Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(9): eadj4698, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38427724

ABSTRACT

Cancers exploit coinhibitory receptors on T cells to escape tumor immunity, and targeting such mechanisms has shown remarkable clinical benefit, but in a limited subset of patients. We hypothesized that cancer cells mimic noncanonical mechanisms of early development such as axon guidance pathways to evade T cell immunity. Using gain-of-function genetic screens, we profiled axon guidance proteins on human T cells and their cognate ligands and identified fibronectin leucine-rich transmembrane protein 3 (FLRT3) as a ligand that inhibits T cell activity. We demonstrated that FLRT3 inhibits T cells through UNC5B, an axon guidance receptor that is up-regulated on activated human T cells. FLRT3 expressed in human cancers favored tumor growth and inhibited CAR-T and BiTE + T cell killing and infiltration in humanized cancer models. An FLRT3 monoclonal antibody that blocked FLRT3-UNC5B interactions reversed these effects in an immune-dependent manner. This study supports the concept that axon guidance proteins mimic T cell checkpoints and can be targeted for cancer immunotherapy.


Subject(s)
Neoplasms , T-Lymphocytes , Humans , Neoplasms/genetics , Neoplasms/therapy , Immunotherapy , Membrane Glycoproteins , Netrin Receptors
2.
J Clin Invest ; 133(22)2023 11 15.
Article in English | MEDLINE | ID: mdl-37966113

ABSTRACT

Effective eradication of leukemic stem cells (LSCs) remains the greatest challenge in treating acute myeloid leukemia (AML). The immune receptor LAIR-1 has been shown to regulate LSC survival; however, the therapeutic potential of this pathway remains unexplored. We developed a therapeutic LAIR-1 agonist antibody, NC525, that induced cell death of LSCs, but not healthy hematopoietic stem cells in vitro, and killed LSCs and AML blasts in both cell- and patient-derived xenograft models. We showed that LAIR-1 agonism drives a unique apoptotic signaling program in leukemic cells that was enhanced in the presence of collagen. NC525 also significantly improved the activity of azacitidine and venetoclax to establish LAIR-1 targeting as a therapeutic strategy for AML that may synergize with standard-of-care therapies.


Subject(s)
Leukemia, Myeloid, Acute , Animals , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Hematopoietic Stem Cells/metabolism , Signal Transduction , Disease Models, Animal , Neoplastic Stem Cells/metabolism
3.
J Genet Genomics ; 48(3): 184-197, 2021 03 20.
Article in English | MEDLINE | ID: mdl-33840602

ABSTRACT

In eukaryotic genome biology, the genomic organization inside the three-dimensional (3D) nucleus is highly complex, and whether this organization governs gene expression is poorly understood. Nuclear lamina (NL) is a filamentous meshwork of proteins present at the lining of inner nuclear membrane that serves as an anchoring platform for genome organization. Large chromatin domains termed as lamina-associated domains (LADs), play a major role in silencing genes at the nuclear periphery. The interaction of the NL and genome is dynamic and stochastic. Furthermore, many genes change their positions during developmental processes or under disease conditions such as cancer, to activate certain sorts of genes and/or silence others. Pericentromeric heterochromatin (PCH) is mostly in the silenced region within the genome, which localizes at the nuclear periphery. Studies show that several genes located at the PCH are aberrantly expressed in cancer. The interesting question is that despite being localized in the pericentromeric region, how these genes still manage to overcome pericentromeric repression. Although epigenetic mechanisms control the expression of the pericentromeric region, recent studies about genome organization and genome-nuclear lamina interaction have shed light on a new aspect of pericentromeric gene regulation through a complex and coordinated interplay between epigenomic remodeling and genomic organization in cancer.


Subject(s)
Epigenomics , Nuclear Lamina , Cell Nucleus , Gene Expression Regulation , Heterochromatin , Humans
4.
Nat Immunol ; 22(1): 53-66, 2021 01.
Article in English | MEDLINE | ID: mdl-33230330

ABSTRACT

Regenerative stem cell-like memory (TSCM) CD8+ T cells persist longer and produce stronger effector functions. We found that MEK1/2 inhibition (MEKi) induces TSCM that have naive phenotype with self-renewability, enhanced multipotency and proliferative capacity. This is achieved by delaying cell division and enhancing mitochondrial biogenesis and fatty acid oxidation, without affecting T cell receptor-mediated activation. DNA methylation profiling revealed that MEKi-induced TSCM cells exhibited plasticity and loci-specific profiles similar to bona fide TSCM isolated from healthy donors, with intermediate characteristics compared to naive and central memory T cells. Ex vivo, antigenic rechallenge of MEKi-treated CD8+ T cells showed stronger recall responses. This strategy generated T cells with higher efficacy for adoptive cell therapy. Moreover, MEKi treatment of tumor-bearing mice also showed strong immune-mediated antitumor effects. In conclusion, we show that MEKi leads to CD8+ T cell reprogramming into TSCM that acts as a reservoir for effector T cells with potent therapeutic characteristics.


Subject(s)
Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Immunologic Memory/drug effects , Immunotherapy, Adoptive , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasms/therapy , Stem Cells/cytology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle/drug effects , Humans , Immunologic Memory/immunology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Receptors, Antigen, T-Cell/physiology , Tumor Microenvironment
5.
Sci Rep ; 9(1): 17213, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748606

ABSTRACT

Transgenic mice that express either a NUP98-PHF23 (NP23) or NUP98-HOXD13 (NHD13) fusion in the hematopoietic compartment develop a wide spectrum of leukemias, including myeloid, erythroid, megakaryocytic and lymphoid, at age 9-14 months. NP23-NHD13 double transgenic mice were generated by interbreeding NP23 and NHD13 mice. Remarkably, 100% of the NP23-NHD13 double transgenic mice developed acute myeloid leukemia (AML) within three months, characterized by replacement of the thymus with leukemic myeloblasts. The marked infiltration of thymus led to the intriguing hypothesis that AML generated in NP23-NHD13 mice arose in the thymus, as opposed to the bone marrow (BM). Transplantation of CD4-CD8- double negative (DN) thymocytes (which were also negative for Mac1 and Gr1) from leukemic NHD13/NP23 mice demonstrated that DN thymocytes could transmit AML, and limiting dilution studies showed that leukemia initiating cells were increased 14-fold in the thymus compared to BM. Further thymocyte fractionation demonstrated that DN1 and DN2, but not DN3 or DN4 fractions transmitted AML, and a marked expansion (100-fold) of Lineage-Sca1 + Kit + (LSK) cells in the thymus of the NP23-NHD13 mice. Taken together, these results show that the thymus of NP23-NHD13 mice acts as a reservoir for AML initiating cells and that thymic progenitors can transmit AML.


Subject(s)
Cell Transformation, Neoplastic/pathology , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Nuclear Pore Complex Proteins/metabolism , Stem Cell Transplantation/methods , Thymocytes/cytology , Transcription Factors/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Transgenic , Nuclear Pore Complex Proteins/genetics , Transcription Factors/genetics
6.
Blood Adv ; 1(20): 1749-1759, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29296821

ABSTRACT

B-1 and B-2 lymphocytes are derived from distinct developmental pathways and represent layered arms of the innate and adaptive immune systems, respectively. In contrast to a majority of murine B-cell malignancies, which stain positive with the B220 antibody, we discovered a novel form of B-cell leukemia in NUP98-PHF23 (NP23) transgenic mice. The immunophenotype (Lin- B220- CD19+ AA4.1+) was identical to that of progenitor (pro) B-1 cells, and VH gene usage was skewed toward 3' V regions, similar to murine fetal liver B cells. Moreover, the gene expression profile of these leukemias was most similar to that of fetal liver pro-B fraction BC, a known source of B-1 B cells, further supporting a pro-B-1 origin of these leukemias. The NP23 pro-B-1 acute lymphoblastic leukemias (ALLs) acquired spontaneous mutations in both Bcor and Janus kinase (Jak) pathway (Jak1/2/3 and Stat5a) genes, supporting a hypothesis that mutations in 3 critical pathways (stem-cell self-renewal, B-cell differentiation, and cytokine signaling) collaborate to induce B-cell precursor (BCP) ALL. Finally, the thymic stromal lymphopoietin (Tslp) cytokine is required for murine B-1 development, and chromosomal rearrangements resulting in overexpression of the TSLP receptor (CRLF2) are present in some patients with high-risk BCP-ALL (referred to as CRLF2r ALL). Gene expression profiles of NP23 pro-B-1 ALL were more similar to that of CRLF2r ALL than non-CRLF2r ALL, and analysis of VH gene usage from patients with CRLF2r ALL demonstrated preferential usage of VH regions used by human B-1 B cells, leading to the suggestion that this subset of patients with BCP-ALL has a malignancy of B-1, rather than B-2, B-cell origin.

7.
Biometals ; 26(6): 897-911, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23948867

ABSTRACT

Apart from cytotoxicity cadmium has no special attributes towards cell's physiological function. The role of cadmium with respect to cell growth is still under debate. Mitogen activated protein kinase and Ca(2+)/calmodulin dependent protein kinase dependent pathways are the two elaborately studied concerning cadmium induced cell proliferation. Low concentration of cadmium chloride (2.5 µM) was applied to mice primary lung epithelial cells and cell proliferation was measured both by cell cycle analysis and Brdu incorporation assay. Effects of differential dose of cadmium chloride on lung epithelial cells were evaluated morphologically by atomic force microscopy. RT-PCR and western blot altogether corroborated the specific signalling pathways concerning cadmium induced lung cell proliferation. Cadmium induced lung epithelial cells which over-expressed EGFR, were transfected with siEGFR, revealed downstream molecules and RNAi induced EGFR silencing. Use of siEGFR effectively prevents expression of proinflammatory and cell proliferative markers. Moreover N-acetyl cysteine and ascorbic acid mediated inhibition of EGFR and downstream signalling molecules indicate the involvement of reactive oxygen species. Exposure to low concentration of cadmium promotes the growth of primary mice lung epithelial cell by EGFR signalling. We have also transfected the primary lung epithelial cell with siRNA against the regulatory subunit of nuclear factor-κB (NF-κB) and the data shows that cadmium induced lung cell proliferation is the effect of EGFR mediated NF-κB activation.


Subject(s)
Cadmium Chloride/pharmacology , Epithelial Cells/drug effects , ErbB Receptors/genetics , Transcription Factor RelA/genetics , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Epithelial Cells/cytology , Epithelial Cells/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Expression Regulation , Lung/cytology , Lung/drug effects , Lung/metabolism , Male , Mice , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Signal Transduction , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism
8.
Cell Biol Int ; 37(5): 449-57, 2013 May.
Article in English | MEDLINE | ID: mdl-23494890

ABSTRACT

Transforming growth factor-ß (TGF-ß) is a potent inhibitor of the growth of normal mammary epithelial cells, and has a pleiotropic, context-dependent, concentration-dependent action. We found attenuation of TGF-ß signalling in mammary adenoma carcinoma cells. Phosphorylation at the linker site of Smad2 occurred in a cooperative way during the attenuation of TGF-ß signalling, and was associated with upregulation of CDK2 and cyclin D1. CDK2 inhibitor restored the anti-proliferative effect of TGF-ß by upregulating p21, with inhibition of linker phosphorylation of Smad2. CDK2-mediated linker phosphorylation of Smad2 may be a plausible mechanism for the attenuation of TGF-ß signalling in breast cancer.


Subject(s)
Signal Transduction/drug effects , Smad2 Protein/metabolism , Transforming Growth Factor beta/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , MCF-7 Cells , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation/drug effects
9.
Inhal Toxicol ; 23(6): 339-48, 2011 May.
Article in English | MEDLINE | ID: mdl-21605009

ABSTRACT

Exposure to cadmium is associated with the development of pulmonary damage such as emphysema and lung cancer. This metal is also a powerful inducer of different proinflammatory and cell cycle regulatory proteins in many biologic models. Previously, we showed that prolonged exposure of low concentration of cadmium resulted in upregulation of proinflammatory cytokines and cell cycle regulatory molecules in mice lung cell. The present study was undertaken to determine molecular mechanism of inflammation and its relation to cell proliferation in a transformed human lung adenocarcinoma epithelial cell line (A549) in response to cadmium chloride. In comparative studies, we examine that short-duration exposure to lower doses of cadmium significantly increase the growth of A549 cells, whereas higher doses are toxic and cause cell death. We also observed that cadmium induced elevated expression of epidermal growth factor receptor (EGFR) along with different proinflammatory cytokines like interleukin-1 beta (IL-1ß), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6). The possible occurrence of cell proliferation events was evaluated via analysis of the physical state of the DNA and the expression of Ki-67 and proliferating cell nuclear antigen (PCNA). We also checked the pattern of expression of different cell cycle regulatory molecules involved in the onset of cell proliferation. Our results indicate that cadmium treatment appears to induce inflammatory and growth responses in transformed A549 cell line by activating EGFR and its downstream modulators. These results may contribute to better understand the toxic mechanism of cadmium; moreover, the expression profile of cadmium-induced regulatory molecules could provide potential biomarkers for cadmium exposure.


Subject(s)
Adenocarcinoma/metabolism , Cadmium Chloride/toxicity , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Cadmium Chloride/administration & dosage , Cell Cycle , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/toxicity , ErbB Receptors/genetics , Gefitinib , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Lethal Dose 50 , Microscopy, Acoustic , Proliferating Cell Nuclear Antigen/metabolism , Quinazolines/pharmacology , Up-Regulation
10.
J Inflamm (Lond) ; 6: 19, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19523218

ABSTRACT

BACKGROUND: Cadmium is one of the inflammation-related xenobiotics and has been regarded as a potent carcinogen. The relationship between inflammation and cell proliferation due to chronic infection has been studied, but the mechanism is not fully clear. Though the mode of cadmium toxicity is well characterized in animal cells, still it requires some further investigations. Previously we reported that cadmium induces immune cell death in Swiss albino mice. In the present study we showed that instead of inducing cell death mechanism, cadmium in low concentration triggers proliferation in mice lung cell and our results reveals that prior to the induction of proliferation it causes severe inflammation. METHODS: Swiss albino mice were treated with different concentrations of cadmium to determine the LD50. Mice were subdivided (5 mice each) according to the exposure period (15, 30, 45, 60 days) and were given sub lethal dose (5 mg/Kg body weight) of cadmium chloride and ibuprofen (50 mg/Kg body weight, recommended dose) once in a week. SEM and histology were performed as evidence of changes in cellular morphology. Inflammation was measured by the expression of Cox-2 and MMPs. Expression of proinflammatory cytokines (Cox-2, IL-6), signaling and cell cycle regulatory molecules (STAT3, Akt, CyclinD1) were measured by western blot, ELISA and immunoprecipitation. Mutagenecity was evidenced by comet assay. Cell proliferation was determined by cell count, cell cycle and DNA analysis. RESULTS: Prolonged exposure of low concentration of cadmium resulted in up regulation of proinflammatory cytokines and cell cycle regulatory molecules. Though NSAIDs like Ibuprofen reduces the expression of inflammatory cytokines, but it did not show any inhibitory effect on cadmium adopted lung cell proliferation. CONCLUSION: Our results prove that cadmium causes both inflammation and cell proliferation when applied in a low dose but proliferative changes occur independent of inflammation.

11.
Mutat Res ; 663(1-2): 22-31, 2009 Apr 26.
Article in English | MEDLINE | ID: mdl-19475715

ABSTRACT

Recently, the role of cadmium (Cd) in immunosupression has gained importance. Nevertheless, the signaling pathways underlying cadmium-induced immune cell death remains largely unclear. In accordance to our previous in vivo report, and to evaluate the further details of the mechanism, we have investigated the effects of cadmium (CdCl(2), H(2)O) on cell cycle regulation and apoptosis in splenocytes in vitro. Our results have revealed that reactive oxygen species (ROS) and p21 are involved in cell cycle arrest in a p53 independent manner but late hour apoptotic response was accompanied by the p53 up-regulation, loss of mitochondrial transmembrane potential (MTP), down-regulation of Bcl-xl, activation of caspase-3 and release of cytochrome c (Cyt c). However, pifithrin alfa (PFT-alpha), an inhibitor of p53, fails to rescue the cells from the cadmium-induced cell cycle arrest but prevents Bcl-xl down-regulation and loss of Deltapsi(m), which indicates that there is an involvement of p53 in apoptosis. In contrast, treatment with N-acetyl cysteine (NAC) can prevent cell cycle arrest and p21 up-regulation at early hours. Although it is clear that, NAC has no effect on apoptosis, p53 expression and MPT changes at late stage events. Taken together, we have demonstrated that cadmium promotes ROS generation, which potently initiates the cell cycle arrest at early hours and finally induces p53-dependent apoptosis at later part of the event.


Subject(s)
Apoptosis/drug effects , Cadmium/toxicity , Cell Cycle/drug effects , Reactive Oxygen Species/metabolism , Animals , Caspase 3/metabolism , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytochromes c/metabolism , Enzyme Activation/drug effects , Flow Cytometry , Inhibitory Concentration 50 , Membrane Potential, Mitochondrial/drug effects , Mice , Models, Biological , Spectrophotometry , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
12.
J Biol Inorg Chem ; 13(7): 1149-55, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18597125

ABSTRACT

An investigation of the molecular mechanism of the anticancer activity demonstrated by the ruthenium(II)-arene compound [Ru(eta(6)-p-cymene)Cl(2)(pta)] (pta is 1,3,5-triaza-7-phosphaadamantane), termed "RAPTA-C", in Ehrlich ascites carcinoma (EAC) bearing mice is described. RAPTA-C exhibits effective cell growth inhibition by triggering G(2)/M phase arrest and apoptosis in cancer cells. Cell cycle arrest is associated with increased levels of p21 and reduced amounts of cyclin E. RAPTA-C treatment also enhances the levels of p53, and its treatment triggers the mitochondrial apoptotic pathway, as shown by the change in Bax to Bcl-2 ratios, resulting in cytochrome c release and caspase-9 activation. c-Jun NH(2)-terminal kinase (JNK) is a critical mediator in RAPTA-C-induced cell growth inhibition. Activation of JNK by RAPTA-C increases significantly during apoptosis. Overall, these results suggest a critical role for JNK and p53 in RAPTA-C-induced G(2)/M arrest and apoptosis of EAC-bearing mice. Consequently, RAPTA-C treatment results in a significant inhibition in the progression of cancer in an animal model, which emulates the human disease, and does so with remarkably low general toxicity; hence, RAPTA-C has potential for clinical application.


Subject(s)
Apoptosis/drug effects , Carcinoma, Ehrlich Tumor/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/drug effects , Organometallic Compounds/pharmacology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cymenes , Cytochromes c/metabolism , DNA/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Mice , Mitochondria/metabolism , bcl-2-Associated X Protein/metabolism
13.
Toxicol Lett ; 177(2): 83-9, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18281164

ABSTRACT

Cadmium is the major component of polluted environment which can be fatal by mechanisms that are not fully clear. Our study indicates immunosupression may be one of the reason for that. It is well known that cadmium (Cd) has toxic and carcinogenic effects in rhondents and humans, but the effects of cadmium on apoptosis are still not clear. Although some studies have shown that cadmium has apoptotic potential, other studies have shown that cadmium can be anti-apoptotic. In the present study, we aimed to determine the mode of cell death and its mechanism in Swiss albino mice splenocyte by cadmium for its toxic effects. To identify the nature of cell death, our result signifies apoptotic mode of killing. In search of the mechanism behind it we found that cadmium increased cell death and lowered the survival of the host in a dose dependent manner. In search of the reason we found increased expression of the pro-apoptotic proteins p53 in splenic lymphocytes. Here we showed that cadmium induced p53-dependent apoptosis through cooperation between Bcl-xl down regulation without changing the Bcl-2 and Bax expression, the common target of p53. The down regulation of Bcl-xl strongly indicating mitochondrial involvement in apoptosis. It is confirmed by the release of cytochrome c and activation of caspase-3. All of these findings establish an important role of p53 and mitochondrial function in cadmium induced toxic environment in the host.


Subject(s)
Apoptosis , Cadmium/toxicity , Environmental Pollutants/toxicity , Lymphocytes/drug effects , Spleen/drug effects , Animals , Caspase 3/metabolism , Cell Cycle , Cell Nucleus/chemistry , Cytochromes c/metabolism , DNA/analysis , Flow Cytometry , Lymphocyte Count , Lymphocytes/immunology , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Spleen/cytology , Spleen/immunology , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...