Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Clin Immunol ; 263: 110203, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38575046

ABSTRACT

Langerhans cell histiocytosis (LCH) is characterized by an expansion and accumulation of pathological histiocytes expressing langerin (CD207) and CD1a in different organs under an inflammatory milieu. The origin of pathognomonic precursors of LCH is widely debated, but monocytes and pre-dendritic cells (pre-DC) play a significant role. Remarkably, we found an expansion of AXLhigh cells in the CD11c+ subset of patients with active LCH, which also express the pathognomonic CD207 and CD1a. Moreover, we obtained a monocyte-derived LC-like (mo-LC-like) expressing high levels of AXL when treated with inflammatory cytokine, or plasma of patients with active disease. Intriguingly, inhibiting the mTOR pathway at the initial stages of monocyte differentiation to LC-like fosters the pathognomonic LCH program, highly increasing CD207 levels, together with NOTCH1 induction. We define here that AXLhigh could also be taken as a strong pathognomonic marker for LCH, and the release of Langerin and NOTCH1 expression depends on the inhibition of the mTOR pathway.


Subject(s)
Antigens, CD , Axl Receptor Tyrosine Kinase , Histiocytosis, Langerhans-Cell , Lectins, C-Type , Mannose-Binding Lectins , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , TOR Serine-Threonine Kinases , Humans , Histiocytosis, Langerhans-Cell/metabolism , TOR Serine-Threonine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Antigens, CD/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Mannose-Binding Lectins/metabolism , Lectins, C-Type/metabolism , Male , Myeloid Cells/metabolism , Biomarkers , Female , Adolescent , Receptor, Notch1/metabolism , Antigens, CD1/metabolism , Child , Monocytes/metabolism , Monocytes/immunology , Adult , Child, Preschool , Signal Transduction , Cell Differentiation
3.
Proc Natl Acad Sci U S A ; 119(51): e2211775119, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36508676

ABSTRACT

Synthetic lethality is a powerful approach for targeting oncogenic drivers in cancer. Recent studies revealed that cancer cells with microsatellite instability (MSI) require Werner (WRN) helicase for survival; however, the underlying mechanism remains unclear. In this study, we found that WRN depletion strongly induced p53 and its downstream apoptotic target PUMA in MSI colorectal cancer (CRC) cells. p53 or PUMA deletion abolished apoptosis induced by WRN depletion in MSI CRC cells. Importantly, correction of MSI abrogated the activation of p53/PUMA and cell killing, while induction of MSI led to sensitivity in isogenic CRC cells. Rare p53-mutant MSI CRC cells are resistant to WRN depletion due to lack of PUMA induction, which could be restored by wildtype (WT) p53 knock in or reconstitution. WRN depletion or treatment with the RecQ helicase inhibitor ML216 suppressed in vitro and in vivo growth of MSI CRCs in a p53/PUMA-dependent manner. ML216 treatment was efficacious in MSI CRC patient-derived xenografts. Interestingly, p53 gene remains WT in the majority of MSI CRCs. These results indicate a critical role of p53/PUMA-mediated apoptosis in the vulnerability of MSI CRCs to WRN loss, and support WRN as a promising therapeutic target in p53-WT MSI CRCs.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Humans , Werner Syndrome Helicase/genetics , Tumor Suppressor Protein p53/genetics , Microsatellite Instability , Colorectal Neoplasms/genetics , RecQ Helicases/genetics
4.
Cancer Res ; 82(7): 1340-1352, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35149588

ABSTRACT

Targeting cyclin-dependent kinases 4 and 6 (CDK4/6) is a successful therapeutic approach against breast and other solid tumors. Inhibition of CDK4/6 halts cell cycle progression and promotes antitumor immunity. However, the mechanisms underlying the antitumor activity of CDK4/6 inhibitors are not fully understood. We found that CDK4/6 bind and phosphorylate the p53 family member p73 at threonine 86, which sequesters p73 in the cytoplasm. Inhibition of CDK4/6 led to dephosphorylation and nuclear translocation of p73, which transcriptionally activated death receptor 5 (DR5), a cytokine receptor and key component of the extrinsic apoptotic pathway. p73-mediated induction of DR5 by CDK4/6 inhibitors promoted immunogenic cell death of cancer cells. Deletion of DR5 in cancer cells in vitro and in vivo abrogated the potentiating effects of CDK4/6 inhibitors on immune cytokine TRAIL, 5-fluorouracil chemotherapy, and anti-PD-1 immunotherapy. Together, these results reveal a previously unrecognized consequence of CDK4/6 inhibition, which may be critical for potentiating the killing and immunogenic effects on cancer cells. SIGNIFICANCE: This work demonstrates how inhibition of CDK4/6 sensitizes cancer cells to chemotherapy and immune checkpoint blockade and may provide a new molecular marker for improving CDK4/6-targeted cancer therapies. See related commentary by Frank, p. 1170.


Subject(s)
Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Immune Checkpoint Inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand , TNF-Related Apoptosis-Inducing Ligand , Tumor Protein p73 , Apoptosis , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Fluorouracil/pharmacology , Humans , Phosphorylation , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Protein p73/metabolism
5.
Oncogene ; 40(48): 6566-6578, 2021 12.
Article in English | MEDLINE | ID: mdl-34615996

ABSTRACT

Bromodomain and extra-terminal domain (BET) family proteins are epigenetic readers that play a critical role in oncogenesis by controlling the expression of oncogenes such as c-Myc. Targeting BET family proteins has recently emerged as a promising anticancer strategy. However, the molecular mechanisms by which cancer cells respond to BET inhibition are not well understood. In this study, we found that inducing the degradation of BET proteins by the proteolysis targeting chimeras (PROTAC) approach potently suppressed the growth of colorectal cancer (CRC) including patient-derived tumors. Mechanistically, BET degradation transcriptionally activates Death Receptor 5 (DR5) to trigger immunogenic cell death (ICD) in CRC cells. Enhanced DR5 induction further sensitizes CRC cells with a mutation in Speckle-type POZ protein (SPOP). Furthermore, DR5 is indispensable for a striking antitumor effect of combining BET degradation and anti-PD-1 antibody, which was well tolerated in mice and almost eradicated syngeneic tumors. Our results demonstrate that BET degradation triggers DR5-mediated ICD to potently suppress CRC and potentiate immune checkpoint blockade. These results provide a rationale, mechanistic insights, and potential biomarkers for developing a precision CRC therapy by inducing BET protein degradation.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Immune Checkpoint Inhibitors/pharmacology , Immunogenic Cell Death , Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Proteins/genetics , Proteolysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Oncogene ; 40(11): 2035-2050, 2021 03.
Article in English | MEDLINE | ID: mdl-33603166

ABSTRACT

Use of non-steroidal anti-inflammatory drugs (NSAIDs) is associated with reduced risk of colorectal cancer (CRC). However, the mechanism by which NSAIDs suppress colorectal tumorigenesis remains unclear. We previously showed that NSAIDs selectively kill emerging tumor cells via death receptor (DR) signaling and a synthetic lethal interaction mediated by the proapoptotic Bcl-2 family protein BID. In this study, we found NSAIDs induce endoplasmic reticulum (ER) stress to activate DR signaling and BID in tumor suppression. Importantly, our results unveiled an ER stress- and BID-dependent immunogenic effect of NSAIDs, which may be critical for tumor suppression. NSAID treatment induced hallmarks of immunogenic cell death (ICD) in CRC cells and colonic epithelial cells upon loss of APC tumor suppressor, and elevated tumor-infiltrating lymphocytes (TILs) in the polyps of APCMin/+ mice. ER stress inhibition or BID deletion abrogated the antitumor and immunogenic effects of NSAIDs. Furthermore, increased ER stress and TILs were detected in human advanced adenomas from NSAID-treated patients. Together, our results suggest that NSAIDs induce ER stress- and BID-mediated ICD to restore immunosurveillance and suppress colorectal tumor formation.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carcinogenesis/genetics , Colorectal Neoplasms/drug therapy , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Stress/drug effects , Humans , Immunogenic Cell Death/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Signal Transduction/drug effects
8.
Leuk Lymphoma ; 61(10): 2409-2418, 2020 10.
Article in English | MEDLINE | ID: mdl-32306816

ABSTRACT

Ibrutinib is a BTK/ITK inhibitor with efficacy for the treatment of various lymphoid cancers, including CLL. Considering that innate and adaptative immune defects are a dominant feature of CLL patients, we evaluated whether in vitro ibrutinib affects the survival and function of neutrophils and γδ T cells, key players of the early immune response against microbes. Neutrophils and γδ T cells were obtained from peripheral blood of healthy donors and CLL patients. We found that ibrutinib reduces the production of reactive oxygen species (ROS) and bacteria killing capacity, and slightly impairs neutrophil extracellular traps (NETs) production without affecting bacteria-uptake and CD62L-downregulation induced by fMLP or aggregated IgG. In addition, ibrutinib reduces γδ T cell activation and CD107a degranulation induced by phosphoantigens or anti-CD3. These findings are in agreement with previous data suggesting that ibrutinib interferes with the protective immune response to pathogens, particularly Mycobacteria and Aspergillus.


Subject(s)
Neutrophils , T-Lymphocytes , Adenine/analogs & derivatives , Humans , Lymphocyte Activation , Piperidines , Reactive Oxygen Species
9.
Cancer Immunol Immunother ; 69(5): 813-824, 2020 May.
Article in English | MEDLINE | ID: mdl-32055920

ABSTRACT

Despite significant therapeutic improvements chronic lymphocytic leukemia (CLL) remains an incurable disease and there is a persistent pursuit of new treatment alternatives. Lurbinectedin, a selective inhibitor of active transcription of protein-coding genes, is currently in phase II/III clinical trials for solid tumors such as small-cell lung cancer (SCLC). In this study, we aimed to evaluate the activity of Lurbinectedin on circulating mononuclear cells from CLL patients and to determine whether Lurbinectedin could affect the cross-talk between B-CLL cells and the tumor microenvironment. We found that Lurbinectedin induced a dose- and time-dependent death in all cell types evaluated, with B cells, monocytes and monocytic myeloid derived suppressor cells (Mo-MDSC) being the most susceptible populations. At sub-apoptotic doses, Lurbinectedin decreased the expression of CCR7 in B-CLL cells and impaired their migration towards CCL19 and CCL21. Furthermore, low concentrations of Lurbinectedin stimulated the synthesis of pro-IL1ß in monocytes and nurse-like cells, without inducing the inflammasome activation. Altogether, these results indicate that Lurbinectedin might have antitumor activity in CLL due to its direct action on leukemic cells in combination with its effects on the tumor microenvironment. Our findings encourage further investigation of Lurbinectedin as a potential therapy for CLL.


Subject(s)
Carbolines/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Tumor Microenvironment/drug effects , Apoptosis/drug effects , Apoptosis/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Survival/drug effects , Cell Survival/immunology , Chemokine CCL19/immunology , Chemokine CCL19/metabolism , Chemokine CCL21/immunology , Chemokine CCL21/metabolism , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Primary Cell Culture , Receptors, CCR7/immunology , Receptors, CCR7/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/immunology
11.
Int J Cancer ; 144(5): 1128-1134, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30178523

ABSTRACT

Reprogramming of neutrophils by malignant cells is well-described for many types of solid tumors, but data remain scarce for hematological diseases. Chronic lymphocytic leukemia (CLL) is characterized for a deep immune dysregulation mediated by leukemic cells that compromises patient's outcome. Murine models of CLL highlight the relevance of myeloid cells as tumor-driven reprogramming targets. In our study, we evaluated neutrophil reprogramming by CLL cells. We first show that the proportion of the CD16high CD62Ldim neutrophil subset in peripheral blood of CLL patients is increased compared to age-matched healthy donors (HD). In vitro, neutrophils from HD cultured in the presence of CLL cells or conditioned media (CM) from CLL cells exhibited a longer lifespan. Depletion of G-CSF and GM-CSF from CM partially reversed the protective effect. In addition, the proportion of viable neutrophils that displayed a CD16high CD62Ldim phenotype was increased in the presence of CM from CLL cells, being TGF-ß/IL-10 responsible for this effect. Altogether, our results describe a novel mechanism through which CLL cells can manipulate neutrophils.


Subject(s)
Cell Differentiation/physiology , Immune Tolerance/physiology , L-Selectin/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neutrophils/pathology , Receptors, IgG/metabolism , Aged , Cell Line, Tumor , Female , GPI-Linked Proteins/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Male , Middle Aged , Neutrophils/metabolism , Transforming Growth Factor beta/metabolism
13.
Haematologica ; 103(10): e458-e461, 2018 10.
Article in English | MEDLINE | ID: mdl-29748439
14.
Sci Rep ; 7(1): 15714, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29146966

ABSTRACT

The proliferation and survival of malignant B cells in chronic lymphocytic leukemia (CLL) depend on signals from the microenvironment in lymphoid tissues. Among a plethora of soluble factors, IL-8 has been considered one of the most relevant to support CLL B cell progression in an autocrine fashion, even though the expression of IL-8 receptors, CXCR1 and CXCR2, on leukemic B cells has not been reported. Here we show that circulating CLL B cells neither express CXCR1 or CXCR2 nor they respond to exogenous IL-8 when cultured in vitro alone or in the presence of monocytes/nurse-like cells. By intracellular staining and ELISA we show that highly purified CLL B cells do not produce IL-8 spontaneously or upon activation through the B cell receptor. By contrast, we found that a minor proportion (<0.5%) of contaminating monocytes in enriched suspensions of leukemic cells might be the actual source of IL-8 due to their strong capacity to release this cytokine. Altogether our results indicate that CLL B cells are not able to secrete or respond to IL-8 and highlight the importance of methodological details in in vitro experiments.


Subject(s)
Interleukin-8/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Aged , Aged, 80 and over , Apoptosis , Cell Line, Tumor , Cell Survival , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Monocytes/metabolism , Receptors, Interleukin-8/metabolism
16.
Cancer Immunol Immunother ; 66(4): 461-473, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28011996

ABSTRACT

Small molecules targeting kinases involved in B cell receptor signaling are showing encouraging clinical activity in chronic lymphocytic leukemia (CLL) patients. Fostamatinib (R406) and entospletinib (GS-9973) are ATP-competitive inhibitors designed to target spleen tyrosine kinase (Syk) that have shown clinical activity with acceptable toxicity in trials with CLL patients. Preclinical studies with these inhibitors in CLL have focused on their effect in patient-derived leukemic B cells. In this work we show that clinically relevant doses of R406 and GS-9973 impaired the activation and proliferation of T cells from CLL patients. This effect could not be ascribed to Syk-inhibition given that we show that T cells from CLL patients do not express Syk protein. Interestingly, ζ-chain-associated protein kinase (ZAP)-70 phosphorylation was diminished by both inhibitors upon TCR stimulation on T cells. In addition, we found that both agents reduced macrophage-mediated phagocytosis of rituximab-coated CLL cells. Overall, these results suggest that in CLL patients treated with R406 or GS-9973 T cell functions, as well as macrophage-mediated anti-tumor activity of rituximab, might be impaired. The potential consequences for CLL-treated patients are discussed.


Subject(s)
Indazoles/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Macrophages/immunology , Oxazines/pharmacology , Pyrazines/pharmacology , Pyridines/pharmacology , Syk Kinase/antagonists & inhibitors , T-Lymphocytes/drug effects , ZAP-70 Protein-Tyrosine Kinase/metabolism , Aged , Aged, 80 and over , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Lymphocyte Activation/drug effects , Male , Middle Aged , Phagocytosis/drug effects , Phosphorylation/drug effects , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Rituximab/pharmacology , T-Lymphocytes/immunology
17.
Cancer Immunol Immunother ; 66(1): 77-89, 2017 01.
Article in English | MEDLINE | ID: mdl-27796477

ABSTRACT

Chronic lymphocytic leukemia (CLL) is characterized by immune defects that contribute to a high rate of infections and autoimmune cytopenias. Neutrophils are the first line of innate immunity and respond to pathogens through multiple mechanisms, including the release of neutrophil extracellular traps (NETs). These web-like structures composed of DNA, histones, and granular proteins are also produced under sterile conditions and play important roles in thrombosis and autoimmune disorders. Here we show that neutrophils from CLL patients are more prone to release NETs compared to those from age-matched healthy donors (HD). Increased generation of NETs was not due to higher levels of elastase, myeloperoxidase, or reactive oxygen species production. Instead, we found that plasma from CLL patients was able to prime neutrophils from HD to generate higher amounts of NETs upon activation. Plasmatic IL-8 was involved in the priming effect since its depletion reduced plasma capacity to enhance NETs release. Finally, we found that culture with NETs delayed spontaneous apoptosis and increased the expression of activation markers on leukemic B cells. Our study provides new insights into the immune dysregulation in CLL and suggests that the chronic inflammatory environment typical of CLL probably underlies this inappropriate neutrophil priming.


Subject(s)
Extracellular Traps/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Neutrophils/immunology , Aged , Aged, 80 and over , Case-Control Studies , Humans , Interleukin-8/immunology , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...