93 results on '"Martinez Climent JA"'
Search Results
2. Identification and molecular characterization of CALM/AF10fusion products in T cell acute lymphoblastic leukemia and acute myeloid leukemia
- Author
-
Carlson, KM, Vignon, C, Bohlander, S, Martinez-Climent, JA, Beau, M M Le, and Rowley, JD
- Published
- 2000
- Full Text
- View/download PDF
3. Deep MRD profiling defines outcome and unveils different modes of treatment resistance in standard and high risk myeloma
- Author
-
Goicoechea I, Puig N, Cedena MT, Burgos L, Cordón L, Vidriales MB, Flores-Montero J, Gutierrez NC, Calasanz MJ, Martin Ramos ML, Lara-Astiaso D, Vilas-Zornoza A, Alignani D, Rodriguez I, Sarvide S, Alameda D, Garcés JJG, Rodriguez S, Fresquet V, Celay J, Garcia-Sanz R, Martinez-Lopez J, Oriol A, Rios R, Martin-Sanchez J, Martinez-Martinez R, Sarra J, Hernandez MT, de la Rubia J, Krsnik I, Moraleda JM, Palomera L, Bargay J, Martinez-Climent JA, Orfao A, Rosiñol L, Mateos MV, Lahuerta JJ, Blade J, San Miguel J, and Paiva B
- Subjects
body regions ,hemic and lymphatic diseases - Abstract
Patients with multiple myeloma (MM) carrying high-risk cytogenetic abnormalities (CA) have inferior outcome despite achieving similar complete response (CR) rates when compared to cases with standard-risk CA. This questions the legitimacy of CR as treatment endpoint for high-risk MM, and represents a biological conundrum regarding the nature of tumor reservoirs persisting after therapy in patients with standard- and high-risk CA. Here, we used next-generation flow (NGF) to evaluate measurable residual disease (MRD) in MM patients with standard- (N=300) vs high-risk CA (N=90) enrolled in the PETHEMA/GEM2012MENOS65 trial (NCT01916252), and to identify mechanisms determining MRD resistance in both patient subgroups (N=40). The 36-month progression-free and overall survival rates were higher than 90% in patients with undetectable MRD, with no significant differences (P=0.202) between cases having standard- vs high-risk CA. Persistent MRD resulted in median progression-free survival of approximately three and two years in patients with standard- and high-risk CA, respectively (P
- Published
- 2020
4. Molecular cytogenetics of childhood hematological malignancies
- Author
-
Martinez-Climent, JA
- Published
- 1997
- Full Text
- View/download PDF
5. Cytogenetic response induced by interferon alpha in the myeloproliferative disorder with eosinophilia, T cell lymphoma and the chromosomal translocation t(8;13)(p11;q12)
- Author
-
Martinez-Climent, JA, Vizcarra, E, Benet, I, Marugan, I, Terol, MJ, Solano, C, Arbona, C, Tormo, M, Comes, AM, and García-Conde, J
- Published
- 1998
- Full Text
- View/download PDF
6. Intrinsic resistance to PIM kinase inhibition in AML through p38a-mediated feedback activation of mTOR signaling
- Author
-
Brunen, D, Garcia-Barchino, MJ, Malani, D, Jagalur Basheer, Noorjahan, Lieftink, C, Beijersbergen, RL, Murumagi, A, Porkka, K, Wolf, M, Zwaan, C.M., Fornerod, M.W.J., Kallioniemi, O, Martinez-Climent, JA, Bernards, R, Brunen, D, Garcia-Barchino, MJ, Malani, D, Jagalur Basheer, Noorjahan, Lieftink, C, Beijersbergen, RL, Murumagi, A, Porkka, K, Wolf, M, Zwaan, C.M., Fornerod, M.W.J., Kallioniemi, O, Martinez-Climent, JA, and Bernards, R
- Published
- 2016
7. MALT1 is deregulated by both chromosomal translocation and amplification in B-cell non-Hodgkin lymphoma
- Author
-
UCL, Sanchez-Izquierdo, D, Buchonnet, G, Siebert, R, Gascoyne, RD, Climent, J, Karran, L, Marin, M., Blesa, D, Horsman, D, Rosenwald, A, Staudt, LM, Albertson, DG, Du, MQ, Ye, HT, Marynen, Peter, Garcia-Conde, J, Pinkel, D, Dyer, MJS, Martinez-Climent, JA, UCL, Sanchez-Izquierdo, D, Buchonnet, G, Siebert, R, Gascoyne, RD, Climent, J, Karran, L, Marin, M., Blesa, D, Horsman, D, Rosenwald, A, Staudt, LM, Albertson, DG, Du, MQ, Ye, HT, Marynen, Peter, Garcia-Conde, J, Pinkel, D, Dyer, MJS, and Martinez-Climent, JA
- Abstract
The MALT1 gene was identified through its involvement in t(11;18)(q21;q21), seen in 30% of cases of mucosa-associated, lymphoid tissue (MALT) lymphoma. Here, we show that deregulated MALT1 expression may occur in B-cell non-Hodgkin lymphoma (B-NHL) of various histologic subtypes either through translocation to the immunoglobulin heavy chain (IGH) locus or by genomic amplification, First, 2 cases, one case of MALT lymphoma and another of aggressive marginal zone lymphoma (MZL) with t(14;1 8)(q32;q21), cytogenetically identical to the translocation involving BCL2 Were shown by fluorescence in situ hybridization (FISH) to involve MALT1, which lies about 5 Mb centromeric of BCL2. Molecular cloning of both by long-distance inverse polymerase chain reaction showed breakpoints lying 1 to 2 kilobase (kb) centromeric of the first 5' MALT1 exon; both cases showed MALT1 overexpression at either RNA or protein levels. Second, we examined the structure and gene expression profile of genomic amplifications involving 18q21 in a panel of 40 B-NHL cell lines using comparative genomic hybridization to microarrays (array CGH) and gene expression profiling techniques. Using array CGH, 2 peaks of genomic amplification were observed one centered around BCL2 and the other around MALT1. Of the 3 cell lines with MALT1 Amplification, 2 showed MALT1 overexpression as assessed by gene profiling, quantitative reverse transcription-polymerase chain re action (QRT-PCR), and Western blotting. To determine if comparable events occurred in primary MALT and splenic MZL tumors, 40 cases were analyzed by FISH or QRT-PCR; genomic amplification and MALT1 overexpression were seen in 2 cases. Together, these data implicate MALT1 as a dominant oncogene that may play a role in the pathogenesis of B-NHL. (C) 2003 by The American Society of Hematology.
- Published
- 2003
8. The alternative RelB NF-κB subunit is a novel critical player in diffuse large B-cell lymphoma
- Author
-
Eluard, B, Nuan-Aliman, S, Faumont, N, Collares, D, Bordereaux, D, Montagne, A, Martins, I, Cagnard, N, Caly, M, Taoui, O, Lordello, L, Lehmann-Che, J, Tesson, B, Martinez-Climent, JA, Copie-Bergman, C, Haioun, C, Tilly, H, Bonsang, B, Vincent-Salomon, A, Jais, JP, Jardin, F, Leroy, K, Maiuri, MC, Kroemer, G, Molina, TJ, Feuillard, J, and Baud, V
- Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most frequent lymphoid malignancy affecting adults. NF-κB transcription factor family is activated by two main pathways, the canonical and the alternative NF-κB activation pathways with different functions. The alternative NF-κB pathway leads to the activation of the transcriptionally active RelB NF-κB subunit. Alternative NF-kB activation status and its role in DLBCL pathogenesis remain undefined. Here, we reveal a frequent activation of RelB in a large cohort of DLBCL patients and cell lines, independently of their ABC or GCB subtypes. RelB activity defines a new subset of DLBCL patients with a peculiar gene expression profile and mutational pattern. Importantly, RelB activation does not correlate with the MCD genetic subtype, enriched for ABC tumors carrying MYD88L265Pand CD79B mutations that cooperatively activate canonical NF-κB, thus indicating that current genetic tools to evaluate NF-κB activity in DLBCL do not provide information on the alternative NF-κB activation. Further, the newly defined RelB-positive subgroup of DLBCL patients exhibits a dismal outcome following immunochemotherapy. Functional studies revealed that RelB confers DLBCL cell resistance to DNA-damage induced apoptosis in response to doxorubicin, a genotoxic agent used in front-line treatment for DLBCL. We also show that RelB positivity is associated with high expression of cIAP2. Altogether, RelB activation can be used to refine the prognostic stratification of DLBCL and may contribute to subvert the therapeutic DNA damage response in a segment of DLBCL patients.
- Published
- 2021
- Full Text
- View/download PDF
9. Additional cytogenetic changes do not influence the outcome of patients with newly diagnosed acute promyelocytic leukemia treated with an ATRA plus anthracycline-based protocol. A report from the Spanish group PETHEMA
- Author
-
Hernandez, Jm, Martin, G., Gutierrez, Nc, Cervera, J., Ferro, Mt, MJ Calasanz, Martinez-Climent, Ja, Luno, E., Tormo, M., Rayon, C., Diaz-Mediavilla, J., Gonzalez, M., Gonzalez-San Miguel, Jd, Perez-Equiza, K., Rivas, C., Esteve, J., Alvarez, Md, Odriozola, J., Ribera, Jm, Sanz, Ma, and Pethema, Cooperative Grp
10. Abnormalities on 1q are associated with a bad prognosis in sporadic Burkitt lymphoma. A cytogenetic and comparative genomic hybridization study
- Author
-
Hernandez, Jm, Garcia, Jl, Lopez-Capitan, C., MJ Calasanz, Martinez-Climent, Ja, Gutierrez, Nc, Flores, T., Odero, Md, Piris, Ma, and San Miguel, Jf
11. LMO2 expression reflects the different stages of blast maturation and genetic features in B-cell acute lymphoblastic leukemia and predicts clinical outcome
- Author
-
Maria Angela Aznar, Raquel Malumbres, Jose A. Martinez-Climent, Izidore S. Lossos, Erlend B. Smeland, M. Jose Calasanz, Jose Roman-Gomez, Giovanna Giuseppina Altobelli, Xabier Agirre, Eloy F. Robles, Miriam Bobadilla, Vanesa Martín-Palanco, Felipe Prosper, Vicente Fresquet, Malumbres, R, Fresquet, V, Roman Gomez, J, Bobadilla, M, Robles, Ef, Altobelli, GIOVANNA GIUSEPPINA, Calasanz, Mj, Smeland, Eb, Aznar, Ma, Agirre, X, Martin Palanco, V, Prosper, F, Lossos, I, and Martinez Climent, Ja
- Subjects
LMO2 ,Adult ,Adolescent ,B-Lymphocyte Subsets ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Immunophenotyping ,Young Adult ,hemic and lymphatic diseases ,Cell Line, Tumor ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Proto-Oncogene Proteins ,Metalloproteins ,Humans ,Clinical significance ,acute leukemia ,Lymphopoiesis ,Stage (cooking) ,Child ,Survival rate ,Adaptor Proteins, Signal Transducing ,Aged ,Aged, 80 and over ,B lymphocyte ,Gene Expression Regulation, Leukemic ,Age Factors ,Germinal center ,Infant ,Karyotype ,Cell Differentiation ,Hematology ,Original Articles ,LIM Domain Proteins ,Middle Aged ,Prognosis ,Survival Analysis ,DNA-Binding Proteins ,Haematopoiesis ,Treatment Outcome ,Child, Preschool ,Karyotyping ,Immunology ,gene expression ,Cancer research - Abstract
Background LMO2 is highly expressed at the most immature stages of lymphopoiesis. In T-lymphocytes, aberrant LMO2 expression beyond those stages leads to T-cell acute lymphoblastic leukemia, while in B cells LMO2 is also expressed in germinal center lymphocytes and diffuse large B-cell lymphomas, where it predicts better clinical outcome. The implication of LMO2 in B-cell acute lymphoblastic leukemia must still be explored. Design and Methods We measured LMO2 expression by real time RT-PCR in 247 acute lymphoblastic leukemia patient samples with cytogenetic data (144 of them also with survival and immunophenotypical data) and in normal hematopoietic and lymphoid cells. Results B-cell acute lymphoblastic leukemia cases expressed variable levels of LMO2 depending on immunophenotypical and cytogenetic features. Thus, the most immature subtype, pro-B cells, displayed three-fold higher LMO2 expression than pre-B cells, common-CD10+ or mature subtypes. Additionally, cases with TEL-AML1 or MLL rearrangements exhibited two-fold higher LMO2 expression compared to cases with BCR-ABL rearrangements or hyperdyploid karyotype. Clinically, high LMO2 expression correlated with better overall survival in adult patients (5-year survival rate 64.8% (42.5%–87.1%) vs . 25.8% (10.9%–40.7%), P = 0.001) and constituted a favorable independent prognostic factor in B-ALL with normal karyotype: 5-year survival rate 80.3% (66.4%–94.2%) vs. 63.0% (46.1%–79.9%) ( P = 0.043). Conclusions Our data indicate that LMO2 expression depends on the molecular features and the differentiation stage of B-cell acute lymphoblastic leukemia cells. Furthermore, assessment of LMO2 expression in adult patients with a normal karyotype, a group which lacks molecular prognostic factors, could be of clinical relevance.
- Published
- 2011
12. Metabolization of microbial postbiotic pentanoate drives anti-cancer CAR T cells.
- Author
-
Staudt S, Nikolka F, Perl M, Franz J, Leblay N, Yuan XK, Larrayoz M, Lozano T, Warmuth L, Fante MA, Skorpskaite A, Fei T, Bromberg M, San Martin-Uriz P, Rodriguez-Madoz JR, Ziegler-Martin K, Adil-Gholam N, Benz P, Tran Huu P, Freitag F, Riester Z, Stein-Thoeringer C, Schmitt M, Kleigrewe K, Weber J, Mangold K, Ho P, Einsele H, Prosper F, Ellmeier W, Busch D, Visekruna A, Slingerland J, Shouval R, Hiller K, Lasarte JJ, Martinez-Climent JA, Pausch P, Neri P, van den Brink M, Poeck H, Hudecek M, and Luu M
- Abstract
The microbiome is a complex host factor and key determinant of the outcome of antibody-based and cellular immunotherapy. Its postbiotics are a blend of soluble commensal byproducts that are released into the host environment and have been associated with the regulation of immune homeostasis, particularly through impacts on epigenetics and cell signaling. In this study, we show that the postbiotic pentanoate is metabolized to citrate within the TCA cycle via both the acetyl- and succinyl-CoA entry points, a feature uniquely enabled by the chemical structure of the C5 aliphatic chain. We identified ATP-citrate lyase as the crucial factor that redirects pentanoate-derived citrate from the succinyl-CoA route to the nucleus, thereby linking metabolic output and histone acetylation. This epigenetic-metabolic crosstalk mitigated T cell exhaustion and promoted naive-like differentiation in pentanoate-programmed chimeric antigen receptor (CAR) T cells. The predictive and therapeutic potential of pentanoate was corroborated in two independent patient cohorts and three syngeneic models of CAR T adoptive therapy. Our data demonstrate that postbiotics are integrated into mitochondrial metabolism and subsequently incorporated as epigenetic imprints. This bridge between microbial and mammalian interspecies communication can ultimately impact T cell differentiation and efficacy., Competing Interests: Competing interests ML, MH and AV are listed as inventors on patent application WO2021/058811A1. MH is listed as an inventor on patent applications and granted patents related to CAR-T technologies that have been filed by the Fred Hutchinson Cancer Research Center, Seattle, WA and by the University of Würzburg, Würzburg, Germany. MH is a co-founder and equity owner of T-CURX GmbH, Würzburg, Germany. MH received honoraria from Celgene/BMS, Janssen, Kite/Gilead. MvdB has received research support and stock options from Seres Therapeutics and stock options from Notch Therapeutics and Pluto Therapeutics; he has received royalties from Wolters Kluwer; has consulted, received honorarium from or participated in advisory boards for Seres Therapeutics, Rheos Medicines, Ceramedix, Pluto Therapeutics, Thymofox, Garuda, Novartis (Spouse), Synthekine (Spouse), Beigene (Spouse), Kite (Spouse); he has IP Licensing with Seres Therapeutics and Juno Therapeutics; and holds a fiduciary role on the Foundation Board of DKMS (a nonprofit organization). PN received honoraria from BMS, Janssen, Sanofi and Pfizer as a consultant/advisory board member. MAF received honoraria from Novartis and Sanofi and travel grants from Sanofi. HP is a consultant for Gilead, Abbvie, Pfizer, Novartis, Servier, and Bristol Myers-Squibb. J.A.M.-C. has received research funding from Roche-Genentech, Bristol Myers Squibb-Celgene, Janssen, Regeneron, Priothera Pharmaceuticals, Palleon Pharmaceuticals, AstraZeneca, and K36 Therapeutics, and is founder and equity owner of MIMO Biosciences. The remaining authors declare no financial conflict of interest.
- Published
- 2025
- Full Text
- View/download PDF
13. Not so natural, not so killers.
- Author
-
Paiva B and Martinez-Climent JA
- Published
- 2024
- Full Text
- View/download PDF
14. Round Table Discussion on Optimal Clinical Trial Design in Precursor Multiple Myeloma.
- Author
-
Ghobrial IM, Gormley N, Kumar SK, Mateos MV, Bergsagel PL, Chesi M, Dhodapkar MV, Dispenzieri A, Fonseca R, Getz G, Kastritis E, Kristinsson SY, Martinez-Climent JA, Manier S, Marinac CR, Maura F, Morgan GJ, Davies FE, Nadeem O, Nuvolone M, Paiva B, O'Donnell E, Prosper F, Shah UA, Sklavenitis-Pistofidis R, Sperling AS, Vassiliou GS, Munshi NC, Castle PE, Anderson KC, and San Miguel JF
- Subjects
- Humans, Research Design, Quality of Life, Multiple Myeloma therapy, Multiple Myeloma drug therapy, Clinical Trials as Topic methods
- Abstract
Summary: While the current approach to precursor hematologic conditions is to "watch and wait," this may change with the development of therapies that are safe and extend survival or delay the onset of symptomatic disease. The goal of future therapies in precursor hematologic conditions is to improve survival and prevent or delay the development of symptomatic disease while maximizing safety. Clinical trial considerations in this field include identifying an appropriate at-risk population, safety assessments, dose selection, primary and secondary trial endpoints including surrogate endpoints, control arms, and quality-of-life metrics, all of which may enable more precise benefit-risk assessment., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
15. Large T cell clones expressing immune checkpoints increase during multiple myeloma evolution and predict treatment resistance.
- Author
-
Botta C, Perez C, Larrayoz M, Puig N, Cedena MT, Termini R, Goicoechea I, Rodriguez S, Zabaleta A, Lopez A, Sarvide S, Blanco L, Papetti DM, Nobile MS, Besozzi D, Gentile M, Correale P, Siragusa S, Oriol A, González-Garcia ME, Sureda A, de Arriba F, Rios Tamayo R, Moraleda JM, Gironella M, Hernandez MT, Bargay J, Palomera L, Pérez-Montaña A, Goldschmidt H, Avet-Loiseau H, Roccaro A, Orfao A, Martinez-Lopez J, Rosiñol L, Lahuerta JJ, Blade J, Mateos MV, San-Miguel JF, Martinez Climent JA, and Paiva B
- Subjects
- Adult, Humans, Animals, Mice, T-Lymphocytes, Programmed Cell Death 1 Receptor genetics, Lenalidomide, Clone Cells, Multiple Myeloma drug therapy, Multiple Myeloma genetics
- Abstract
Tumor recognition by T cells is essential for antitumor immunity. A comprehensive characterization of T cell diversity may be key to understanding the success of immunomodulatory drugs and failure of PD-1 blockade in tumors such as multiple myeloma (MM). Here, we use single-cell RNA and T cell receptor sequencing to characterize bone marrow T cells from healthy adults (n = 4) and patients with precursor (n = 8) and full-blown MM (n = 10). Large T cell clones from patients with MM expressed multiple immune checkpoints, suggesting a potentially dysfunctional phenotype. Dual targeting of PD-1 + LAG3 or PD-1 + TIGIT partially restored their function in mice with MM. We identify phenotypic hallmarks of large intratumoral T cell clones, and demonstrate that the CD27
- and CD27+ T cell ratio, measured by flow cytometry, may serve as a surrogate of clonal T cell expansions and an independent prognostic factor in 543 patients with MM treated with lenalidomide-based treatment combinations., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
16. Targeting the immune microenvironment in Waldenström macroglobulinemia via halting the CD40/CD40-ligand axis.
- Author
-
Sacco A, Desantis V, Celay J, Giustini V, Rigali F, Savino FD, Cea M, Soncini D, Cagnetta A, Solimando AG, D'Aliberti D, Spinelli S, Ramazzotti D, Almici C, Todoerti K, Neri A, Anastasia A, Tucci A, Motta M, Chiarini M, Kawano Y, Martinez-Climent JA, Piazza R, and Roccaro AM
- Subjects
- Humans, Animals, Mice, CD40 Ligand genetics, Phosphatidylinositol 3-Kinases, Ligands, Signal Transduction, Tumor Microenvironment, Waldenstrom Macroglobulinemia pathology, Lymphoma, B-Cell complications
- Abstract
Recent investigations have improved our understanding of the molecular aberrations supporting Waldenström macroglobulinemia (WM) biology; however, whether the immune microenvironment contributes to WM pathogenesis remains unanswered. First, we showed how a transgenic murine model of human-like lymphoplasmacytic lymphoma/WM exhibits an increased number of regulatory T cells (Tregs) relative to control mice. These findings were translated into the WM clinical setting, in which the transcriptomic profiling of Tregs derived from patients with WM unveiled a peculiar WM-devoted messenger RNA signature, with significant enrichment for genes related to nuclear factor κB-mediated tumor necrosis factor α signaling, MAPK, and PI3K/AKT, which was paralleled by a different Treg functional phenotype. We demonstrated significantly higher Treg induction, expansion, and proliferation triggered by WM cells, compared with their normal cellular counterpart; with a more profound effect within the context of CXCR4C1013G-mutated WM cells. By investigating the B-cell-to-T-cell cross talk at single-cell level, we identified the CD40/CD40-ligand as a potentially relevant axis that supports WM cell-Tregs interaction. Our findings demonstrate the existence of a Treg-mediated immunosuppressive phenotype in WM, which can be therapeutically reversed by blocking the CD40L/CD40 axis to inhibit WM cell growth., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
17. Preclinical models for prediction of immunotherapy outcomes and immune evasion mechanisms in genetically heterogeneous multiple myeloma.
- Author
-
Larrayoz M, Garcia-Barchino MJ, Celay J, Etxebeste A, Jimenez M, Perez C, Ordoñez R, Cobaleda C, Botta C, Fresquet V, Roa S, Goicoechea I, Maia C, Lasaga M, Chesi M, Bergsagel PL, Larrayoz MJ, Calasanz MJ, Campos-Sanchez E, Martinez-Cano J, Panizo C, Rodriguez-Otero P, Vicent S, Roncador G, Gonzalez P, Takahashi S, Katz SG, Walensky LD, Ruppert SM, Lasater EA, Amann M, Lozano T, Llopiz D, Sarobe P, Lasarte JJ, Planell N, Gomez-Cabrero D, Kudryashova O, Kurilovich A, Revuelta MV, Cerchietti L, Agirre X, San Miguel J, Paiva B, Prosper F, and Martinez-Climent JA
- Subjects
- Mice, Animals, CD8-Positive T-Lymphocytes, Immune Evasion, T-Lymphocytes, Regulatory, Immunotherapy adverse effects, Tumor Microenvironment genetics, Multiple Myeloma therapy, Multiple Myeloma drug therapy
- Abstract
The historical lack of preclinical models reflecting the genetic heterogeneity of multiple myeloma (MM) hampers the advance of therapeutic discoveries. To circumvent this limitation, we screened mice engineered to carry eight MM lesions (NF-κB, KRAS, MYC, TP53, BCL2, cyclin D1, MMSET/NSD2 and c-MAF) combinatorially activated in B lymphocytes following T cell-driven immunization. Fifteen genetically diverse models developed bone marrow (BM) tumors fulfilling MM pathogenesis. Integrative analyses of ∼500 mice and ∼1,000 patients revealed a common MAPK-MYC genetic pathway that accelerated time to progression from precursor states across genetically heterogeneous MM. MYC-dependent time to progression conditioned immune evasion mechanisms that remodeled the BM microenvironment differently. Rapid MYC-driven progressors exhibited a high number of activated/exhausted CD8
+ T cells with reduced immunosuppressive regulatory T (Treg ) cells, while late MYC acquisition in slow progressors was associated with lower CD8+ T cell infiltration and more abundant Treg cells. Single-cell transcriptomics and functional assays defined a high ratio of CD8+ T cells versus Treg cells as a predictor of response to immune checkpoint blockade (ICB). In clinical series, high CD8+ T/Treg cell ratios underlie early progression in untreated smoldering MM, and correlated with early relapse in newly diagnosed patients with MM under Len/Dex therapy. In ICB-refractory MM models, increasing CD8+ T cell cytotoxicity or depleting Treg cells reversed immunotherapy resistance and yielded prolonged MM control. Our experimental models enable the correlation of MM genetic and immunological traits with preclinical therapy responses, which may inform the next-generation immunotherapy trials., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
18. Venetoclax improves CD20 immunotherapy in a mouse model of MYC/BCL2 double-expressor diffuse large B-cell lymphoma.
- Author
-
Melchor J, Garcia-Lacarte M, Grijalba SC, Arnaiz-Leché A, Pascual M, Panizo C, Blanco O, Segura V, Novo FJ, Valero JG, Pérez-Galán P, Martinez-Climent JA, and Roa S
- Subjects
- Animals, Mice, Disease Models, Animal, Proto-Oncogene Proteins c-bcl-2, Tumor Microenvironment, Proto-Oncogene Proteins c-myc, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Immunotherapy methods, Lymphoma, Large B-Cell, Diffuse drug therapy
- Abstract
Background: Approximately one-third of diffuse large B cell lymphoma (DLBCL) patients exhibit co-expression of MYC and BCL2 (double-expressor lymphoma, DEL) and have a dismal prognosis. Targeted inhibition of the anti-apoptotic protein BCL2 with venetoclax (ABT-199) has been approved in multiple B-cell malignancies and is currently being investigated in clinical trials for DLBCL. Whether BCL2 anti-apoptotic function represents a multifaceted vulnerability for DEL-DLBCL, affecting both lymphoma B cells and T cells within the tumor microenvironment, remains to be elucidated., Methods: Here, we present novel genetically engineered mice that preclinically recapitulate DEL-DLBCL lymphomagenesis, and evaluate their sensitivity ex vivo and in vivo to the promising combination of venetoclax with anti-CD20-based standard immunotherapy., Results: Venetoclax treatment demonstrated specific killing of MYC
+ /BCL2+ lymphoma cells by licensing their intrinsically primed apoptosis, and showed previously unrecognized immunomodulatory activity by specifically enriching antigen-activated effector CD8 T cells infiltrating the tumors. Whereas DEL-DLBCL mice were refractory to venetoclax alone, inhibition of BCL2 significantly extended overall survival of mice that were simultaneously treated with a murine surrogate for anti-CD20 rituximab., Conclusions: These results suggest that the combination of anti-CD20-based immunotherapy and BCL2 inhibition leads to cooperative immunomodulatory effects and improved preclinical responses, which may offer promising therapeutic opportunities for DEL-DLBCL patients., Competing Interests: Competing interests: SR has received research funding from Roche/Genentech (imCORE) and Gilead; JAM-C has received research funding from Roche/Genentech (imCORE) and Janssen Pharmaceuticals; CP has acted as a consultant for Roche, Janssen, Celgene/BMS and Kyowa Kirin. The remaining authors declare no conflicting financial interests., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2023
- Full Text
- View/download PDF
19. Preneoplastic somatic mutations including MYD88 L265P in lymphoplasmacytic lymphoma.
- Author
-
Rodriguez S, Celay J, Goicoechea I, Jimenez C, Botta C, Garcia-Barchino MJ, Garces JJ, Larrayoz M, Santos S, Alignani D, Vilas-Zornoza A, Perez C, Garate S, Sarvide S, Lopez A, Reinhardt HC, Carrasco YR, Sanchez-Garcia I, Larrayoz MJ, Calasanz MJ, Panizo C, Prosper F, Lamo-Espinosa JM, Motta M, Tucci A, Sacco A, Gentile M, Duarte S, Vitoria H, Geraldes C, Paiva A, Puig N, Garcia-Sanz R, Roccaro AM, Fuerte G, San Miguel JF, Martinez-Climent JA, and Paiva B
- Subjects
- Aged, Animals, Humans, Mice, Mutation, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 metabolism, Lymphoma, Lymphoma, B-Cell metabolism, Waldenstrom Macroglobulinemia diagnosis, Waldenstrom Macroglobulinemia genetics, Waldenstrom Macroglobulinemia pathology
- Abstract
Normal cell counterparts of solid and myeloid tumors accumulate mutations years before disease onset; whether this occurs in B lymphocytes before lymphoma remains uncertain. We sequenced multiple stages of the B lineage in elderly individuals and patients with lymphoplasmacytic lymphoma, a singular disease for studying lymphomagenesis because of the high prevalence of mutated MYD88 . We observed similar accumulation of random mutations in B lineages from both cohorts and unexpectedly found MYD88
L265P in normal precursor and mature B lymphocytes from patients with lymphoma. We uncovered genetic and transcriptional pathways driving malignant transformation and leveraged these to model lymphoplasmacytic lymphoma in mice, based on mutated MYD88 in B cell precursors and BCL2 overexpression. Thus, MYD88L265P is a preneoplastic event, which challenges the current understanding of lymphomagenesis and may have implications for early detection of B cell lymphomas.- Published
- 2022
- Full Text
- View/download PDF
20. The alternative RelB NF-κB subunit is a novel critical player in diffuse large B-cell lymphoma.
- Author
-
Eluard B, Nuan-Aliman S, Faumont N, Collares D, Bordereaux D, Montagne A, Martins I, Cagnard N, Caly M, Taoui O, Lordello L, Lehmann-Che J, Tesson B, Martinez-Climent JA, Copie-Bergman C, Haioun C, Tilly H, Bonsang B, Vincent-Salomon A, Jais JP, Jardin F, Leroy K, Maiuri MC, Kroemer G, Molina TJ, Feuillard J, and Baud V
- Subjects
- Apoptosis, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Humans, Lymphoma, Large B-Cell, Diffuse genetics, NF-kappa B genetics, Transcription Factor RelB genetics, Transcriptional Activation, Lymphoma, Large B-Cell, Diffuse metabolism, NF-kappa B metabolism, Transcription Factor RelB metabolism
- Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most frequent lymphoid malignancy affecting adults. The NF-κB transcription factor family is activated by 2 main pathways, the canonical and the alternative NF-κB activation pathway, with different functions. The alternative NF-κB pathway leads to activation of the transcriptionally active RelB NF-κB subunit. Alternative NF-κB activation status and its role in DLBCL pathogenesis remain undefined. Here, we reveal a frequent activation of RelB in a large cohort of DLBCL patients and cell lines, independently of their activated B-cell-like or germinal center B-cell-like subtype. RelB activity defines a new subset of patients with DLBCL and a peculiar gene expression profile and mutational pattern. Importantly, RelB activation does not correlate with the MCD genetic subtype, enriched for activated B-cell-like tumors carrying MYD88L265P and CD79B mutations that cooperatively activate canonical NF-κB, thus indicating that current genetic tools to evaluate NF-κB activity in DLBCL do not provide information on the alternative NF-κB activation. Furthermore, the newly defined RelB-positive subgroup of patients with DLBCL exhibits a dismal outcome after immunochemotherapy. Functional studies revealed that RelB confers DLBCL cell resistance to DNA damage-induced apoptosis in response to doxorubicin, a genotoxic agent used in the front-line treatment of DLBCL. We also show that RelB positivity is associated with high expression of cellular inhibitor of apoptosis protein 2 (cIAP2). Altogether, RelB activation can be used to refine the prognostic stratification of DLBCL and may contribute to subvert the therapeutic DNA damage response in a segment of patients with DLBCL., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
21. Endogenous Retroelement Activation by Epigenetic Therapy Reverses the Warburg Effect and Elicits Mitochondrial-Mediated Cancer Cell Death.
- Author
-
Fresquet V, Garcia-Barchino MJ, Larrayoz M, Celay J, Vicente C, Fernandez-Galilea M, Larrayoz MJ, Calasanz MJ, Panizo C, Junza A, Han J, Prior C, Fortes P, Pio R, Oyarzabal J, Martinez-Baztan A, Paiva B, Moreno-Aliaga MJ, Odero MD, Agirre X, Yanes O, Prosper F, and Martinez-Climent JA
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Humans, Antineoplastic Agents pharmacology, Epigenesis, Genetic drug effects, Mitochondria drug effects, Neoplasms drug therapy
- Abstract
For millions of years, endogenous retroelements have remained transcriptionally silent within mammalian genomes by epigenetic mechanisms. Modern anticancer therapies targeting the epigenetic machinery awaken retroelement expression, inducing antiviral responses that eliminate tumors through mechanisms not completely understood. Here, we find that massive binding of epigenetically activated retroelements by RIG-I and MDA5 viral sensors promotes ATP hydrolysis and depletes intracellular energy, driving tumor killing independently of immune signaling. Energy depletion boosts compensatory ATP production by switching glycolysis to mitochondrial oxidative phosphorylation, thereby reversing the Warburg effect. However, hyperfunctional succinate dehydrogenase in mitochondrial electron transport chain generates excessive oxidative stress that unleashes RIP1-mediated necroptosis. To maintain ATP generation, hyperactive mitochondrial membrane blocks intrinsic apoptosis by increasing BCL2 dependency. Accordingly, drugs targeting BCL2 family proteins and epigenetic inhibitors yield synergistic responses in multiple cancer types. Thus, epigenetic therapy kills cancer cells by rewiring mitochondrial metabolism upon retroelement activation, which primes mitochondria to apoptosis by BH3-mimetics. SIGNIFICANCE: The state of viral mimicry induced by epigenetic therapies in cancer cells remodels mitochondrial metabolism and drives caspase-independent tumor cell death, which sensitizes to BCL2 inhibitor drugs. This novel mechanism underlies clinical efficacy of hypomethylating agents and venetoclax in acute myeloid leukemia, suggesting similar combination therapies for other incurable cancers. This article is highlighted in the In This Issue feature, p. 995 ., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
22. Deep MRD profiling defines outcome and unveils different modes of treatment resistance in standard- and high-risk myeloma.
- Author
-
Goicoechea I, Puig N, Cedena MT, Burgos L, Cordón L, Vidriales MB, Flores-Montero J, Gutierrez NC, Calasanz MJ, Ramos MM, Lara-Astiaso D, Vilas-Zornoza A, Alignani D, Rodriguez I, Sarvide S, Alameda D, Garcés JJ, Rodriguez S, Fresquet V, Celay J, Garcia-Sanz R, Martinez-Lopez J, Oriol A, Rios R, Martin-Sanchez J, Martinez-Martinez R, Sarra J, Hernandez MT, de la Rubia J, Krsnik I, Moraleda JM, Palomera L, Bargay J, Martinez-Climent JA, Orfao A, Rosiñol L, Mateos MV, Lahuerta JJ, Blade J, San Miguel J, and Paiva B
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Boron Compounds therapeutic use, Bortezomib therapeutic use, Chromosome Aberrations, Dexamethasone therapeutic use, Female, Flow Cytometry, Glycine analogs & derivatives, Glycine therapeutic use, Humans, Lenalidomide therapeutic use, Male, Middle Aged, Progression-Free Survival, Treatment Outcome, Drug Resistance, Neoplasm genetics, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Multiple Myeloma pathology, Neoplasm, Residual pathology
- Abstract
Patients with multiple myeloma (MM) carrying standard- or high-risk cytogenetic abnormalities (CAs) achieve similar complete response (CR) rates, but the later have inferior progression-free survival (PFS). This questions the legitimacy of CR as a treatment endpoint and represents a biological conundrum regarding the nature of tumor reservoirs that persist after therapy in high-risk MM. We used next-generation flow (NGF) cytometry to evaluate measurable residual disease (MRD) in MM patients with standard- vs high-risk CAs (n = 300 and 90, respectively) enrolled in the PETHEMA/GEM2012MENOS65 trial, and to identify mechanisms that determine MRD resistance in both patient subgroups (n = 40). The 36-month PFS rates were higher than 90% in patients with standard- or high-risk CAs achieving undetectable MRD. Persistent MRD resulted in a median PFS of ∼3 and 2 years in patients with standard- and high-risk CAs, respectively. Further use of NGF to isolate MRD, followed by whole-exome sequencing of paired diagnostic and MRD tumor cells, revealed greater clonal selection in patients with standard-risk CAs, higher genomic instability with acquisition of new mutations in high-risk MM, and no unifying genetic event driving MRD resistance. Conversely, RNA sequencing of diagnostic and MRD tumor cells uncovered the selection of MRD clones with singular transcriptional programs and reactive oxygen species-mediated MRD resistance in high-risk MM. Our study supports undetectable MRD as a treatment endpoint for patients with MM who have high-risk CAs and proposes characterizing MRD clones to understand and overcome MRD resistance. This trial is registered at www.clinicaltrials.gov as #NCT01916252., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
23. B-cell leukemia transdifferentiation to macrophage involves reconfiguration of DNA methylation for long-range regulation.
- Author
-
Bueno-Costa A, Piñeyro D, Soler M, Javierre BM, Raurell-Vila H, Subirana-Granés M, Pasquali L, Martinez-Climent JA, and Esteller M
- Subjects
- Humans, Macrophages metabolism, Tumor Cells, Cultured, Cell Lineage, Cell Transdifferentiation, DNA Methylation, Genome, Human, Leukemia, B-Cell genetics, Leukemia, B-Cell pathology, Macrophages pathology
- Published
- 2020
- Full Text
- View/download PDF
24. CLL intraclonal fractions exhibit established and recently acquired patterns of DNA methylation.
- Author
-
Bartholdy BA, Wang X, Yan XJ, Pascual M, Fan M, Barrientos J, Allen SL, Martinez-Climent JA, Rai KR, Chiorazzi N, Scharff MD, and Roa S
- Subjects
- B-Lymphocytes, DNA Methylation, Humans, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Abstract
Intraclonal subpopulations of circulating chronic lymphocytic leukemia (CLL) cells with different proliferative histories and reciprocal surface expression of CXCR4 and CD5 have been observed in the peripheral blood of CLL patients and named proliferative (PF), intermediate (IF), and resting (RF) cellular fractions. Here, we found that these intraclonal circulating fractions share persistent DNA methylation signatures largely associated with the mutation status of the immunoglobulin heavy chain locus (IGHV) and their origins from distinct stages of differentiation of antigen-experienced B cells. Increased leukemic birth rate, however, showed a very limited impact on DNA methylation of circulating CLL fractions independent of IGHV mutation status. Additionally, DNA methylation heterogeneity increased as leukemic cells advanced from PF to RF in the peripheral blood. This frequently co-occurred with heterochromatin hypomethylation and hypermethylation of Polycomb-repressed regions in the PF, suggesting accumulation of longevity-associated epigenetic features in recently born cells. On the other hand, transcriptional differences between paired intraclonal fractions confirmed their proliferative experience and further supported a linear advancement from PF to RF in the peripheral blood. Several of these differentially expressed genes showed unique associations with clinical outcome not evident in the bulk clone, supporting the pathological and therapeutic relevance of studying intraclonal CLL fractions. We conclude that independent methylation and transcriptional landscapes reflect both preexisting cell-of-origin fingerprints and more recently acquired hallmarks associated with the life cycle of circulating CLL cells., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
25. PD-1/PD-L1 immune checkpoint and p53 loss facilitate tumor progression in activated B-cell diffuse large B-cell lymphomas.
- Author
-
Pascual M, Mena-Varas M, Robles EF, Garcia-Barchino MJ, Panizo C, Hervas-Stubbs S, Alignani D, Sagardoy A, Martinez-Ferrandis JI, Bunting KL, Meier S, Sagaert X, Bagnara D, Guruceaga E, Blanco O, Celay J, Martínez-Baztan A, Casares N, Lasarte JJ, MacCarthy T, Melnick A, Martinez-Climent JA, and Roa S
- Subjects
- Animals, B-Lymphocytes pathology, B7-H1 Antigen genetics, Female, Humans, Immunotherapy, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Lymphoma, Large B-Cell, Diffuse therapy, Male, Mice, Mice, Transgenic, Programmed Cell Death 1 Receptor genetics, T-Lymphocytes immunology, T-Lymphocytes pathology, Tumor Suppressor Protein p53 genetics, B-Lymphocytes immunology, B7-H1 Antigen immunology, Gene Expression Regulation, Neoplastic, Lymphocyte Activation, Lymphoma, Large B-Cell, Diffuse immunology, Programmed Cell Death 1 Receptor immunology, Tumor Escape, Tumor Suppressor Protein p53 immunology
- Abstract
Refractory or relapsed diffuse large B-cell lymphoma (DLBCL) often associates with the activated B-cell-like (ABC) subtype and genetic alterations that drive constitutive NF-κB activation and impair B-cell terminal differentiation. Here, we show that DNA damage response by p53 is a central mechanism suppressing the pathogenic cooperation of IKK2ca-enforced canonical NF-κB and impaired differentiation resulting from Blimp1 loss in ABC-DLBCL lymphomagenesis. We provide evidences that the interplay between these genetic alterations and the tumor microenvironment select for additional molecular addictions that promote lymphoma progression, including aberrant coexpression of FOXP1 and the B-cell mutagenic enzyme activation-induced deaminase, and immune evasion through major histocompatibility complex class II downregulation, PD-L1 upregulation, and T-cell exhaustion. Consistently, PD-1 blockade cooperated with anti-CD20-mediated B-cell cytotoxicity, promoting extended T-cell reactivation and antitumor specificity that improved long-term overall survival in mice. Our data support a pathogenic cooperation among NF-κB-driven prosurvival, genetic instability, and immune evasion mechanisms in DLBCL and provide preclinical proof of concept for including PD-1/PD-L1 blockade in combinatorial immunotherapy for ABC-DLBCL., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
26. G-protein coupled receptor (GPCR) mutations in lymphoid malignancies: linking immune signaling activation and genetic abnormalities.
- Author
-
Martinez-Climent JA
- Subjects
- Genetic Profile, Humans, Mutation, Receptors, CCR6, Receptors, G-Protein-Coupled genetics, Signal Transduction, Lymphoma, B-Cell, Marginal Zone
- Published
- 2018
- Full Text
- View/download PDF
27. Richter transformation driven by Epstein-Barr virus reactivation during therapy-related immunosuppression in chronic lymphocytic leukaemia.
- Author
-
García-Barchino MJ, Sarasquete ME, Panizo C, Morscio J, Martinez A, Alcoceba M, Fresquet V, Gonzalez-Farre B, Paiva B, Young KH, Robles EF, Roa S, Celay J, Larrayoz M, Rossi D, Gaidano G, Montes-Moreno S, Piris MA, Balanzategui A, Jimenez C, Rodriguez I, Calasanz MJ, Larrayoz MJ, Segura V, Garcia-Muñoz R, Rabasa MP, Yi S, Li J, Zhang M, Xu-Monette ZY, Puig-Moron N, Orfao A, Böttcher S, Hernandez-Rivas JM, Miguel JS, Prosper F, Tousseyn T, Sagaert X, Gonzalez M, and Martinez-Climent JA
- Subjects
- Adult, Aged, B-Lymphocytes drug effects, B-Lymphocytes pathology, Cell Transformation, Neoplastic pathology, Epstein-Barr Virus Infections drug therapy, Epstein-Barr Virus Infections pathology, Female, Herpesvirus 4, Human genetics, Humans, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics, Male, Middle Aged, Cell Transformation, Neoplastic drug effects, Herpesvirus 4, Human drug effects, Immunosuppressive Agents pharmacology, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy
- Abstract
The increased risk of Richter transformation (RT) in patients with chronic lymphocytic leukaemia (CLL) due to Epstein-Barr virus (EBV) reactivation during immunosuppressive therapy with fludarabine other targeted agents remains controversial. Among 31 RT cases classified as diffuse large B-cell lymphoma (DLBCL), seven (23%) showed EBV expression. In contrast to EBV
- tumours, EBV+ DLBCLs derived predominantly from IGVH-hypermutated CLL, and they also showed CLL-unrelated IGVH sequences more frequently. Intriguingly, despite having different cellular origins, clonally related and unrelated EBV+ DLBCLs shared a previous history of immunosuppressive chemo-immunotherapy, a non-germinal centre DLBCL phenotype, EBV latency programme type II or III, and very short survival. These data suggested that EBV reactivation during therapy-related immunosuppression can transform either CLL cells or non-tumoural B lymphocytes into EBV+ DLBCL. To investigate this hypothesis, xenogeneic transplantation of blood cells from 31 patients with CLL and monoclonal B-cell lymphocytosis (MBL) was performed in Rag2-/- IL2γc-/- mice. Remarkably, the recipients' impaired immunosurveillance favoured the spontaneous outgrowth of EBV+ B-cell clones from 95% of CLL and 64% of MBL patients samples, but not from healthy donors. Eventually, these cells generated monoclonal tumours (mostly CLL-unrelated but also CLL-related), recapitulating the principal features of EBV+ DLBCL in patients. Accordingly, clonally related and unrelated EBV+ DLBCL xenografts showed indistinguishable cellular, virological and molecular features, and synergistically responded to combined inhibition of EBV replication with ganciclovir and B-cell receptor signalling with ibrutinib in vivo. Our study underscores the risk of RT driven by EBV in CLL patients receiving immunosuppressive therapies, and provides the scientific rationale for testing ganciclovir and ibrutinib in EBV+ DLBCL. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd., (Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)- Published
- 2018
- Full Text
- View/download PDF
28. Discovery of first-in-class reversible dual small molecule inhibitors against G9a and DNMTs in hematological malignancies.
- Author
-
San José-Enériz E, Agirre X, Rabal O, Vilas-Zornoza A, Sanchez-Arias JA, Miranda E, Ugarte A, Roa S, Paiva B, Estella-Hermoso de Mendoza A, Alvarez RM, Casares N, Segura V, Martín-Subero JI, Ogi FX, Soule P, Santiveri CM, Campos-Olivas R, Castellano G, de Barrena MGF, Rodriguez-Madoz JR, García-Barchino MJ, Lasarte JJ, Avila MA, Martinez-Climent JA, Oyarzabal J, and Prosper F
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Apoptosis genetics, Apoptosis immunology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Crystallography, X-Ray, DNA Modification Methylases chemistry, DNA Modification Methylases genetics, DNA Modification Methylases metabolism, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical, Enzyme Inhibitors chemistry, Enzyme Inhibitors therapeutic use, Epigenesis, Genetic drug effects, Female, Hematologic Neoplasms genetics, Hematologic Neoplasms immunology, Hematologic Neoplasms mortality, Histocompatibility Antigens chemistry, Histocompatibility Antigens genetics, Histocompatibility Antigens metabolism, Histone-Lysine N-Methyltransferase chemistry, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Humans, Interferons immunology, Interferons metabolism, Mice, Mice, Inbred BALB C, Microsomes, Liver, Molecular Docking Simulation, Survival Analysis, Treatment Outcome, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, DNA Modification Methylases antagonists & inhibitors, Drug Design, Enzyme Inhibitors pharmacology, Hematologic Neoplasms drug therapy, Histone-Lysine N-Methyltransferase antagonists & inhibitors
- Abstract
The indisputable role of epigenetics in cancer and the fact that epigenetic alterations can be reversed have favoured development of epigenetic drugs. In this study, we design and synthesize potent novel, selective and reversible chemical probes that simultaneously inhibit the G9a and DNMTs methyltransferase activity. In vitro treatment of haematological neoplasia (acute myeloid leukaemia-AML, acute lymphoblastic leukaemia-ALL and diffuse large B-cell lymphoma-DLBCL) with the lead compound CM-272, inhibits cell proliferation and promotes apoptosis, inducing interferon-stimulated genes and immunogenic cell death. CM-272 significantly prolongs survival of AML, ALL and DLBCL xenogeneic models. Our results represent the discovery of first-in-class dual inhibitors of G9a/DNMTs and establish this chemical series as a promising therapeutic tool for unmet needs in haematological tumours.
- Published
- 2017
- Full Text
- View/download PDF
29. C/EBPα Induces Highly Efficient Macrophage Transdifferentiation of B Lymphoma and Leukemia Cell Lines and Impairs Their Tumorigenicity.
- Author
-
Rapino F, Robles EF, Richter-Larrea JA, Kallin EM, Martinez-Climent JA, and Graf T
- Published
- 2017
- Full Text
- View/download PDF
30. ODZ1 allows glioblastoma to sustain invasiveness through a Myc-dependent transcriptional upregulation of RhoA.
- Author
-
Talamillo A, Grande L, Ruiz-Ontañon P, Velasquez C, Mollinedo P, Torices S, Sanchez-Gomez P, Aznar A, Esparis-Ogando A, Lopez-Lopez C, Lafita C, Berciano MT, Montero JA, Vazquez-Barquero A, Segura V, Villagra NT, Pandiella A, Lafarga M, Leon J, Martinez-Climent JA, Sanz-Moreno V, and Fernandez-Luna JL
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Gene Knockout Techniques, Glioblastoma mortality, Glioblastoma pathology, Heterografts, Humans, Mice, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Nerve Tissue Proteins deficiency, Nerve Tissue Proteins metabolism, Prognosis, Protein Transport, Proteolysis, Signal Transduction, Tenascin deficiency, Tenascin metabolism, Up-Regulation, rho-Associated Kinases metabolism, Gene Expression Regulation, Neoplastic, Glioblastoma genetics, Glioblastoma metabolism, Nerve Tissue Proteins genetics, Proto-Oncogene Proteins c-myc metabolism, Tenascin genetics, Transcription, Genetic, rhoA GTP-Binding Protein genetics
- Abstract
Long-term survival remains low for most patients with glioblastoma (GBM), which reveals the need for markers of disease outcome and novel therapeutic targets. We describe that ODZ1 (also known as TENM1), a type II transmembrane protein involved in fetal brain development, plays a crucial role in the invasion of GBM cells. Differentiation of glioblastoma stem-like cells drives the nuclear translocation of an intracellular fragment of ODZ1 through proteolytic cleavage by signal peptide peptidase-like 2a. The intracellular fragment of ODZ1 promotes cytoskeletal remodelling of GBM cells and invasion of the surrounding environment both in vitro and in vivo. Absence of ODZ1 by gene deletion or downregulation of ODZ1 by small interfering RNAs drastically reduces the invasive capacity of GBM cells. This activity is mediated by an ODZ1-triggered transcriptional pathway, through the E-box binding Myc protein, that promotes the expression and activation of Ras homolog family member A (RhoA) and subsequent activation of Rho-associated, coiled-coil containing protein kinase (ROCK). Overexpression of ODZ1 in GBM cells reduced survival of xenografted mice. Consistently, analysis of 122 GBM tumour samples revealed that the number of ODZ1-positive cells inversely correlated with overall and progression-free survival. Our findings establish a novel marker of invading GBM cells and consequently a potential marker of disease progression and a therapeutic target in GBM.
- Published
- 2017
- Full Text
- View/download PDF
31. Divergence of Vascular Specification in Visceral Lymphoid Organs-Genetic Determinants and Differentiation Checkpoints.
- Author
-
Kellermayer Z, Hayasaka H, Kajtár B, Simon D, Robles EF, Martinez-Climent JA, and Balogh P
- Subjects
- Animals, Animals, Genetically Modified, Endothelium, Vascular cytology, High-Throughput Nucleotide Sequencing, Homeodomain Proteins genetics, Homeodomain Proteins immunology, Humans, Intestines blood supply, Intestines embryology, Intestines physiology, Leukocytes immunology, Lymphoid Tissue embryology, Mice, Organogenesis, Peyer's Patches blood supply, Peyer's Patches embryology, Peyer's Patches physiology, Sequence Analysis, RNA, Spleen blood supply, Spleen embryology, Spleen physiology, Transcription Factors genetics, Transcription Factors immunology, Cell Differentiation immunology, Endothelial Cells physiology, Endothelium, Vascular physiology, Gene Expression Regulation immunology, Lymphoid Tissue blood supply, Lymphoid Tissue physiology
- Abstract
Despite their functional similarities, peripheral lymphoid tissues are remarkably different according to their developmental properties and structural characteristics, including their specified vasculature. Access of leukocytes to these organs critically depends on their interactions with the local endothelium, where endothelial cells are patterned to display a restricted set of adhesion molecules and other regulatory compounds necessary for extravasation. Recent advances in high throughput analyses of highly purified endothelial subsets in various lymphoid tissues as well as the expansion of various transgenic animal models have shed new light on the transcriptional complexities of lymphoid tissue vascular endothelium. This review is aimed at providing a comprehensive analysis linking the functional competence of spleen and intestinal lymphoid tissues with the developmental programming and functional divergence of their vascular specification, with particular emphasis on the transcriptional control of endothelial cells exerted by Nkx2.3 homeodomain transcription factor.
- Published
- 2016
- Full Text
- View/download PDF
32. Homeobox NKX2-3 promotes marginal-zone lymphomagenesis by activating B-cell receptor signalling and shaping lymphocyte dynamics.
- Author
-
Robles EF, Mena-Varas M, Barrio L, Merino-Cortes SV, Balogh P, Du MQ, Akasaka T, Parker A, Roa S, Panizo C, Martin-Guerrero I, Siebert R, Segura V, Agirre X, Macri-Pellizeri L, Aldaz B, Vilas-Zornoza A, Zhang S, Moody S, Calasanz MJ, Tousseyn T, Broccardo C, Brousset P, Campos-Sanchez E, Cobaleda C, Sanchez-Garcia I, Fernandez-Luna JL, Garcia-Muñoz R, Pena E, Bellosillo B, Salar A, Baptista MJ, Hernandez-Rivas JM, Gonzalez M, Terol MJ, Climent J, Ferrandez A, Sagaert X, Melnick AM, Prosper F, Oscier DG, Carrasco YR, Dyer MJ, and Martinez-Climent JA
- Subjects
- Animals, Cell Adhesion Molecules genetics, Cell Adhesion Molecules metabolism, Gene Expression Profiling, Homeodomain Proteins metabolism, Humans, Kaplan-Meier Estimate, Lymphoid Tissue metabolism, Lymphoma, B-Cell, Marginal Zone metabolism, Mice, Inbred C57BL, Mice, Knockout, Phosphorylation, Receptors, Antigen, B-Cell metabolism, Syk Kinase genetics, Syk Kinase metabolism, Transcription Factors metabolism, Homeodomain Proteins genetics, Lymphocytes metabolism, Lymphoma, B-Cell, Marginal Zone genetics, Receptors, Antigen, B-Cell genetics, Signal Transduction genetics, Transcription Factors genetics
- Abstract
NKX2 homeobox family proteins have a role in cancer development. Here we show that NKX2-3 is overexpressed in tumour cells from a subset of patients with marginal-zone lymphomas, but not with other B-cell malignancies. While Nkx2-3-deficient mice exhibit the absence of marginal-zone B cells, transgenic mice with expression of NKX2-3 in B cells show marginal-zone expansion that leads to the development of tumours, faithfully recapitulating the principal clinical and biological features of human marginal-zone lymphomas. NKX2-3 induces B-cell receptor signalling by phosphorylating Lyn/Syk kinases, which in turn activate multiple integrins (LFA-1, VLA-4), adhesion molecules (ICAM-1, MadCAM-1) and the chemokine receptor CXCR4. These molecules enhance migration, polarization and homing of B cells to splenic and extranodal tissues, eventually driving malignant transformation through triggering NF-κB and PI3K-AKT pathways. This study implicates oncogenic NKX2-3 in lymphomagenesis, and provides a valid experimental mouse model for studying the biology and therapy of human marginal-zone B-cell lymphomas.
- Published
- 2016
- Full Text
- View/download PDF
33. Combined clinical and genomic signatures for the prognosis of early stage non-small cell lung cancer based on gene copy number alterations.
- Author
-
Aramburu A, Zudaire I, Pajares MJ, Agorreta J, Orta A, Lozano MD, Gúrpide A, Gómez-Román J, Martinez-Climent JA, Jassem J, Skrzypski M, Suraokar M, Behrens C, Wistuba II, Pio R, Rubio A, and Montuenga LM
- Subjects
- Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung surgery, Female, Gene Expression Regulation, Neoplastic, Genome, Human, Genomics, Humans, Kaplan-Meier Estimate, Male, Neoplasm Proteins biosynthesis, Neoplasm Staging, Carcinoma, Non-Small-Cell Lung genetics, Gene Dosage genetics, Neoplasm Proteins genetics, Prognosis
- Abstract
Background: The development of a more refined prognostic methodology for early non-small cell lung cancer (NSCLC) is an unmet clinical need. An accurate prognostic tool might help to select patients at early stages for adjuvant therapies., Results: A new integrated bioinformatics searching strategy, that combines gene copy number alterations and expression, together with clinical parameters was applied to derive two prognostic genomic signatures. The proposed methodology combines data from patients with and without clinical data with a priori information on the ability of a gene to be a prognostic marker. Two initial candidate sets of 513 and 150 genes for lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC), respectively, were generated by identifying genes which have both: a) significant correlation between copy number and gene expression, and b) significant prognostic value at the gene expression level in external databases. From these candidates, two panels of 7 (ADC) and 5 (SCC) genes were further identified via semi-supervised learning. These panels, together with clinical data (stage, age and sex), were used to construct the ADC and SCC hazard scores combining clinical and genomic data. The signatures were validated in two independent datasets (n = 73 for ADC, n = 97 for SCC), confirming that the prognostic value of both clinical-genomic models is robust, statistically significant (P = 0.008 for ADC and P = 0.019 for SCC) and outperforms both the clinical models (P = 0.060 for ADC and P = 0.121 for SCC) and the genomic models applied separately (P = 0.350 for ADC and P = 0.269 for SCC)., Conclusion: The present work provides a methodology to generate a robust signature using copy number data that can be potentially used to any cancer. Using it, we found new prognostic scores based on tumor DNA that, jointly with clinical information, are able to predict overall survival (OS) in patients with early-stage ADC and SCC.
- Published
- 2015
- Full Text
- View/download PDF
34. KLF2 mutation is the most frequent somatic change in splenic marginal zone lymphoma and identifies a subset with distinct genotype.
- Author
-
Clipson A, Wang M, de Leval L, Ashton-Key M, Wotherspoon A, Vassiliou G, Bolli N, Grove C, Moody S, Escudero-Ibarz L, Gundem G, Brugger K, Xue X, Mi E, Bench A, Scott M, Liu H, Follows G, Robles EF, Martinez-Climent JA, Oscier D, Watkins AJ, and Du MQ
- Subjects
- Biopsy, CARD Signaling Adaptor Proteins metabolism, DNA-Binding Proteins metabolism, Exome, Frameshift Mutation, Gene Rearrangement, B-Lymphocyte, Heavy Chain, Genetic Variation, Genotype, Guanylate Cyclase metabolism, Humans, Intracellular Signaling Peptides and Proteins metabolism, Lymphoma metabolism, Lymphoma, B-Cell, Marginal Zone diagnosis, Mutation, Missense, Nuclear Proteins metabolism, Polymerase Chain Reaction, Receptor, Notch2 metabolism, Recurrence, Sequence Analysis, DNA, Signal Transduction, Splenic Neoplasms diagnosis, Tumor Necrosis Factor alpha-Induced Protein 3, Kruppel-Like Transcription Factors genetics, Lymphoma, B-Cell, Marginal Zone genetics, Mutation, Splenic Neoplasms genetics
- Abstract
To characterise the genetics of splenic marginal zone lymphoma (SMZL), we performed whole exome sequencing of 16 cases and identified novel recurrent inactivating mutations in Kruppel-like factor 2 (KLF2), a gene whose deficiency was previously shown to cause splenic marginal zone hyperplasia in mice. KLF2 mutation was found in 40 (42%) of 96 SMZLs, but rarely in other B-cell lymphomas. The majority of KLF2 mutations were frameshift indels or nonsense changes, with missense mutations clustered in the C-terminal zinc finger domains. Functional assays showed that these mutations inactivated the ability of KLF2 to suppress NF-κB activation by TLR, BCR, BAFFR and TNFR signalling. Further extensive investigations revealed common and distinct genetic changes between SMZL with and without KLF2 mutation. IGHV1-2 rearrangement and 7q deletion were primarily seen in SMZL with KLF2 mutation, while MYD88 and TP53 mutations were nearly exclusively found in those without KLF2 mutation. NOTCH2, TRAF3, TNFAIP3 and CARD11 mutations were observed in SMZL both with and without KLF2 mutation. Taken together, KLF2 mutation is the most common genetic change in SMZL and identifies a subset with a distinct genotype characterised by multi-genetic changes. These different genetic changes may deregulate various signalling pathways and generate cooperative oncogenic properties, thereby contributing to lymphomagenesis.
- Published
- 2015
- Full Text
- View/download PDF
35. DNA methylation profiling identifies two splenic marginal zone lymphoma subgroups with different clinical and genetic features.
- Author
-
Arribas AJ, Rinaldi A, Mensah AA, Kwee I, Cascione L, Robles EF, Martinez-Climent JA, Oscier D, Arcaini L, Baldini L, Marasca R, Thieblemont C, Briere J, Forconi F, Zamò A, Bonifacio M, Mollejo M, Facchetti F, Dirnhofer S, Ponzoni M, Bhagat G, Piris MA, Gaidano G, Zucca E, Rossi D, and Bertoni F
- Subjects
- Adult, Aged, Aged, 80 and over, Cell Proliferation, Cell Transformation, Neoplastic, Cluster Analysis, DNA Mutational Analysis, Female, Gene Expression Profiling, Humans, Kaplan-Meier Estimate, Kruppel-Like Factor 4, Lymphoma, B-Cell, Marginal Zone diagnosis, Lymphoma, B-Cell, Marginal Zone mortality, Male, Middle Aged, Mutation, Phenotype, Prognosis, Promoter Regions, Genetic, Splenic Neoplasms diagnosis, Splenic Neoplasms mortality, Treatment Outcome, DNA Methylation, Lymphoma, B-Cell, Marginal Zone genetics, Splenic Neoplasms genetics
- Abstract
Splenic marginal zone lymphoma is a rare lymphoma. Loss of 7q31 and somatic mutations affecting the NOTCH2 and KLF2 genes are the commonest genomic aberrations. Epigenetic changes can be pharmacologically reverted; therefore, identification of groups of patients with specific epigenomic alterations might have therapeutic relevance. Here we integrated genome-wide DNA-promoter methylation profiling with gene expression profiling, and clinical and biological variables. An unsupervised clustering analysis of a test series of 98 samples identified 2 clusters with different degrees of promoter methylation. The cluster comprising samples with higher-promoter methylation (High-M) had a poorer overall survival compared with the lower (Low-M) cluster. The prognostic relevance of the High-M phenotype was confirmed in an independent validation set of 36 patients. In the whole series, the High-M phenotype was associated with IGHV1-02 usage, mutations of NOTCH2 gene, 7q31-32 loss, and histologic transformation. In the High-M set, a number of tumor-suppressor genes were methylated and repressed. PRC2 subunit genes and several prosurvival lymphoma genes were unmethylated and overexpressed. A model based on the methylation of 3 genes (CACNB2, HTRA1, KLF4) identified a poorer-outcome patient subset. Exposure of splenic marginal zone lymphoma cell lines to a demethylating agent caused partial reversion of the High-M phenotype and inhibition of proliferation., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
36. The origin and targeting of mucosa-associated lymphoid tissue lymphomas.
- Author
-
Martinez-Climent JA
- Subjects
- Animals, Caspases genetics, Chronic Disease, Humans, Inflammation complications, Lymphoma, B-Cell, Marginal Zone drug therapy, Lymphoma, B-Cell, Marginal Zone metabolism, Molecular Targeted Therapy, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, Mutation, NF-kappa B metabolism, Neoplasm Proteins genetics, Signal Transduction, Translocation, Genetic, Lymphoma, B-Cell, Marginal Zone etiology
- Abstract
Purpose of Review: Extranodal mucosa-associated lymphoid tissue (MALT lymphoma) is a distinct clinical-pathological entity that can be distinguished from other lymphomas by a number of unique features, including their location in various extranodal sites, being preceded by chronic inflammatory or infection processes; a characteristic histopathological picture; and the presence of exclusive chromosomal translocations which increase MALT1 proteolytic activity to promote constitutive NF-κB signaling and eventually drive lymphomagenesis., Recent Findings: This review explores the major molecular and cellular events that participate in MALT lymphoma pathogenesis, focusing on gastric MALT lymphoma as a model of chronic inflammation-induced tumor development. In addition, the pivotal roles of activated MALT1 protease, its substrate TNFAIP3/A20, and the MyD88 adaptor protein in abnormally triggering downstream NF-κB pathway are overviewed. These new insights provide a mechanistic basis for using novel therapies targeting MALT1 protease or IRAK4 kinase activities. Finally, the putative cellular origin of MALT lymphomas is also discussed., Summary: Over the last decade, unraveling the biological complexity of MALT lymphomas has shed light on the fundamental cellular and molecular aspects of the disease that are to be translated into clinical diagnostics and therapy.
- Published
- 2014
- Full Text
- View/download PDF
37. Acquired mutations in BCL2 family proteins conferring resistance to the BH3 mimetic ABT-199 in lymphoma.
- Author
-
Fresquet V, Rieger M, Carolis C, García-Barchino MJ, and Martinez-Climent JA
- Subjects
- Amino Acid Substitution, Animals, Antineoplastic Agents, Apoptosis drug effects, Apoptosis genetics, Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Humans, Lymphoma drug therapy, Lymphoma genetics, Lymphoma pathology, Mice, Mice, Knockout, Mitochondria genetics, Mitochondria metabolism, Mitochondria pathology, bcl-2-Associated X Protein genetics, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Drug Resistance, Neoplasm drug effects, Lymphoma metabolism, Mutation, Missense, Sulfonamides pharmacology, bcl-2-Associated X Protein metabolism
- Abstract
Acquired resistance to targeted drugs is emerging as an obstacle to successful cancer treatment. Recently, a BCL2-selective BH3 mimetic termed ABT-199 showed promising therapeutic results in BCL2-dependent tumors. Based on its high affinity for BCL2, we studied potential mechanisms conferring resistance upon ABT-199 therapy, aiming to anticipate its occurrence in the clinic. Two models of resistant lymphomas were established by continuous ABT-199 exposure. In resistant Bcl2-expressing mouse lymphoma cells, 2 missense mutations within the Bcl2 BH3 domain were identified. Both F101C and F101L mutations impeded ABT-199 binding to the BH3 domain, therefore suppressing mitochondrial apoptosis. In resistant human lymphoma cells, a missense mutation in the C-terminal transmembrane domain of proapoptotic BAX (G179E) was found, which abrogated BAX anchoring to mitochondria and blocked ABT-199-induced apoptosis both in vitro and in vivo. Importantly, G179E BAX mutation also induced partial cross-resistance to other antineoplastic drugs. Our study reveals the acquisition of mutations in BCL2 family proteins as a novel mechanism of apoptosis resistance in cancer. These results anticipate the potential development of such mutations in patients treated with ABT-199, providing a basis to preventing their occurrence and to designing drugs able to circumvent the acquired resistance., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
38. Involvement of miRNAs in the differentiation of human glioblastoma multiforme stem-like cells.
- Author
-
Aldaz B, Sagardoy A, Nogueira L, Guruceaga E, Grande L, Huse JT, Aznar MA, Díez-Valle R, Tejada-Solís S, Alonso MM, Fernandez-Luna JL, Martinez-Climent JA, and Malumbres R
- Subjects
- Animals, Apoptosis genetics, Astrocytes metabolism, Astrocytes pathology, Biomarkers, Tumor metabolism, Brain Neoplasms surgery, Cell Line, Tumor, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Glial Fibrillary Acidic Protein metabolism, Glioblastoma surgery, Humans, Membrane Proteins metabolism, Mice, Mice, Inbred BALB C, MicroRNAs genetics, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Neoplastic Stem Cells pathology, Nestin metabolism, Neurons metabolism, Neurons pathology, Oligonucleotide Array Sequence Analysis, Phosphoproteins metabolism, Spheroids, Cellular metabolism, Spheroids, Cellular pathology, Tubulin metabolism, Brain Neoplasms genetics, Brain Neoplasms pathology, Cell Differentiation genetics, Glioblastoma genetics, Glioblastoma pathology, MicroRNAs metabolism, Neoplastic Stem Cells metabolism
- Abstract
Glioblastoma multiforme (GBM)-initiating cells (GICs) represent a tumor subpopulation with neural stem cell-like properties that is responsible for the development, progression and therapeutic resistance of human GBM. We have recently shown that blockade of NFκB pathway promotes terminal differentiation and senescence of GICs both in vitro and in vivo, indicating that induction of differentiation may be a potential therapeutic strategy for GBM. MicroRNAs have been implicated in the pathogenesis of GBM, but a high-throughput analysis of their role in GIC differentiation has not been reported. We have established human GIC cell lines that can be efficiently differentiated into cells expressing astrocytic and neuronal lineage markers. Using this in vitro system, a microarray-based high-throughput analysis to determine global expression changes of microRNAs during differentiation of GICs was performed. A number of changes in the levels of microRNAs were detected in differentiating GICs, including over-expression of hsa-miR-21, hsa-miR-29a, hsa-miR-29b, hsa-miR-221 and hsa-miR-222, and down-regulation of hsa-miR-93 and hsa-miR-106a. Functional studies showed that miR-21 over-expression in GICs induced comparable cell differentiation features and targeted SPRY1 mRNA, which encodes for a negative regulator of neural stem-cell differentiation. In addition, miR-221 and miR-222 inhibition in differentiated cells restored the expression of stem cell markers while reducing differentiation markers. Finally, miR-29a and miR-29b targeted MCL1 mRNA in GICs and increased apoptosis. Our study uncovers the microRNA dynamic expression changes occurring during differentiation of GICs, and identifies miR-21 and miR-221/222 as key regulators of this process.
- Published
- 2013
- Full Text
- View/download PDF
39. LITAF, a BCL6 target gene, regulates autophagy in mature B-cell lymphomas.
- Author
-
Bertolo C, Roa S, Sagardoy A, Mena-Varas M, Robles EF, Martinez-Ferrandis JI, Sagaert X, Tousseyn T, Orta A, Lossos IS, Amar S, Natkunam Y, Briones J, Melnick A, Malumbres R, and Martinez-Climent JA
- Subjects
- B-Lymphocyte Subsets metabolism, Cell Line, Tumor, DNA-Binding Proteins metabolism, Gene Expression Regulation, Neoplastic physiology, Humans, Introns, Lymphoma, B-Cell metabolism, Lymphoma, B-Cell pathology, Neoplasm Proteins metabolism, Nuclear Proteins metabolism, Proto-Oncogene Proteins c-bcl-6, Transcription Factors metabolism, Transcription, Genetic, Tumor Necrosis Factor-alpha metabolism, Autophagy genetics, Lymphoma, B-Cell genetics, Neoplasm Proteins genetics, Nuclear Proteins genetics, Transcription Factors genetics
- Abstract
We have previously reported that LITAF is silenced by promoter hypermethylation in germinal centre-derived B-cell lymphomas, but beyond these data the regulation and function of lipopolysaccharide-induced tumour necrosis factor (TNF) factor (LITAF) in B cells are unknown. Gene expression and immunohistochemical studies revealed that LITAF and BCL6 show opposite expression in tonsil B-cell subpopulations and B-cell lymphomas, suggesting that BCL6 may regulate LITAF expression. Accordingly, BCL6 silencing increased LITAF expression, and chromatin immunoprecipitation and luciferase reporter assays demonstrated a direct transcriptional repression of LITAF by BCL6. Gain- and loss-of-function experiments in different B-cell lymphoma cell lines revealed that, in contrast to its function in monocytes, LITAF does not induce lipopolysaccharide-mediated TNF secretion in B cells. However, gene expression microarrays defined a LITAF-related transcriptional signature containing genes regulating autophagy, including MAP1LC3B (LC3B). In addition, immunofluorescence analysis co-localized LITAF with autophagosomes, further suggesting a possible role in autophagy modulation. Accordingly, ectopic LITAF expression in B-cell lymphoma cells enhanced autophagy responses to starvation, which were impaired upon LITAF silencing. Our results indicate that the BCL6-mediated transcriptional repression of LITAF may inhibit autophagy in B cells during the germinal centre reaction, and suggest that the constitutive repression of autophagy responses in BCL6-driven lymphomas may contribute to lymphomagenesis., (© 2013 John Wiley & Sons Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
40. Cellular plasticity confers migratory and invasive advantages to a population of glioblastoma-initiating cells that infiltrate peritumoral tissue.
- Author
-
Ruiz-Ontañon P, Orgaz JL, Aldaz B, Elosegui-Artola A, Martino J, Berciano MT, Montero JA, Grande L, Nogueira L, Diaz-Moralli S, Esparís-Ogando A, Vazquez-Barquero A, Lafarga M, Pandiella A, Cascante M, Segura V, Martinez-Climent JA, Sanz-Moreno V, and Fernandez-Luna JL
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Line, Cell Line, Tumor, Cell Movement genetics, Chick Embryo, Down-Regulation, Female, Glioblastoma genetics, Glioblastoma metabolism, Heterografts, Humans, Integrin alphaVbeta3 genetics, Integrin alphaVbeta3 metabolism, Mice, Mice, Inbred BALB C, Neoplasm Invasiveness, Neoplastic Stem Cells metabolism, Signal Transduction, Tumor Cells, Cultured, Up-Regulation, rac GTP-Binding Proteins genetics, rac GTP-Binding Proteins metabolism, rhoA GTP-Binding Protein genetics, rhoA GTP-Binding Protein metabolism, Brain Neoplasms pathology, Cell Movement physiology, Glioblastoma pathology, Neoplastic Stem Cells pathology
- Abstract
Glioblastoma (GBM) is associated with infiltration of peritumoral (PT) parenchyma by isolated tumor cells that leads to tumor regrowth. Recently, GBM stem-like or initiating cells (GICs) have been identified in the PT area, but whether these GICs have enhanced migratory and invasive capabilities compared with GICs from the tumor mass (TM) is presently unknown. We isolated GICs from the infiltrated PT tissue and the TM of three patients and found that PT cells have an advantage over TM cells in two-dimensional and three-dimensional migration and invasion assays. Interestingly, PT cells display a high plasticity in protrusion formation and cell shape and their migration is insensitive to substrate stiffness, which represent advantages to infiltrate microenvironments of different rigidity. Furthermore, mouse and chicken embryo xenografts revealed that only PT cells showed a dispersed distribution pattern, closely associated to blood vessels. Consistent with cellular plasticity, simultaneous Rac and RhoA activation are required for the enhanced invasive capacity of PT cells. Moreover, Rho GTPase signaling modulators αVβ3 and p27 play key roles in GIC invasiveness. Of note, p27 is upregulated in TM cells and inhibits RhoA activity. Gene silencing of p27 increased the invasive capacity of TM GICs. Additionally, β3 integrin is upregulated in PT cells. Blockade of dimeric integrin αVβ3, a Rac activator, reduced the invasive capacity of PT GICs in vitro and abrogated the spreading of PT cells into chicken embryos. Thus, our results describe the invasive features acquired by a unique subpopulation of GICs that infiltrate neighboring tissue., (Copyright © 2013 AlphaMed Press.)
- Published
- 2013
- Full Text
- View/download PDF
41. Downregulation of FOXP1 is required during germinal center B-cell function.
- Author
-
Sagardoy A, Martinez-Ferrandis JI, Roa S, Bunting KL, Aznar MA, Elemento O, Shaknovich R, Fontán L, Fresquet V, Perez-Roger I, Robles EF, De Smedt L, Sagaert X, Melnick A, and Martinez-Climent JA
- Subjects
- Animals, Cell Differentiation immunology, Cell Line, DNA-Binding Proteins immunology, DNA-Binding Proteins metabolism, Down-Regulation immunology, Forkhead Transcription Factors immunology, Germinal Center immunology, Humans, Lymphoma metabolism, Mice, Mice, Transgenic, Palatine Tonsil cytology, Proto-Oncogene Proteins c-bcl-6, Repressor Proteins immunology, Transcriptional Activation immunology, B-Lymphocytes immunology, B-Lymphocytes metabolism, Forkhead Transcription Factors metabolism, Germinal Center cytology, Lymphoma immunology, Repressor Proteins metabolism
- Abstract
B-cell maturation and germinal center (GC) formation are dependent on the interplay between BCL6 and other transcriptional regulators. FOXP1 is a transcription factor that regulates early B-cell development, but whether it plays a role in mature B cells is unknown. Analysis of human tonsillar B-cell subpopulations revealed that FOXP1 shows the opposite expression pattern to BCL6, suggesting that FOXP1 regulates the transition from resting follicular B cell to activated GC B cell. Chromatin immunoprecipitation-on-chip and gene expression assays on B cells indicated that FOXP1 acts as a transcriptional activator and repressor of genes involved in the GC reaction, half of which are also BCL6 targets. To study FOXP1 function in vivo, we developed transgenic mice expressing human FOXP1 in lymphoid cells. These mice exhibited irregular formation of splenic GCs, showing a modest increase in naïve and marginal-zone B cells and a significant decrease in GC B cells. Furthermore, aberrant expression of FOXP1 impaired transcription of noncoding γ1 germline transcripts and inhibited efficient class switching to the immunoglobulin G1 isotype. These studies show that FOXP1 is physiologically downregulated in GC B cells and that aberrant expression of FOXP1 impairs mechanisms triggered by B-cell activation, potentially contributing to B-cell lymphomagenesis.
- Published
- 2013
- Full Text
- View/download PDF
42. C/EBPα induces highly efficient macrophage transdifferentiation of B lymphoma and leukemia cell lines and impairs their tumorigenicity.
- Author
-
Rapino F, Robles EF, Richter-Larrea JA, Kallin EM, Martinez-Climent JA, and Graf T
- Subjects
- Animals, Antineoplastic Agents, Hormonal therapeutic use, Antineoplastic Agents, Hormonal toxicity, Cell Line, Tumor, Cell Lineage, Cell Transdifferentiation drug effects, Humans, Leukemia metabolism, Leukemia pathology, Lymphoma, B-Cell drug therapy, Lymphoma, B-Cell metabolism, Lymphoma, B-Cell mortality, Macrophages metabolism, Mice, Phagocytosis, Tamoxifen therapeutic use, Tamoxifen toxicity, Transcriptome, Transplantation, Heterologous, CCAAT-Enhancer-Binding Protein-alpha metabolism, Macrophages cytology
- Abstract
Earlier work demonstrated that the transcription factor C/EBPα can convert immature and mature murine B lineage cells into functional macrophages. Testing >20 human lymphoma and leukemia B cell lines, we found that most can be transdifferentiated at least partially into macrophage-like cells, provided that C/EBPα is expressed at sufficiently high levels. A tamoxifen-inducible subclone of the Seraphina Burkitt lymphoma line, expressing C/EBPαER, could be efficiently converted into phagocytic and quiescent cells with a transcriptome resembling normal macrophages. The converted cells retained their phenotype even when C/EBPα was inactivated, a hallmark of cell reprogramming. Interestingly, C/EBPα induction also impaired the cells' tumorigenicity. Likewise, C/EBPα efficiently converted a lymphoblastic leukemia B cell line into macrophage-like cells, again dramatically impairing their tumorigenicity. Our experiments show that human cancer cells can be induced by C/EBPα to transdifferentiate into seemingly normal cells at high frequencies and provide a proof of principle for a potential new therapeutic strategy for treating B cell malignancies., (Copyright © 2013 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
43. Genomic profiling of mantle cell lymphoma.
- Author
-
Menanteau MR and Martinez-Climent JA
- Subjects
- Chromosome Aberrations, Comparative Genomic Hybridization methods, Cyclin D1 genetics, Humans, Lymphoma, Mantle-Cell diagnosis, Lymphoma, Mantle-Cell pathology, Lymphoma, Mantle-Cell therapy, Prognosis, Uniparental Disomy genetics, Gene Expression Profiling methods, Lymphoma, Mantle-Cell genetics
- Abstract
Genomic profiling of mantle cell lymphoma (MCL) cells has enabled a better understanding of the complex mechanisms underlying the pathogenesis of disease. Besides the t(11;14)(q13;q32) leading to cyclin D1 overexpression, MCL exhibits a characteristic pattern of DNA copy number aberrations that differs from those detected in other B-cell lymphomas. These genomic changes disrupt selected oncogenes and suppressor genes that are required for lymphoma development and progression, many of which are components of cell cycle, DNA damage response and repair, apoptosis, and cell-signaling pathways. Additionally, some of them may represent effective therapeutic targets. A number of genomic and molecular abnormalities have been correlated with the clinical outcome of patients with MCL and are considered prognostic factors. However, only a few genomic markers have been shown to predict the response to current or novel targeted therapies. One representative example is the high-level amplification of the BCL2 gene, which predicts a good response to pro-apoptotic BH3 mimetic drugs. In summary, genomic analyses have contributed to the substantial advances made in the comprehension of the pathogenesis of MCL, providing a solid basis for the identification of optimal therapeutic targets and for the design of new molecular therapies aiming to cure this fatal disease.
- Published
- 2013
- Full Text
- View/download PDF
44. MALT1 small molecule inhibitors specifically suppress ABC-DLBCL in vitro and in vivo.
- Author
-
Fontan L, Yang C, Kabaleeswaran V, Volpon L, Osborne MJ, Beltran E, Garcia M, Cerchietti L, Shaknovich R, Yang SN, Fang F, Gascoyne RD, Martinez-Climent JA, Glickman JF, Borden K, Wu H, and Melnick A
- Subjects
- Animals, B-Lymphocytes metabolism, Caspases metabolism, Catalysis, Cell Line, Tumor, Cell Proliferation drug effects, DNA-Binding Proteins antagonists & inhibitors, DNA-Binding Proteins metabolism, Down-Regulation drug effects, Humans, Lymphoma, Large B-Cell, Diffuse metabolism, Lymphoma, Large B-Cell, Diffuse pathology, Male, Mice, Mice, Inbred NOD, Mice, SCID, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, NF-kappa B metabolism, Neoplasm Proteins metabolism, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins metabolism, Proteolysis, Proto-Oncogene Proteins c-rel, Xenograft Model Antitumor Assays, B-Lymphocytes drug effects, Lymphoma, Large B-Cell, Diffuse drug therapy, Neoplasm Proteins antagonists & inhibitors, Protease Inhibitors pharmacology
- Abstract
MALT1 cleavage activity is linked to the pathogenesis of activated B cell-like diffuse large B cell lymphoma (ABC-DLBCL), a chemoresistant form of DLBCL. We developed a MALT1 activity assay and identified chemically diverse MALT1 inhibitors. A selected lead compound, MI-2, featured direct binding to MALT1 and suppression of its protease function. MI-2 concentrated within human ABC-DLBCL cells and irreversibly inhibited cleavage of MALT1 substrates. This was accompanied by NF-κB reporter activity suppression, c-REL nuclear localization inhibition, and NF-κB target gene downregulation. Most notably, MI-2 was nontoxic to mice, and displayed selective activity against ABC-DLBCL cell lines in vitro and xenotransplanted ABC-DLBCL tumors in vivo. The compound was also effective against primary human non-germinal center B cell-like DLBCLs ex vivo., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
45. High-throughput sequencing analysis of the chromosome 7q32 deletion reveals IRF5 as a potential tumour suppressor in splenic marginal-zone lymphoma.
- Author
-
Fresquet V, Robles EF, Parker A, Martinez-Useros J, Mena M, Malumbres R, Agirre X, Catarino S, Arteta D, Osaba L, Mollejo M, Hernandez-Rivas JM, Calasanz MJ, Daibata M, Dyer MJ, Prosper F, Vizcarra E, Piris MÁ, Oscier D, and Martinez-Climent JA
- Subjects
- Animals, Apoptosis genetics, Cell Division drug effects, Cell Line, Tumor transplantation, Chromosomes, Human, Pair 7 ultrastructure, Comparative Genomic Hybridization, Gene Expression Regulation, Neoplastic, Genes, Immunoglobulin, Humans, Interferon Regulatory Factors biosynthesis, Interferon Regulatory Factors deficiency, Interferon Regulatory Factors physiology, Kaplan-Meier Estimate, Lymphoma, B-Cell, Marginal Zone mortality, Lymphoma, B-Cell, Marginal Zone pathology, Mice, Mice, Knockout, Neoplasm Proteins biosynthesis, Neoplasm Proteins physiology, Point Mutation, Real-Time Polymerase Chain Reaction, Splenic Neoplasms mortality, Splenic Neoplasms pathology, Translocation, Genetic, Chromosomes, Human, Pair 7 genetics, Genes, Tumor Suppressor, Genetic Association Studies, High-Throughput Nucleotide Sequencing, Interferon Regulatory Factors genetics, Lymphoma, B-Cell, Marginal Zone genetics, Neoplasm Proteins genetics, Oligonucleotide Array Sequence Analysis methods, Sequence Deletion, Splenic Neoplasms genetics
- Abstract
Using high-resolution genomic microarray analysis, a distinct genomic profile was defined in 114 samples from patients with splenic marginal zone lymphoma (SMZL). Deletion or uniparental disomy of chromosome 7q were detected in 42 of 114 (37%) SMZLs but in only nine of 170 (5%) mature B-cell lymphomas (P < 0·00001). The presence of unmutated IGHV, genomic complexity, 17p13-TP53 deletion and 8q-MYC gain, but not 7q deletion, correlated with shorter overall survival of SMZL patients. Mapping studies narrowed down a commonly deleted region of 2·7 Mb in 7q32.1-q32.2 spanning a region between the SND1 and COPG2 genes. High-throughput sequencing analysis of the 7q32-deleted segment did not identify biallelic deletions/insertions or clear pathogenic gene mutations, but detected six nucleotide changes in IRF5 (n = 2), TMEM209 (n = 2), CALU (n = 1) and ZC3HC1 (n = 1) not found in healthy individuals. Comparative expression analysis found a fourfold down-regulation of IRF5 gene in lymphomas with 7q32 deletion versus non-deleted tumours (P = 0·032). Ectopic expression of IRF5 in marginal-zone lymphoma cells decreased proliferation and increased apoptosis in vitro, and impaired lymphoma development in vivo. These results show that cryptic deletions, insertions and/or point mutations inactivating genes within 7q32 are not common in SMZL, and suggest that IRF5 may be a haploinsufficient tumour suppressor in this lymphoma entity., (© 2012 Blackwell Publishing Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
46. Preclinical activity of LBH589 alone or in combination with chemotherapy in a xenogeneic mouse model of human acute lymphoblastic leukemia.
- Author
-
Vilas-Zornoza A, Agirre X, Abizanda G, Moreno C, Segura V, De Martino Rodriguez A, José-Eneriz ES, Miranda E, Martín-Subero JI, Garate L, Blanco-Prieto MJ, García de Jalón JA, Rio P, Rifón J, Cigudosa JC, Martinez-Climent JA, Román-Gómez J, Calasanz MJ, Ribera JM, and Prósper F
- Subjects
- Animals, Antineoplastic Agents, Hormonal pharmacology, Antineoplastic Agents, Phytogenic pharmacology, Antineoplastic Combined Chemotherapy Protocols, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Proliferation drug effects, DNA Methylation, DNA-Binding Proteins physiology, Drug Synergism, Female, Gene Expression Profiling, Histones metabolism, Humans, Immunoenzyme Techniques, Indoles, Mice, Mice, Inbred BALB C, Oligonucleotide Array Sequence Analysis, Panobinostat, Polymorphism, Single Nucleotide genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Apoptosis drug effects, Dexamethasone pharmacology, Histone Deacetylase Inhibitors pharmacology, Hydroxamic Acids pharmacology, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Vincristine pharmacology
- Abstract
Histone deacetylases (HDACs) have been identified as therapeutic targets due to their regulatory function in chromatin structure and organization. Here, we analyzed the therapeutic effect of LBH589, a class I-II HDAC inhibitor, in acute lymphoblastic leukemia (ALL). In vitro, LBH589 induced dose-dependent antiproliferative and apoptotic effects, which were associated with increased H3 and H4 histone acetylation. Intravenous administration of LBH589 in immunodeficient BALB/c-RAG2(-/-)γc(-/-) mice in which human-derived T and B-ALL cell lines were injected induced a significant reduction in tumor growth. Using primary ALL cells, a xenograft model of human leukemia in BALB/c-RAG2(-/-)γc(-/-) mice was established, allowing continuous passages of transplanted cells to several mouse generations. Treatment of mice engrafted with T or B-ALL cells with LBH589 induced an in vivo increase in the acetylation of H3 and H4, which was accompanied with prolonged survival of LBH589-treated mice in comparison with those receiving vincristine and dexamethasone. Notably, the therapeutic efficacy of LBH589 was significantly enhanced in combination with vincristine and dexamethasone. Our results show the therapeutic activity of LBH589 in combination with standard chemotherapy in pre-clinical models of ALL and suggest that this combination may be of clinical value in the treatment of patients with ALL.
- Published
- 2012
- Full Text
- View/download PDF
47. Expression of MALT1 oncogene in hematopoietic stem/progenitor cells recapitulates the pathogenesis of human lymphoma in mice.
- Author
-
Vicente-Dueñas C, Fontán L, Gonzalez-Herrero I, Romero-Camarero I, Segura V, Aznar MA, Alonso-Escudero E, Campos-Sanchez E, Ruiz-Roca L, Barajas-Diego M, Sagardoy A, Martinez-Ferrandis JI, Abollo-Jimenez F, Bertolo C, Peñuelas I, Garcia-Criado FJ, García-Cenador MB, Tousseyn T, Agirre X, Prosper F, Garcia-Bragado F, McPhail ED, Lossos IS, Du MQ, Flores T, Hernandez-Rivas JM, Gonzalez M, Salar A, Bellosillo B, Conde E, Siebert R, Sagaert X, Cobaleda C, Sanchez-Garcia I, and Martinez-Climent JA
- Subjects
- Animals, Humans, Mice, Mice, Transgenic, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, NF-kappa B metabolism, Transcription, Genetic, Caspases genetics, Hematopoietic Stem Cells metabolism, Lymphoma pathology, Neoplasm Proteins genetics, Oncogenes
- Abstract
Chromosomal translocations involving the MALT1 gene are hallmarks of mucosa-associated lymphoid tissue (MALT) lymphoma. To date, targeting these translocations to mouse B cells has failed to reproduce human disease. Here, we induced MALT1 expression in mouse Sca1(+)Lin(-) hematopoietic stem/progenitor cells, which showed NF-κB activation and early lymphoid priming, being selectively skewed toward B-cell differentiation. These cells accumulated in extranodal tissues and gave rise to clonal tumors recapitulating the principal clinical, biological, and molecular genetic features of MALT lymphoma. Deletion of p53 gene accelerated tumor onset and induced transformation of MALT lymphoma to activated B-cell diffuse large-cell lymphoma (ABC-DLBCL). Treatment of MALT1-induced lymphomas with a specific inhibitor of MALT1 proteolytic activity decreased cell viability, indicating that endogenous Malt1 signaling was required for tumor cell survival. Our study shows that human-like lymphomas can be modeled in mice by targeting MALT1 expression to hematopoietic stem/progenitor cells, demonstrating the oncogenic role of MALT1 in lymphomagenesis. Furthermore, this work establishes a molecular link between MALT lymphoma and ABC-DLBCL, and provides mouse models to test MALT1 inhibitors. Finally, our results suggest that hematopoietic stem/progenitor cells may be involved in the pathogenesis of human mature B-cell lymphomas.
- Published
- 2012
- Full Text
- View/download PDF
48. PIM2 inhibition as a rational therapeutic approach in B-cell lymphoma.
- Author
-
Gómez-Abad C, Pisonero H, Blanco-Aparicio C, Roncador G, González-Menchén A, Martinez-Climent JA, Mata E, Rodríguez ME, Muñoz-González G, Sánchez-Beato M, Leal JF, Bischoff JR, and Piris MA
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Apoptosis drug effects, Apoptosis physiology, Biomarkers, Tumor metabolism, Cell Cycle Proteins, Cell Line, Tumor, Gene Expression Regulation, Neoplastic genetics, Humans, Lymph Nodes pathology, Lymphoma, Follicular genetics, Lymphoma, Follicular pathology, Lymphoma, Follicular therapy, Lymphoma, Large B-Cell, Diffuse pathology, Lymphoma, Mantle-Cell genetics, Lymphoma, Mantle-Cell pathology, Lymphoma, Mantle-Cell therapy, Palatine Tonsil pathology, Phosphoproteins metabolism, Proto-Oncogene Proteins c-akt metabolism, RNA, Small Interfering genetics, RNA, Small Interfering pharmacology, bcl-Associated Death Protein metabolism, Enzyme Inhibitors pharmacology, Genetic Therapy methods, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics
- Abstract
PIM serine/threonine kinases are overexpressed, translocated, or amplified in multiple B-cell lymphoma types. We have explored the frequency and relevance of PIM expression in different B-cell lymphoma types and investigated whether PIM inhibition could be a rational therapeutic approach. Increased expression of PIM2 was detected in subsets of mantle cell lymphoma, diffuse large B-cell lymphoma (DLBLC), follicular lymphoma, marginal zone lymphoma-mucosa-associated lymphoid tissue type, chronic lymphocytic leukemia, and nodal marginal zone lymphoma cases. Increased PIM2 protein expression was associated with an aggressive clinical course in activated B-like-DLBCL patients. Pharmacologic and genetic inhibition of PIM2 revealed p4E-BP1(Thr37/46) and p4E-BP1(Ser65) as molecular biomarkers characteristic of PIM2 activity and indicated the involvement of PIM2 kinase in regulating mammalian target of rapamycin complex 1. The simultaneous genetic inhibition of all 3 PIM kinases induced changes in apoptosis and cell cycle. In conclusion, we show that PIM2 kinase inhibition is a rational approach in DLBCL treatment, identify appropriate biomarkers for pharmacodynamic studies, and provide a new marker for patient stratification.
- Published
- 2011
- Full Text
- View/download PDF
49. Blockade of the NFκB pathway drives differentiating glioblastoma-initiating cells into senescence both in vitro and in vivo.
- Author
-
Nogueira L, Ruiz-Ontañon P, Vazquez-Barquero A, Lafarga M, Berciano MT, Aldaz B, Grande L, Casafont I, Segura V, Robles EF, Suarez D, Garcia LF, Martinez-Climent JA, and Fernandez-Luna JL
- Subjects
- Animals, Blotting, Western, Carbazoles pharmacology, Cell Differentiation genetics, Cell Proliferation drug effects, Cellular Senescence drug effects, Cyclin D1 genetics, Cyclin D1 metabolism, Female, Gene Expression Profiling, Glioblastoma drug therapy, Glioblastoma pathology, Glycosides pharmacology, Humans, I-kappa B Kinase antagonists & inhibitors, I-kappa B Kinase genetics, I-kappa B Kinase metabolism, I-kappa B Proteins genetics, I-kappa B Proteins metabolism, Mice, Mice, Inbred BALB C, Mice, Knockout, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, Neural Stem Cells drug effects, Neural Stem Cells metabolism, Neural Stem Cells pathology, Nitriles pharmacology, Oligonucleotide Array Sequence Analysis, Protein Kinase Inhibitors pharmacology, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction drug effects, Sulfones pharmacology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Cellular Senescence genetics, Glioblastoma genetics, NF-kappa B genetics, Signal Transduction genetics
- Abstract
Glioblastoma multiforme is one of the most devastating cancers and presents unique challenges to therapy because of its aggressive behavior. Cancer-initiating or progenitor cells have been described to be the only cell population with tumorigenic capacity in glioblastoma. Therefore, effective therapeutic strategies targeting these cells or the early precursors may be beneficial. We have established different cultures of glioblastoma-initiating cells (GICs) derived from surgical specimens and found that, after induction of differentiation, the NFκB transcriptional pathway was activated, as determined by analyzing key proteins such as p65 and IκB and the upregulation of a number of target genes. We also showed that blockade of nuclear factor (NF)κB signaling in differentiating GICs by different genetic strategies or treatment with small-molecule inhibitors, promoted replication arrest and senescence. This effect was partly mediated by reduced levels of the NFκB target gene cyclin D1, because its downregulation by RNA interference reproduced a similar phenotype. Furthermore, these results were confirmed in a xenograft model. Intravenous treatment of immunodeficient mice bearing human GIC-derived tumors with a novel small-molecule inhibitor of the NFκB pathway induced senescence of tumor cells but no ultrastructural alterations of the brain parenchyma were detected. These findings reveal that activation of NFκB may keep differentiating GICs from acquiring a mature postmitotic phenotype, thus allowing cell proliferation, and support the rationale for therapeutic strategies aimed to promote premature senescence of differentiating GICs by blocking key factors within the NFκB pathway.
- Published
- 2011
- Full Text
- View/download PDF
50. A cyclin-D1 interaction with BAX underlies its oncogenic role and potential as a therapeutic target in mantle cell lymphoma.
- Author
-
Beltran E, Fresquet V, Martinez-Useros J, Richter-Larrea JA, Sagardoy A, Sesma I, Almada LL, Montes-Moreno S, Siebert R, Gesk S, Calasanz MJ, Malumbres R, Rieger M, Prosper F, Lossos IS, Piris MA, Fernandez-Zapico ME, and Martinez-Climent JA
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Biphenyl Compounds pharmacology, Cell Cycle, Cell Line, Tumor, Cell Survival, Cyclin D1 antagonists & inhibitors, Cyclin D1 genetics, Disease Models, Animal, Gene Amplification, Genes, bcl-2, Humans, Lymphoma, Mantle-Cell etiology, Lymphoma, Mantle-Cell pathology, Lymphoma, Mantle-Cell therapy, Mice, Nitrophenols pharmacology, Piperazines pharmacology, Sulfonamides pharmacology, Xenograft Model Antitumor Assays, bcl-2-Associated X Protein antagonists & inhibitors, Cyclin D1 metabolism, Lymphoma, Mantle-Cell metabolism, bcl-2-Associated X Protein metabolism
- Abstract
The chromosomal translocation t(11;14)(q13;q32) leading to cyclin-D1 overexpression plays an essential role in the development of mantle cell lymphoma (MCL), an aggressive tumor that remains incurable with current treatment strategies. Cyclin-D1 has been postulated as an effective therapeutic target, but the evaluation of this target has been hampered by our incomplete understanding of its oncogenic functions and by the lack of valid MCL murine models. To address these issues, we generated a cyclin-D1-driven mouse model in which cyclin-D1 expression can be regulated externally. These mice developed cyclin-D1-expressing lymphomas capable of recapitulating features of human MCL. We found that cyclin-D1 inactivation was not sufficient to induce lymphoma regression in vivo; however, using a combination of in vitro and in vivo assays, we identified a novel prosurvival cyclin-D1 function in MCL cells. Specifically, we found that cyclin-D1, besides increasing cell proliferation through deregulation of the cell cycle at the G(1)-S transition, sequestrates the proapoptotic protein BAX in the cytoplasm, thereby favoring BCL2's antiapoptotic function. Accordingly, cyclin-D1 inhibition sensitized the lymphoma cells to apoptosis through BAX release. Thus, genetic or pharmacologic targeting of cyclin-D1 combined with a proapoptotic BH3 mimetic synergistically killed the cyclin-D1-expressing murine lymphomas, human MCL cell lines, and primary lymphoma cells. Our study identifies a role of cyclin-D1 in deregulating apoptosis in MCL cells, and highlights the potential benefit of simultaneously targeting cyclin-D1 and survival pathways in patients with MCL. This effective combination therapy also might be exploited in other cyclin-D1-expressing tumors.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.