adaptation in Escherichia coli, re-synthesized chemically (GeneArt, http://www.geneart.com), cloned into the pGEX4T-1 vector (Amersham Pharmacia, http://www.gelifesciences.com), and expressed in E. coli BL21 CodonPlus-RIL (Stratagene, http://www.stratagene.com) as fusion protein with the GST. Non-recombinant pGEX4T-1 was used to produce GST as control protein. Protein purification was carried out according to an optimized protocol [35], and the recombinant protein was purified on glutathione-sepharose column GSTrap FF (Amersham Pharmacia) according to the manufacturer's instructions. Throughout the paper, we refer to the recombinant CIDR1α-GST fusion protein as CIDR1α, while GST is referred to as control protein. Binding assays. Akata iRBCs: Akata cells and enriched iRBCs (trophozoite stage) were stained with PKH67 (green) and PKH26 (red) (Sigma-Aldrich), respectively, according to manufacturer's instructions. The binding was evaluated by fluorescence microscopy after co-incubation in PBS at room temperature (RT) for 1 h at a 1:2 ratio (Akata:iRBC). Akata CIDR1α: Cells incubated for 1 h at RT in PBS containing GST or CIDR1α (1 μM) were washed twice in PBS and incubated for 30 min at RT with anti-GST Abs (Sigma-Aldrich) diluted 1:500 in PBS. After two washes with PBS, anti-mouse IgG Alexa-488-conjugated Ab (Molecular Probes, http://probes.invitrogen.com) diluted 1:100 in PBS was added for 30 min at 4 °C. Cell binding was analyzed by fluorescence microscopy, and the fluorescence intensity was measured with a FACSCalibur flow cytometer and analyzed with Cell Quest Pro software (Becton Dickinson, http://www.bd.com). Stimulation assays. Twenty-four hours before experiments, Akata and Akata-GFP cells were suspended at a concentration of 106/mL in complete medium and complete medium containing G418 (500 μg/mL), respectively. Fresh PBMCs were washed with PBS and resuspended in complete medium at a concentration of 105 cells per ml. The cells were then seeded in 96-well plates and cultured in medium alone or with increasing concentrations of CIDR1α, GST (range 0–4 μM, corresponding to 0–200 μg/mL), or with anti-Ig (10 μg/mL) (Jackson ImmunoResearch, http://www.jacksonimmuno.com). After 48 h, cells were harvested for analysis. All tests were set up in multiple replicates. EBV DNA detection by real-time PCR. DNA was extracted from cells and supernatants using the QIAamp Blood kit (Qiagen, http://www.qiagen.com) according to manufacturer's instructions and eluted in 50 μL of DEPC-treated water (Ambion, http://www.ambion.com). Purity and DNA concentration were evaluated using a NanoDrop ND-1000 spectrophotometer (http://www.nanodrop.com). The PCR primers and probe used for the quantification of EBV genomes were selected from the LMP-1 gene as previously described [13,14]. The primers used were the EBV-LMP1 forward primer 5′-AAGGTCAAAGAACAAGGCCAAG-3′ and the EBV-LMP1 reverse primer 5′-GCATCGGAGTCGG-3′. The fluorogenic probe (PE Applied Biosystems, http://www.appliedbiosystems.com) was synthesized using a FAM reporter molecule attached to the 5′ end and a TAMRA quench-er linked to the 3′ end (5′-AGGAGCGTGTCCCCGTGGAGG-3′). A standard curve was prepared using serial dilutions of DNA derived from the EBV-positive BL line Namalwa that contains two copies of EBV genome per cell. Detection was performed using an ABI Prism 7700 Sequence detection System (PE Applied Biosystems). Briefly, cycling parameters were 50 °C for 2 min, 95 °C for 10 min, 45 cycles 95 °C for 15 s, and 60 °C for 1 min. The EBV DNA copy number was calculated as mean of triplicates. Cell cycle analysis. Cells were washed twice in ice-cold PBS and resuspended in 100 μL of PBS containing 50 μg propidium iodide/mL and 0.1% (v/v) triton X-100. After incubation at 4 °C for 8 h, cell DNA analysis was performed by flow cytometry (FACSCalibur, Becton Dickinson) using Cell Quest Pro software (Becton Dickinson). p-Values ≤ 0,05 were regarded as statistically significant. Western blot analysis. Expression of BZLF1, the early lytic antigen, was assessed using a specific mouse Ab (DAKO, http://www.dako.com). Total extracts from 106 cells were separated in 10% SDS polyacrylamide gels, blotted onto nitrocellulose filters probed with 1:500 dilution of the DAKO monoclonal Ab for 1 h at RT, and then reacted with horseradish-peroxidase–labelled anti-mouse Abs (Amersham). Immunocomplexes were visualized by enhanced chemiluminescence according to the manufacturer's instructions (Amersham). To control for equal loading, the total protein concentration of each sample was checked using Bradford assay. Statistical analysis. Computations were performed with the Prism 4 package (GraphPad Software, http://www.graphpad.com). For parametric data, differences between groups were analyzed with Student t-test; for nonparametric data, differences between groups were analyzed with the Wilcoxon rank sum test. Results are expressed as mean ± SEM, and unless otherwise stated, considered statistically significant at p < 0,05.
                Bookmark

                Author and article information

                Journal
                Case Rep Oncol
                Case Rep Oncol
                CRO
                CRO
                Case Reports in Oncology
                S. Karger AG (Basel, Switzerland )
                1662-6575
                12 May 2023
                Jan-Dec 2023
                12 May 2023
                : 16
                : 1
                : 302-307
                Affiliations
                [a ]Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
                [b ]Department of Internal Medicine, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
                [c ]Department of Radiology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
                [d ]Division of Tropical Medicine and Infectious Disease, Department of Internal Medicine, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
                [e ]Department of Pathological Anatomy, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University/Dr. Sardjito Hospital, Yogyakarta, Indonesia
                Author notes
                Correspondence to: Ibnu Purwanto, ibnupurwanto@ 123456ugm.ac.id
                Article
                530337
                10.1159/000530337
                10176193
                37187685
                ae476cc1-d8d7-4428-a85a-1c9ce8dc1739
                © 2023 The Author(s). Published by S. Karger AG, Basel

                This article is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License (CC BY-NC) ( http://www.karger.com/Services/OpenAccessLicense). Usage and distribution for commercial purposes requires written permission.

                History
                : 12 December 2022
                : 21 March 2023
                : 2023
                Page count
                Figures: 3, References: 17, Pages: 6
                Funding
                No funding was received specifically for this case report.
                Categories
                Case Report

                Oncology & Radiotherapy
                malaria,relapse,diffuse large b-cell lymphoma,risk factor,complication
                Oncology & Radiotherapy
                malaria, relapse, diffuse large b-cell lymphoma, risk factor, complication

                Comments

                Comment on this article