Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 915
1.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 155-160, 2024 May 27.
Article En | MEDLINE | ID: mdl-38814221

In order to explore a new mode for the diagnosis of angioimmunoblastic T-cell lymphoma (AITL), 31 cases of AITL and 28 cases of peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS) were used as the study subjects. Identifying T follicular helper (TFH) cells with CD4, CD10, Bcl-6, and PD-1, identifying proliferative B cells with CD20 and EZH2, identifying proliferative follicular dendritic cells (FDCs) with CD21 and CD23, and analyzing the value of TFH/B/FDC proliferation and immunolocalization in the diagnosis of AITL. (1) Outside the inherent lymphoid follicles, simultaneous proliferation of TFH/B/FDC (a new diagnostic mode) were observed in AITL [83.87%; 26/31], with their immunolocalizations in the same site [83.87%; 26/31], while this phenomenon was not observed in 28 cases of PTCL-NOS (P<0.05). (2) The sensitivity and specificity of using this new mode to diagnose AITL were both high (83.87%, 100%), which was superior to CD2 (100%, 0%), CD3 (100%, 0%), CD4 (100%, 32.14%), CD5 (100%, 25%), CD10 (61.9%, 100%), Bcl-6 (42.86%, 100%), PD-1 (83.87%, 96.43%), and its Youden Index (0.84) was the highest. The areas under the curve (AUC) of CD10, Bcl-6, PD-1, and new mode to diagnosis AITL were 0.81, 0.71, 0.90, and 0.92, respectively, while the new mode had the highest AUC. The simultaneous proliferation of TFH/B/FDC cells outside the inherent lymphoid follicles can be used to assist in the diagnosis of AITL, and the simultaneous spatiotemporal proliferation of TFH/B/FDC cells is a specific immunomorphology of AITL.


Proto-Oncogene Proteins c-bcl-6 , Humans , Female , Male , Middle Aged , Aged , Proto-Oncogene Proteins c-bcl-6/metabolism , Neprilysin/metabolism , Immunoblastic Lymphadenopathy/diagnosis , Immunoblastic Lymphadenopathy/pathology , Dendritic Cells, Follicular/pathology , Dendritic Cells, Follicular/metabolism , Programmed Cell Death 1 Receptor/metabolism , Adult , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Cell Proliferation , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , T Follicular Helper Cells/immunology , T Follicular Helper Cells/metabolism , Receptors, Complement 3d/metabolism , Receptors, Complement 3d/analysis , Antigens, CD20/metabolism , Antigens, CD20/analysis , Lymphoma, T-Cell, Peripheral/diagnosis , Lymphoma, T-Cell, Peripheral/pathology , CD4 Antigens/metabolism , Sensitivity and Specificity , Aged, 80 and over , Immunohistochemistry/methods , ROC Curve
2.
J Exp Clin Cancer Res ; 43(1): 43, 2024 Feb 06.
Article En | MEDLINE | ID: mdl-38321568

BACKGROUND: Angioimmunoblastic T-cell lymphoma (AITL) is a malignancy with very poor survival outcome, in urgent need of more specific therapeutic strategies. The drivers of malignancy in this disease are CD4+ follicular helper T cells (Tfh). The metabolism of these malignant Tfh cells was not yet elucidated. Therefore, we decided to identify their metabolic requirements with the objective to propose a novel therapeutic option. METHODS: To reveal the prominent metabolic pathways used by the AITL lymphoma cells, we relied on metabolomic and proteomic analysis of murine AITL (mAITL) T cells isolated from our established mAITL model. We confirmed these results using AITL patient and healthy T cell expression data. RESULTS: Strikingly, the mAITL Tfh cells were highly dependent on the second branch of the Kennedy pathway, the choline lipid pathway, responsible for the production of the major membrane constituent phosphatidylcholine. Moreover, gene expression data from Tfh cells isolated from AITL patient tumors, confirmed the upregulation of the choline lipid pathway. Several enzymes involved in this pathway such as choline kinase, catalyzing the first step in the phosphatidylcholine pathway, are upregulated in multiple tumors other than AITL. Here we showed that treatment of our mAITL preclinical mouse model with a fatty acid oxydation inhibitor, significantly increased their survival and even reverted the exhausted CD8 T cells in the tumor into potent cytotoxic anti-tumor cells. Specific inhibition of Chokα confirmed the importance of the phosphatidylcholine production pathway in neoplastic CD4 + T cells, nearly eradicating mAITL Tfh cells from the tumors. Finally, the same inhibitor induced in human AITL lymphoma biopsies cell death of the majority of the hAITL PD-1high neoplastic cells. CONCLUSION: Our results suggest that interfering with choline metabolism in AITL reveals a specific metabolic vulnerability and might represent a new therapeutic strategy for these patients.


Immunoblastic Lymphadenopathy , Lymphoma, T-Cell , Lymphoma , Humans , Animals , Mice , Proteomics , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , Immunoblastic Lymphadenopathy/genetics , Immunoblastic Lymphadenopathy/metabolism , Immunoblastic Lymphadenopathy/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Phosphatidylcholines/metabolism , Lymphoma/metabolism , Lymphoma/pathology
3.
Environ Toxicol ; 39(4): 1909-1922, 2024 Apr.
Article En | MEDLINE | ID: mdl-38059649

In the past years, PDE5 has emerged as a promising therapeutic target for many cancers due to its highly upregulated expression. Interestingly, a recent in vitro study by our group has shown the antitumor and chemopotentiating action of sildenafil against T cell lymphoma. Our study showed that lower doses of sildenafil (50 µM) and cisplatin (0.5 µg/mL) exhibited 4% and 23% cytotoxicity against HuT78 cells, respectively, which was dramatically increased up to 50% when treated with both. Hence, the present study was designed to evaluate the antitumor and chemo-potentiating action of sildenafil in a murine model of T cell lymphoma (popularly called as Dalton's lymphoma [DL]). In the present study, DL-bearing mice were administered with vehicle (PBS), sildenafil (5 mg/kg bw), cisplatin (5 mg/kg bw), and sildenafil and cisplatin followed by evaluation of their impact on tumor growth by analyzing various parameters. The apoptosis was assessed by Wright-Giemsa, annexin-V, and DAPI staining. Reactive oxygen species (ROS) level was examined through DCFDA staining. The expression of genes and proteins were estimated by RT-PCR and Western blotting, respectively. The experimental findings of the study demonstrate for the first time that sildenafil inhibits tumor growth and potentiates tumor inhibitory ability of cisplatin by altering apoptosis, glycolysis, ROS homeostasis, and pH regulation in T cell lymphoma-carrying host. In addition, our investigation also showed amelioration of tumor-induced liver and kidney damage by sildenafil. Overall, the experimental data of our study strongly advocate the use and repurposing of SDF in designing promising chemotherapeutic regimens against malignancies of T cells.


Lymphoma, T-Cell , Lymphoma , Mice , Animals , Cisplatin/pharmacology , Reactive Oxygen Species/metabolism , Sildenafil Citrate/pharmacology , Sildenafil Citrate/therapeutic use , Cyclic Nucleotide Phosphodiesterases, Type 5/therapeutic use , Apoptosis , Lymphoma, T-Cell/metabolism , Homeostasis , Glucose/metabolism , Hydrogen-Ion Concentration , Cell Line, Tumor
4.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166722, 2023 08.
Article En | MEDLINE | ID: mdl-37084822

Long intergenic noncoding RNAs (lincRNAs) are differentially expressed in EBV-infected cells and play an essential role in tumor progression. Molecular pathogenesis of lincRNAs in EBV-driven natural killer T cell lymphoma (NKTCL) remains unclear. Here we investigated the ncRNA profile using high-throughput RNA sequencing data of 439 lymphoma samples and screened out LINC00486, whose downregulation was further validated by quantitative real-time polymerase chain reaction in EBV-encoded RNA (EBER)-positive lymphoma, particularly NKTCL. Both in vitro and in vivo studies revealed the tumor suppressive function of LINC00486 through inhibiting tumor cell growth and inducing G0/G1 cell cycle arrest. As mechanism of action, LINC00486 specifically interacted with NKRF to abrogate its binding with phosphorylated p65, activated NF-κB/TNF-α signaling and subsequently enhanced EBV eradication. Solute carrier family 1 member 1 (SLC1A1), upregulated and mediated the glutamine-addiction and tumor progression in NKTCL, was negatively correlated with the expression of NKRF. NKRF specifically bound to the promoter and transcriptionally downregulated the expression of SLC1A1, as evidenced by Chromatin Immunoprecipitation (ChIP) and luciferase assay. Collectively, LINC00486 functioned as a tumor suppressor and counteracted EBV infection in NKTCL. Our study improved the knowledge of EBV-driven oncogenesis in NKTCL and provided the clinical rationale of EBV eradication in anti-cancer treatment.


Lymphoma, T-Cell , Lymphoma , Natural Killer T-Cells , RNA, Long Noncoding , Humans , Herpesvirus 4, Human/genetics , RNA, Long Noncoding/genetics , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Lymphoma/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology
5.
Front Immunol ; 14: 1118681, 2023.
Article En | MEDLINE | ID: mdl-36936927

Introduction: Most T cell receptor (TCR)Vß chain-expressing T cell lymphomas (TCL) including those caused by Human T cell leukaemia virus type-1 (HTLV-1) have poor prognosis. We hypothesised that chimeric antigen receptor (CAR)-mediated targeting of the clonal, lymphoma-associated TCRß chains would comprise an effective cell therapy for TCL that would minimally impact the physiological TCR repertoire. Methods: As proof of concept, we generated CAR constructs to target four TCRVß subunits. Efficacy of the CAR constructs was tested using conventional T cells as effectors (CAR-T). Since invariant NKT (iNKT) cell do not incite acute graft-versus-host disease and are suitable for 'off-the-shelf' immunotherapy, we generated anti-TCRVß CAR-iNKT cells. Results: We show that anti-TCRVß CAR-T cells selectively kill their cognate tumour targets while leaving >90% of the physiological TCR repertoire intact. CAR-iNKT cells inhibited the growth of TCL in vivo, and were also selectively active against malignant cells from Adult T cell leukaemia/lymphoma patients without activating expression of HTLV-1. Discussion: Thus we provide proof-of-concept for effective and selective anti-TCRVß CAR-T and -iNKT cell-based therapy of TCL with the latter providing the option for 'off-the-shelf' immunotherapy.


Human T-lymphotropic virus 1 , Leukemia-Lymphoma, Adult T-Cell , Lymphoma, T-Cell, Peripheral , Lymphoma, T-Cell , Natural Killer T-Cells , Receptors, Chimeric Antigen , Adult , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Leukemia-Lymphoma, Adult T-Cell/therapy , Lymphoma, T-Cell/metabolism
6.
Chem Biol Interact ; 369: 110278, 2023 Jan 05.
Article En | MEDLINE | ID: mdl-36423730

In recent years, studies have reported the role of stress-regulatory hormones, including epinephrine, in regulating the progression of a few cancers. However, the tumor-promoting action of epinephrine is not yet investigated in T cell malignancy, a rare and complicated neoplastic disorder. More so, very little is known regarding the implication of epinephrine in the glucose metabolic rewiring in tumor cells. The present investigation showed that epinephrine enhanced the proliferation of T lymphoma cells through up- and down-regulating the expression of PCNA, cyclin D, and p53, respectively. In addition, epinephrine inhibited apoptosis in T lymphoma cells possibly by increasing the level of BCL2 (an anti-apoptotic protein) and decreasing PARP level (a pro-apoptotic protein). Intriguingly, epinephrine is reported to stimulate glycolysis in T lymphoma cells by increasing the expression of crucial glycolysis regulatory molecules, namely HKII and PKM2, in a HIF-1α-dependent manner. Moreover, augmented production of ROS has been observed in T lymphoma cells, which might be a central player in epinephrine-mediated T cell lymphoma growth. Taken together, our study demonstrates that epinephrine might have a significant role in the progression of T cell lymphoma.


Apoptosis , Lymphoma, T-Cell , Humans , Cell Proliferation , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/metabolism , Apoptosis Regulatory Proteins/metabolism , Glucose/metabolism , Glycolysis , Cell Line, Tumor , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
7.
Oncoimmunology ; 11(1): 2134536, 2022.
Article En | MEDLINE | ID: mdl-36249275

Angioimmunoblastic T-cell lymphoma (AITL) is a malignant hematologic tumor arising from T follicular helper (Tfh) cells. High-throughput genomic sequencing studies have shown that AITL is characterized by a novel highly recurring somatic mutation in RHOA, encoding p.Gly17Val (RHOA G17V). However, the specific role of RHOA G17V in AITL remains unknown. Here, we demonstrated that expression of Rhoa G17V in CD4+ T cells increased cell proliferation and induces Tfh cell specification associated with Pon2 upregulation through an NF-κB-dependent mechanism. Further, loss of Pon2 attenuated oncogenic function induced by genetic lesions in Rhoa. In addition, an abnormality of RHOA G17V mutation and PON2 expression is also detected in patients with AITL. Our findings suggest that PON2 associated with RHOA G17V mutation might control the direction of the molecular agents-based AITL and provide a new therapeutic target in AITL.


Immunoblastic Lymphadenopathy , Lymphoma, T-Cell , Aryldialkylphosphatase/metabolism , Humans , Immunoblastic Lymphadenopathy/genetics , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , NF-kappa B/metabolism , Neoplasm Recurrence, Local , T Follicular Helper Cells , rhoA GTP-Binding Protein/genetics
8.
Apoptosis ; 27(7-8): 606-618, 2022 08.
Article En | MEDLINE | ID: mdl-35725975

Cyclic nucleotide phosphodiesterase 5 (PDE5) has been recently identified to play a crucial role in the progression of many cancers. PDE5 promotes tumorigenesis by dysregulating various cellular processes such as proliferation, apoptosis, angiogenesis, and invasion and migration. Interestingly, multiple studies have reported the promising chemosensitizing potential of PDE5 inhibitor sildenafil in breast, colon, prostate, glioma, and lung cancers. However, to date, the chemosensitizing action of sildenafil is not evaluated in T cell lymphoma, a rare and challenging neoplastic disorder. Hence, the present investigation was undertaken to examine the chemosensitizing potential of sildenafil against T cell lymphoma along with elucidation of possible involvement of altered apoptosis and glucose metabolism. The experimental findings of this study showed that sildenafil enhances the cytotoxic ability of cisplatin by apoptosis induction through altering the levels of apoptosis regulatory molecules: Bcl-2, Bax, cytochrome c (Cyt c), cleaved caspase-3, and poly (ADP-ribose) polymerase (PARP). These molecular alterations were possibly driven by sildenafil through reactive oxygen species (ROS). Sildenafil deregulates glucose metabolism by markedly lowering the expression of glycolysis regulatory molecules, namely glucose transporter 1 (GLUT1), lactate dehydrogenase A (LDHA), hexokinase II (HKII), pyruvate kinase M2 (PKM2), and pyruvate dehydrogenase kinase 1 (PDK1) via suppressing hypoxia-inducible factor 1-alpha (HIF-1α) expression. Hence, sildenafil potentiates the tumor cell killing ability of cisplatin by augmenting ROS production through switching the glucose metabolism from glycolysis to oxidative phosphorylation (OXPHOS). Overall, our study demonstrates that sildenafil might be a promising adjunct therapeutic candidate in designing novel combinatorial chemotherapeutic regimens against T cell lymphoma.


Cisplatin , Lymphoma, T-Cell , Apoptosis , Cell Line, Tumor , Cisplatin/pharmacology , Glucose/metabolism , Glycolysis , Humans , Lymphoma, T-Cell/metabolism , Male , Phosphodiesterase 5 Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Sildenafil Citrate/pharmacology
9.
Am J Pathol ; 192(8): 1186-1198, 2022 08.
Article En | MEDLINE | ID: mdl-35640677

This study reports that hairy and enhancer of split homolog-1 (HES1), known to repress gene transcription in progenitor cells of several cell lineages, was strongly expressed in cells and tissues of T-cell lymphoma expressing the oncogenic chimeric tyrosine kinase nucleophosmin (NPM)-anaplastic lymphoma kinase [ALK; ALK+ T-cell lymphoma (TCL)]. The structural analysis of the Orange domain of HES1 indicated that HES1 formed a highly stable homodimer. Of note, repression of HES1 expression led to inhibition of ALK+ TCL cell growth in vivo. The expression of the HES1 gene was induced by NPM-ALK through activation of STAT3, which bound to the gene's promoter and induced the gene's transcription. NPM-ALK also directly phosphorylated HES1 protein. In turn, HES1 up-regulated and down-regulated in ALK+ TCL cells, the expression of numerous genes, protein products of which are involved in key cell functions, such as cell proliferation and viability. Among the genes inhibited by HES1 was thioredoxin-interacting protein (TXNIP), encoding a protein implicated in promotion of cell death in various types of cells. Accordingly, ALK+ TCL cells and tissues lacked expression of TXNIP, and its transcription was co-inhibited by HES1 and STAT3 in an NPM-ALK-dependent manner. Finally, the induced expression of TXNIP induced massive apoptotic cell death of ALK+ TCL cells. The results reveal a novel NPM-ALK-controlled pro-oncogenic regulatory network and document an important role of HES and TXNIP in the NPM-ALK-driven oncogenesis, with the former protein displaying oncogenic and the latter tumor suppressor properties.


Anaplastic Lymphoma Kinase , Carrier Proteins , Lymphoma, T-Cell , Transcription Factor HES-1 , Anaplastic Lymphoma Kinase/genetics , Carcinogenesis/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Humans , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Oncogenes , Phosphorylation , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
10.
Hum Pathol ; 125: 48-58, 2022 07.
Article En | MEDLINE | ID: mdl-35452696

Normal T cells express high levels of B-cell lymphoma-2 (BCL2) protein, and data regarding BCL2 expression status and its diagnostic utility in T-cell lymphoma are scarce. We evaluated BCL2 expression in a series of mature T-cell lymphoproliferations (TCLs) including indolent and more recently recognized entities (follicular helper T-cell [TFH] lymphomas). Sixty-six neoplastic biopsies (60 patients) representing mature nodal, extranodal, and leukemia T-cell neoplasms were collected from three institutes (2 US and 1 Japan) and were compared with reactive T cells in 8 benign tissues/blood and 9 T cell-rich B-cell proliferations. BCL2 immunostaining was performed and scored based on intensity-weighted H-score (0-300). Next-generation sequencing (NGS; 5 cases), BCL2 gene sequencing, and real-time polymerase chain reaction (PCR; 3 cases) were conducted. Association of H-score with overall survival (using proportional hazards modeling) was assessed in nonleukemic TCLs. Most TCLs showed significantly downregulated median BCL2 H-score (125, range: 18-300) with the exception of T-cell prolymphocytic leukemia and hepatosplenic T-cell lymphoma, both of which showed uniform strong retention of BCL2 as did the 8 reactive tissues (median H-score: 280; p = 0.000). Notably all TFH lymphoma CD4 neoplastic T cells, subcutaneous panniculitis-like T-cell lymphoma, CD8 adipocyte-rimming T cells, and T-cell large lymphocyte leukemia with pathogenic STAT5B and TP53 mutation showed BCL2 downregulation. No BCL2 mutations were observed by NGS or sequencing with decreased BCL2 mRNA transcripts by real-time PCR. BCL2 downregulation is pervasive among many TCLs and unrelated to any mutations. There is utility for BCL2 immunostaining in some challenging situations as discussed in this article.


Leukemia , Lymphoma, B-Cell , Lymphoma, T-Cell, Peripheral , Lymphoma, T-Cell , Proto-Oncogene Proteins c-bcl-2 , Down-Regulation , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/pathology , Phenotype , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
11.
Apoptosis ; 27(5-6): 382-400, 2022 06.
Article En | MEDLINE | ID: mdl-35366141

Lysophosphatidic acid (LPA) is a small phospholipid that acts as an extracellular lipid mediator. It promotes cancer progression by altering a wide array of cellular processes, including apoptosis, survival, angiogenesis, invasion, and migration through binding with its cognate receptors. Intriguingly, our previous study showed that in vitro treatment of LPA induced survival of T lymphoma cells. Hence, the present investigation was designed to investigate the antitumor potential of Ki16425, an antagonist of LPA receptors, against T cell lymphoma. Our in vitro results showed inhibition of LPA-mediated survival and metabolic activity of T lymphoma cells by Ki16425. Further, in vivo experimental findings indicated the tumor retarding potential of Ki16425 against T cell lymphoma through apoptosis induction, glycolysis inhibition, and immunoactivation. The administration of Ki16425 triggered apoptosis by down-regulating the expression of Bcl2 and up-regulating p53, Bax, cleaved caspase-3, and Cyt c expression. Further, Ki16425 suppressed glycolytic activity with concomitantly decreased expression of GLUT3 and MCT1. Moreover, we also noticed an elevated level of NO and iNOS in tumor cells after Ki16425 administration which might also be responsible for apoptosis induction and suppressed glycolysis. Additionally, we observed an increased population of total leukocytes, lymphocytes, and monocytes along with increased thymocytes count and IL-2 and IFN-γ levels. Besides, we observed amelioration of tumor-induced kidney and liver damages by Ki16425. Taken together, this is the first study that demonstrates that LPA receptors could be potential future therapeutic targets for designing promising therapeutic strategies against T cell lymphoma.


Lymphoma, T-Cell , Receptors, Lysophosphatidic Acid , Apoptosis , Glycolysis , Humans , Immunity , Isoxazoles , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lysophospholipids/metabolism , Propionates
12.
Environ Toxicol ; 37(6): 1445-1457, 2022 Jun.
Article En | MEDLINE | ID: mdl-35199915

Nimbolide is reported as one of the potential anticancer candidates of the neem tree (Azadirachta indica A. Juss). The cytotoxic action of nimbolide has been well reported against a wide number of malignancies, including breast, prostate, lung, liver, and cervix cancers. Interestingly, only a few in vivo studies conducted on B cell lymphoma, glioblastoma, pancreatic cancer, and buccal pouch carcinoma have shown the in vivo antitumor efficacy of nimbolide. Therefore, it is highly needed to examine the in vivo antineoplastic activity of nimbolide on a wide variety of cancers to establish nimbolide as a promising anticancer drug. In the present study, we investigated the tumor retarding action of nimbolide in a murine model of T cell lymphoma. We noticed significantly augmented apoptosis in nimbolide- administered tumor-bearing mice, possibly due to down-regulated expression of Bcl2 and up-regulated expression of p53, cleaved caspase-3, Cyt c, and ROS. The nimbolide treatment-induced ROS production by suppressing the expression of antioxidant regulatory enzymes, namely superoxide dismutase and catalase. In addition, nimbolide administration impaired glycolysis and pH homeostasis with concomitant inhibition of crucial glycolysis and pH regulatory molecules such as GLUT3, LDHA, MCT1, and V-ATPase, CAIX and NHE1, respectively. Taken together, the present investigation provides novel insights into molecular mechanisms of nimbolide inhibited T cell lymphoma progression and directs the utility of nimbolide as a potential anticancer therapeutic drug for the treatment of T cell lymphoma.


Antineoplastic Agents , Limonins , Lymphoma, T-Cell , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , Glucose/metabolism , Homeostasis , Hydrogen-Ion Concentration , Limonins/pharmacology , Limonins/therapeutic use , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/metabolism , Male , Mice , Reactive Oxygen Species/metabolism
13.
Int J Mol Sci ; 23(3)2022 Jan 26.
Article En | MEDLINE | ID: mdl-35163302

The discovery of the human histamine H4 receptor (H4R) has contributed to our understanding of the role of histamine in numerous physiological and pathological conditions, including tumor development and progression. The lymph nodes of patients with malignant lymphomas have shown to contain high levels of histamine, however, less is known regarding the expression and function of the H4R in T-cell lymphoma (TCL). In this work we demonstrate the expression of H4R isoforms (mRNA and protein) in three human aggressive TCL (OCI-Ly12, Karpas 299, and HuT78). Histamine and specific H4R agonists (VUF8430 and JNJ28610244) significantly reduced cell viability in a dose-dependent manner (p < 0.05). The combined treatment with the H4R antagonist (JNJ7777120, 10 µM) reversed the effects of the H4R ligands. Importantly, we screened a drug repurposing library of 433 FDA-approved compounds (1 µM) in combination with histamine (10 µM) in Hut78 cells. Histamine produced a favorable antitumor effect with 18 of these compounds, including the histone deacetylase inhibitor panobinostat. Apoptosis, proliferation, and oxidative stress studies confirmed the antitumoral effects of the combination. We conclude that the H4R is expressed in TCL, and it is involved in histamine-mediated responses.


Antineoplastic Agents/pharmacology , Histamine Agonists/pharmacology , Lymphoma, T-Cell/drug therapy , Receptors, Histamine H4/metabolism , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , HEK293 Cells , Histamine/metabolism , Histamine Antagonists/pharmacology , Humans , Lymphoma, T-Cell/metabolism , Oxidative Stress/drug effects
14.
Pharmacol Ther ; 236: 108109, 2022 08.
Article En | MEDLINE | ID: mdl-35007658

T cell lymphomas encompass a diverse group of Non-Hodgkin lymphomas with a wide spectrum of clinical, immunological and pathological manifestations. In the last two decades there has been a progress in our understanding of the cell of origin, genetic abnormalities and their impact on behaviour in T cell lymphomas. Genetic alterations are one of the critical drivers of the pathogenesis of T cell lymphoma. Disease progression has been correlated with multiple genetic abnormalities where malignant clones arise primarily out of the host immune surveillance arsenal. There are many cellular processes involved in disease development, and some of them are T cell signaling, differentiation, epigenetic modifications, and immune regulation. Modulation of these crucial pathways via genetic mutations and chromosomal abnormalities possessing either point or copy number mutations helps tumor cells to develop a niche favourable for their growth via metabolic alterations. Several metabolic pathways especially regulation of redox homeostasis is critical in pathogenesis of lymphoma. Disruption of redox potential and induction of oxidative stress renders malignant cells vulnerable to mitochondrial damage and triggers apoptotic pathways causing cell death. Targeting genetic abnormalities and oxidative stress along with current treatment regime have the potential for improved therapeutics and presents new combination approaches towards selective treatment of T cell lymphomas.


Lymphoma, Non-Hodgkin , Lymphoma, T-Cell , Lymphoma , Humans , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Mutation , Oxidative Stress
15.
Br J Haematol ; 196(1): 31-44, 2022 01.
Article En | MEDLINE | ID: mdl-34169507

Epstein-Barr virus (EBV) is a ubiquitous herpesvirus with rare but severe potential for lymphoproliferative complications. EBV is associated with a variety of presentations of haemophagocytic lymphohistiocytosis (HLH). HLH is a life-threatening hyperinflammatory syndrome that can occur in patients with genetic defects associated with dysregulation of the immune response (familial HLH) or arise in patients with underlying infection or malignancy (non-familial or secondary HLH). EBV can both serve as the incidental trigger of familial HLH or as the driving factor in patients with selective inherited vulnerability (e.g. X-linked lymphoproliferative disease). Alternatively, acute infection can idiosyncratically cause non-neoplastic HLH in patients without inherited predisposition (i.e. secondary HLH), while EBV-associated T/natural killer (NK)-cell lymphoproliferative disorders and lymphomas can cause neoplasia-associated HLH. The present review will discern between EBV-associated familial and non-familial HLH and highlight diagnostic and therapeutic considerations. Non-familial EBV-associated HLH is a major diagnostic dilemma, as it represents a diverse spectrum of disease ranging from highly curable (non-neoplastic EBV-HLH) to indolent but incurable (chronic active EBV) to acutely fatal (systemic EBV-positive T-cell lymphoma of childhood). Increased clinical awareness and understanding of this rare and potentially devastating subset of EBV-related complications is desperately needed to improve survival for patients with neoplasia-associated HLH.


Disease Susceptibility , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/metabolism , Algorithms , Biomarkers , Biopsy , Bone Marrow/pathology , Clinical Decision-Making , Cytokines/metabolism , Disease Management , Genetic Predisposition to Disease , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/therapy , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/metabolism , Mutation , Perforin/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
16.
Exp Hematol ; 106: 3-18, 2022 02.
Article En | MEDLINE | ID: mdl-34879258

T Cells comprise many subtypes of specified lymphocytes, and their differentiation and function take place in different tissues. This cellular diversity is also observed in the multiple ways T-cell transformation gives rise to a variety of T-cell neoplasms. This review covers the main types of T-cell malignancies and their specific characteristics, emphasizing recent advances at the cellular and molecular levels as well as differences and commonalities among them.


Leukemia, T-Cell/pathology , Lymphoma, T-Cell/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , T-Lymphocytes/pathology , Animals , Chromosome Aberrations , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/therapy , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/therapy , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Signal Transduction , T-Lymphocytes/metabolism
17.
J Histochem Cytochem ; 69(10): 645-657, 2021 10.
Article En | MEDLINE | ID: mdl-34617807

Angioimmunoblastic T-cell lymphoma (AITL) is a T-cell lymphoma of follicular helper T-cell origin. Histologically, neoplastic T-cells proliferate to form clusters adjacent to or between arborizing high endothelial venules (HEVs). HEVs in normal lymph nodes express sulfated glycans called peripheral lymph node addressin (PNAd); however, it remains unclear whether PNAd is also expressed on HEVs in AITL. Furthermore, although it is widely accepted that HEVs are conspicuous in AITL due to their proliferation, quantitative histological support for this concept is lacking. To investigate these issues, we employed monoclonal antibodies recognizing PNAd, namely, MECA-79, HECA-452, and 297-11A, and performed quantitative immunohistochemical analysis of HEVs in 36 AITL-affected and 67 normal lymph nodes. Staining with all three antibodies confirmed that AITL HEVs express PNAd. Moreover, AITL HEVs were bound calcium-dependently by L-selectin-IgM fusion proteins, indicating that they function in the recruitment of L-selectin-expressing lymphocytes. Unexpectedly, HEV distribution density was not increased but rather decreased in AITL compared with normal lymph nodes, but HEV cross-sectional area in AITL was significantly greater than that seen in normal lymph nodes. Overall, these results indicate that the prominence of AITL HEVs is likely due to increased cross-sectional area rather than increased distribution density.


Lymphoma, T-Cell/pathology , Venules/cytology , Cell Line , Humans , Lymphoma, T-Cell/metabolism , Venules/metabolism
18.
Front Immunol ; 12: 737849, 2021.
Article En | MEDLINE | ID: mdl-34557199

Capecitabine (CAP) is now widely used in the comprehensive treatment of digestive system tumors. Some clinical observations have shown that CAP may have immunosuppressive effects, but there is still a lack of clear experimental verification. In this study, different doses of CAP were administered to normal mice by gavage. Our results confirmed that CAP did not cause myelosuppression in bone marrow tissue; CAP selectively reduced the proportion of T cells and the concentration of related pro-inflammatory cytokines, while it increased the concentration of anti-inflammatory cytokines. Thymidylate phosphorylase (TP) is the key enzyme for the transformation of CAP in vivo; this study confirmed that T cells express TP, but the bone marrow tissue lacks TP expression, which explains the selectivity in pharmacodynamic effects of CAP. In addition, it was confirmed that CAP can induce T cell apoptosis in vivo and in vitro. In vitro experiments showed that CAP-induced T cell apoptosis was related to TP expression, endoplasmic reticulum stress (ERS) induction, reactive oxygen species (ROS) production, and mitochondria-mediated apoptosis activation. Therefore, this study confirmed that the differential expression of TP in cells and tissues explains why CAP avoids the toxic effects of myelosuppression while inducing T cell apoptosis to exert the immunosuppressive effect. Therefore, CAP may become an immunosuppressive agent with a simultaneous anti-cancer effect, which is worthy of further studies.


Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Capecitabine/pharmacology , Immunosuppressive Agents/pharmacology , Lymphoma, T-Cell/drug therapy , T-Lymphocytes/drug effects , Animals , Cell Line, Tumor , Cytokines/metabolism , Endoplasmic Reticulum Stress/drug effects , Humans , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Male , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thymidine Phosphorylase/metabolism
19.
Exp Cell Res ; 407(1): 112790, 2021 10 01.
Article En | MEDLINE | ID: mdl-34418460

BACKGROUND: Natural killer/T cell lymphoma (NKTCL) is a distinct subtype of Non-Hodgkin's lymphoma with highly aggressive clinical behavior. We aim to investigate the function of Latent transforming growth factor ß binding protein 1 (LTBP1) and transforming growth factor beta1 (TGF-ß1) and complex molecular pathogenesis of this disease. METHODS: NKTCL patients and reactive lymph nodes patients were recruited in this study. The expression of LTBP1 and TGF-ß1 was examined using qRT-PCR, Western blot, IHC and ELISA analyses in biopsied tissues and serum from participants and NKTCL cell lines. Cell proliferation was determined using CFSE. Cell cycle and apoptosis were evaluated using flow cytometric analyses. The expression of Ki-67, CDK4 and cyclinD1 proteins was measured using Western blot analyses. The roles of LTBP-1/TGF-ß1 in EMT program were determined by measuring E-cadherin, N-cadherin and Vimentin using Western blot analyses. The effects of LTBP-1 and TGF-ß1 on tumor progression in vivo were determined by animal experiments. RESULTS: LTBP-1 and TGF-ß1 levels were elevated in NKTCL tissues and serum. The expression of LTBP-1 was positively correlated with the expression of TGF-ß1 in NKTCL tissues. LTBP-1 was overexpressed in NKTCL cells. Knockdown of LTBP-1 suppressed cell proliferation and cell cycle progression, induced cell apoptosis, and suppressed EMT program in NKTCL cells. These effects of LTBP-1 knockdown were attenuated after TGF-ß1 stimulation. Knockdown of LTBP-1 inhibited NKTCL tumor weight and volume in vivo. Also, stimulation of TGF-ß1 attenuated the suppressive effects on tumor growth from sh-LTBP-1. Silencing of LTBP-1 lowered cellular TGF-ß1, phosphorylated-Smad2, phosphorlyatd-Smad3, and phosphorylated-p38 and the suppressive effects were reversed after stimulation of TGF-ß1. CONCLUSION: Our findings suggested that inhibition of LTBP-1/TGF-ß1 suppressed the malignant phenotypes of NKTCL cells and tumor growth via inactivating the canonical TGF-ß/Smad signaling and p38MAPK signaling.


Killer Cells, Natural/metabolism , Latent TGF-beta Binding Proteins/metabolism , Lymphoma, T-Cell/metabolism , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Humans , Signal Transduction/drug effects , Vimentin/metabolism
...