Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biomedicines ; 10(9)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36140319

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) is the most common type of epithelial ovarian cancer. The majority of cases are diagnosed at advanced stages, when intraperitoneal (IP) spread has already occurred. Despite significant surgical and chemotherapeutic advances in HGSOC treatment over the past decades, survival rates with HGSOC have only modestly improved. Chimeric antigen receptor (CAR)-T cells enable T cells to directly bind to tumor-associated antigens in a major histocompatibility complex-independent manner, thereby inducing tumor rejection. While CAR-T cell therapy shows great promise in hematological malignancies, its use in solid tumors is limited. Therefore, innovative approaches are needed to increase the specificity of CAR-modified T cells against solid tumors. The aim of this study was to assess the efficacy and safety of intraperitoneal (IP) versus intravenous (IV) CAR-T cell therapy in the treatment of HGSOC. We constructed a CAR that targets the ErbB2/HER2 protein (ErbB2CAR), which is overexpressed in HGSOC, and evaluated the functionality of ErbB2CAR on ovarian cancer cell lines (OVCAR8, SKOV3, and NAR). Our findings show that an IP injection of ErbB2CAR-T cells to tumor-bearing mice led to disease remission and increased survival compared to the IV route. Moreover, we found that IP-injected ErbB2CART cells circulate to a lesser extent, making them safer for non-tumor tissues than IV-injected cells. Further supporting our findings, we show that the effect of ErbB2CAR-T cells on primary HGSOC tumors is correlated with ErbB2 expression. Together, these data demonstrate the advantages of an IP administration of CAR-T cells over IV administration, offering not only a safer strategy but also the potential for counteracting the effect of ErbB2CAR in HGSOC. Significance: IP-injected ErbB2CAR-T cells led to disease remission and increased survival compared to the IV route. These findings demonstrate the advantages of IP administration, offering a safe treatment strategy with the potential for counteracting the effect of ErbB2CAR in HGSOC.

2.
Hum Gene Ther ; 32(19-20): 1224-1241, 2021 10.
Article in English | MEDLINE | ID: mdl-34494460

ABSTRACT

Chimeric antigen receptor (CAR)-T cells are genetically engineered T cells, directed against a tumor-associated antigen. Extracellular vesicles (EVs) derived from CAR-T cells (CAR-T EVs) may preserve CAR-T activity and overcome one of the major obstacles responsible for CAR-T cell failure in patients with solid tumors. This study aimed to compare CAR-T EVs with their parental cells and explore their cell penetration and cytotoxic activity. Anti-HER-2 CARs were stimulated with specific target cells. EVs were isolated from the cell media and characterized for their content and functions. We found that CAR-T EVs contained a mixture of small and large EVs. Stimulated anti-HER-2+ CAR-T EVs expressed lower cytokine levels compared with their parental CAR-T cells (such as interferon gamma). Higher levels of granzyme B were found in CAR-T EVs (≥20 × ) compared with EVs from unstimulated cells (p < 0.001). Anti-HER-2+ CAR-T EVs bound and penetrated specifically into HER-2 expressing target cells. Similar cytotoxic effects measured by caspase-3/7 activity were found in CAR-T cells and their derived EVs. However, while the CAR-T cells induced massive apoptosis during the first 24 h, CAR-T EVs required 60 - 90 h. In summary, CAR-T EVs provide a novel potent immunotherapy approach that may be effective against solid tumors.


Subject(s)
Extracellular Vesicles , Neoplasms , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
3.
Cancer Immunol Res ; 8(12): 1485-1495, 2020 12.
Article in English | MEDLINE | ID: mdl-33008840

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy has shown remarkable successes in fighting B-cell leukemias/lymphomas. Promising response rates are reported in patients treated with B-cell maturation antigen (BCMA) CAR T cells for multiple myeloma. However, responses appear to be nondurable, highlighting the need to expand the repertoire of multiple myeloma-specific targets for immunotherapy and to generate new CAR T cells. Here, we developed a "dual-CAR" targeting two multiple myeloma-associated antigens and explored its safety and efficacy. To reduce the "off-target" toxicity, we used the recognition of paired antigens that were coexpressed by the tumor to induce efficient CAR T-cell activation. The dual-CAR construct presented here was carefully designed to target the multiple myeloma-associated antigens, taking into consideration the distribution of both antigens on normal human tissues. Our results showed that the CD138/CD38-targeted dual CAR (dCAR138-38) elicited a potent anti-multiple myeloma response both in vitro and in vivo NSG mice transplanted with a multiple myeloma cell line and treated with dCAR138-38 showed median survival of 97 days compared with 31 days in the control group treated with mock-lymphocytes. The dCAR138-38 showed increased specificity toward cells expressing both targeted antigens compared with single-antigen-expressing cells and low activity toward primary cells from healthy tissues. Our findings indicated that the dCAR138-38 may provide a potent and safe alternative therapy for patients with multiple myeloma.


Subject(s)
B-Cell Maturation Antigen/immunology , Multiple Myeloma/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , B-Cell Maturation Antigen/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Immunotherapy , Immunotherapy, Adoptive/methods , Mice , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism
4.
Theranostics ; 10(19): 8744-8756, 2020.
Article in English | MEDLINE | ID: mdl-32754275

ABSTRACT

Erythropoietin (EPO) is a key regulator of erythropoiesis. However, EPO receptors (EPO-Rs) are also expressed on non-erythroid cell types, including myeloid and bone cells. Immune cells also participate in bone homeostasis. B cells produce receptor activator of nuclear factor kappa-Β ligand (RANKL) and osteoprotegerin (OPG), two pivotal regulators of bone metabolism. Here we explored the ability of B cells to transdifferentiate into functional osteoclasts and examined the role of EPO in this process in a murine model. Methods: We have combined specifically-designed experimental mouse models and in vitro based osteoclastogenesis assays, as well as PCR analysis of gene expression. Results: (i) EPO treatment in vivo increased RANKL expression in bone marrow (BM) B cells, suggesting a paracrine effect on osteoclastogenesis; (ii) B cell-derived osteoclastogenesis occured in vivo and in vitro, as demonstrated by B cell lineage tracing in murine models; (iii) B-cell-derived osteoclastogenesis in vitro was restricted to Pro-B cells expressing CD115/CSF1-R and is enhanced by EPO; (iv) EPO treatment increased the number of B-cell-derived preosteoclasts (ß3+CD115+), suggesting a physiological rationale for B cell derived osteoclastogenesis; (v) finally, mice with conditional EPO-R knockdown in the B cell lineage (cKD) displayed a higher cortical and trabecular bone mass. Moreover, cKD displayed attenuated EPO-driven trabecular bone loss, an effect that was observed despite the fact that cKD mice attained higher hemoglobin levels following EPO treatment. Conclusions: Our work highlights B cells as an important extra-erythropoietic target of EPO-EPO-R signaling and suggests their involvement in the regulation of bone homeostasis and possibly in EPO-stimulated erythropoietic response. Importantly, we present here for the first time, histological evidence for B cell-derived osteoclastogenesis in vivo.


Subject(s)
B-Lymphocytes/cytology , Bone Remodeling/drug effects , Erythropoietin/pharmacology , Receptors, Erythropoietin/genetics , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Transdifferentiation/drug effects , Female , Gene Knockout Techniques , Mice , Osteogenesis , RANK Ligand/metabolism , Receptors, Erythropoietin/metabolism
5.
Int J Mol Sci ; 21(11)2020 May 27.
Article in English | MEDLINE | ID: mdl-32471308

ABSTRACT

Recent studies have demonstrated that erythropoietin (EPO) treatment in mice results in trabecular bone loss. Here, we investigated the dose-response relationship between EPO, hemoglobin (Hgb) and bone loss and examined the reversibility of EPO-induced damage. Increasing doses of EPO over two weeks led to a dose-dependent increase in Hgb in young female mice, accompanied by a disproportionate decrease in trabecular bone mass measured by micro-CT (µCT). Namely, increasing EPO from 24 to 540 IU/week produced a modest 12% rise in Hgb (20.2 ± 1.3 mg/dL vs 22.7 ± 1.3 mg/dL), while trabecular bone volume fraction (BV/TV) in the distal femur decreased dramatically (27 ± 8.5% vs 53 ± 10.2% bone loss). To explore the long-term skeletal effects of EPO, we treated mice for two weeks (540 IU/week) and monitored bone mass changes after treatment cessation. Six weeks post-treatment, there was only a partial recovery of the trabecular microarchitecture in the femur and vertebra. EPO-induced bone loss is therefore dose-dependent and mostly irreversible at doses that offer only a minor advantage in the treatment of anemia. Because patients requiring EPO therapy are often prone to osteoporosis, our data advocate for using the lowest effective EPO dose for the shortest period of time to decrease thromboembolic complications and minimize the adverse skeletal outcome.


Subject(s)
Bone Resorption/etiology , Erythropoietin/adverse effects , Animals , Bone Resorption/diagnostic imaging , Bone Resorption/pathology , Cancellous Bone/diagnostic imaging , Cancellous Bone/drug effects , Cells, Cultured , Erythropoietin/administration & dosage , Erythropoietin/pharmacology , Female , Femur/diagnostic imaging , Femur/drug effects , Hemoglobins/metabolism , Mice , Mice, Inbred C57BL , Spine/diagnostic imaging , Spine/drug effects
6.
Sci Rep ; 7(1): 10379, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871174

ABSTRACT

Erythropoietin (EPO) is the main hormone driving mammalian erythropoiesis, with activity mediated via the surface receptor, EPO-R, on erythroid progenitor cells. Recombinant human EPO is currently used clinically for the treatment of anemia in patients with end-stage renal disease, and in certain cancer patients suffering from anemia induced either by the tumor itself or by chemotherapy. EPO-R expression is also detected in non-erythroid cells, including macrophages present in the peritoneum, spleen, and bone marrow (BM). Here we demonstrate that Kupffer cells (KCs) - the liver-resident macrophages - are EPO targets. We show that, in vitro, EPO initiated intracellular signalling and enhanced phagocytosis in a rat KC line (RKC-2) and in sorted KCs. Moreover, continuous EPO administration in mice, resulted in an increased number of KCs, up-regulation of liver EPO-R expression and elevated production of the monocyte chemoattractant CCL2, with corresponding egress of Ly6Chi monocytes from the BM. In a model of acute acetaminophen-induced liver injury, EPO administration increased the recruitment of Ly6Chi monocytes and neutrophils to the liver. Taken together, our results reveal a new role for EPO in stimulating KC proliferation and phagocytosis, and in recruiting Ly6Chi monocytes in response to liver injury.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Erythropoietin/genetics , Kupffer Cells/cytology , Receptors, Erythropoietin/metabolism , Recombinant Proteins/administration & dosage , Animals , Antigens, Ly/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Disease Models, Animal , Erythropoietin/administration & dosage , Erythropoietin/pharmacology , Humans , Kupffer Cells/metabolism , Liver/drug effects , Liver/metabolism , Mice , Phagocytosis , Rats , Recombinant Proteins/pharmacology , Signal Transduction , Up-Regulation
7.
Leuk Res ; 52: 20-27, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27870945

ABSTRACT

The immune system is impaired in myelodysplastic syndromes (MDS) and plays a role in the pathogenesis of the disease. Here we show effects of recombinant human erythropoietin (rHuEPO) on T cell (CD4+, CD8+ and CD4+CD25+) number and function in MDS patients. Healthy (20 subjects), MDS patients without rHuEPO treatment ('MDS', 13), and MDS patients treated with rHuEPO ('MDS+EPO', 17) were examined. CD4+ and CD8+ T cell numbers were reduced and increased respectively in MDS compared to healthy subjects. EPO treatment normalized these levels. CD4+CD25+ cell numbers, lower in MDS, were normalized in MDS+EPO. In vitro activation of CD4+ and CD8+ cells with phytohemagglutinin as measured by CD69 expression, demonstrated a 7.2 fold increase in CD4+ activation vs 13.6 fold for MDS and MDS+EPO respectively (p=0.004); and 10.2 fold (MDS) vs 18.6 fold (MDS+EPO, p<0.003) for CD8+ T cells. Expression of the co-stimulatory marker CD28, decreased in CD4+ and CD8+ T cells in MDS, was normalized in MDS+EPO CD4+ T cells. Subgroup analysis of milder disease (WHO RA and RARS) and more advanced disease revealed no difference in CD4+ and CD8+ T cell numbers. However, the activation of these cells in the RA/RARS subgroup was impaired in EPO-untreated and enhanced in EPO-treated MDS patients. Our data suggest that EPO treatment improves immune abnormalities in MDS and may depend on disease severity.


Subject(s)
Erythropoietin/administration & dosage , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/immunology , T-Lymphocytes/pathology , Aged , Aged, 80 and over , CD28 Antigens/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured , Erythropoietin/immunology , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Count , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , T-Lymphocytes/immunology
8.
Sci Rep ; 6: 30998, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27481313

ABSTRACT

Multiple myeloma (MM) is a plasma cell malignancy, characterized by osteolytic lesions and monoclonal immunoglobulins. The anemia, accompanying the disease is often treated with recombinant human EPO. Diverse non-erythropoietic effects of EPO have led us to question its combined action on the immune system and bone in the 5T33MM mouse model. EPO administration to MM mice attenuated disease progression as demonstrated by a decrease in serum MM IgG2b, splenic CD138 expressing cells, IL-6 and RORγτ transcripts in bone marrow (BM). IFN-γ transcript levels and macrophages (F4/80(+)CD11b(+)) in the BM both increased ~1.5 fold in the EPO-treated MM mice. In-vitro, EPO stimulated phagocytosis of 5T33MM cells (+30%) by BM-derived macrophages. In contrast, high-resolution microCT analysis of distal femurs revealed EPO-associated bone loss in both healthy and 5T33MM mice. EPO significantly increased expression of the osteoclastogenic nuclear factor-kappa B ligand (RANKL) in healthy mice, but not in MM mice, likely due to antagonizing effects on MM progression. Thus, in MM, EPO may act as a double-edged-sword stimulating immune response, while accelerating bone resorption, possibly via direct action on BM macrophages. This study supports a prudent approach of treating anemia in MM patients, aiming to maintain EPO-associated anti-MM effects, while considering bone damage.


Subject(s)
Anemia/drug therapy , Bone Marrow/immunology , Bone Resorption/etiology , Erythropoietin/pharmacology , Macrophages/immunology , Multiple Myeloma/complications , Anemia/etiology , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Cells, Cultured , Female , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Multiple Myeloma/immunology , Phagocytosis/physiology , Signal Transduction/drug effects
9.
FASEB J ; 29(5): 1890-900, 2015 May.
Article in English | MEDLINE | ID: mdl-25630969

ABSTRACT

Erythropoietin (EPO) primarily regulates red blood cell formation, and EPO serum levels are increased on hypoxic stress (e.g., anemia and altitude). In addition to anemia, recent discoveries suggest new therapeutic indications for EPO, unrelated to erythropoiesis. We investigated the skeletal role of EPO using several models of overexpression (Tg6 mice) and EPO administration (intermittent/continuous, high/low doses) in adult C57Bl6 female mice. Using microcomputed tomography, histology, and serum markers, we found that EPO induced a 32%-61% trabecular bone loss caused by increased bone resorption (+60%-88% osteoclast number) and reduced bone formation rate (-19 to -74%; P < 0.05 throughout). EPO targeted the monocytic lineage by increasing the number of bone monocytes/macrophages, preosteoclasts, and mature osteoclasts. In contrast to the attenuated bone formation in vivo, EPO treatment in vitro did not inhibit osteoblast differentiation and activity, suggesting an indirect effect of EPO on osteoblasts. However, EPO had a direct effect on preosteoclasts by stimulating osteoclastogenesis in isolated cultures (+60%) via the Jak2 and PI3K pathways. In summary, our findings demonstrate that EPO negatively regulates bone mass and thus bears significant clinical implications for the potential management of patients with endogenously or therapeutically elevated EPO levels.


Subject(s)
Bone Resorption/etiology , Erythropoietin/physiology , Osteoclasts/cytology , Receptors, Erythropoietin/metabolism , Animals , Apoptosis , Blotting, Western , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoclasts/metabolism , Osteogenesis/physiology , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Erythropoietin/genetics , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
10.
RNA ; 19(5): 591-604, 2013 May.
Article in English | MEDLINE | ID: mdl-23474544

ABSTRACT

Alternative mRNA splicing is a major mechanism for gene regulation and transcriptome diversity. Despite the extent of the phenomenon, the regulation and specificity of the splicing machinery are only partially understood. Adenosine-to-inosine (A-to-I) RNA editing of pre-mRNA by ADAR enzymes has been linked to splicing regulation in several cases. Here we used bioinformatics approaches, RNA-seq and exon-specific microarray of ADAR knockdown cells to globally examine how ADAR and its A-to-I RNA editing activity influence alternative mRNA splicing. Although A-to-I RNA editing only rarely targets canonical splicing acceptor, donor, and branch sites, it was found to affect splicing regulatory elements (SREs) within exons. Cassette exons were found to be significantly enriched with A-to-I RNA editing sites compared with constitutive exons. RNA-seq and exon-specific microarray revealed that ADAR knockdown in hepatocarcinoma and myelogenous leukemia cell lines leads to global changes in gene expression, with hundreds of genes changing their splicing patterns in both cell lines. This global change in splicing pattern cannot be explained by putative editing sites alone. Genes showing significant changes in their splicing pattern are frequently involved in RNA processing and splicing activity. Analysis of recently published RNA-seq data from glioblastoma cell lines showed similar results. Our global analysis reveals that ADAR plays a major role in splicing regulation. Although direct editing of the splicing motifs does occur, we suggest it is not likely to be the primary mechanism for ADAR-mediated regulation of alternative splicing. Rather, this regulation is achieved by modulating trans-acting factors involved in the splicing machinery.


Subject(s)
Adenosine Deaminase/genetics , Alternative Splicing/genetics , RNA Precursors/genetics , RNA, Messenger/genetics , Adenosine/genetics , Adenosine/metabolism , Adenosine Deaminase/metabolism , Base Sequence , Cell Line, Tumor , Exons/genetics , Gene Expression Regulation , Humans , Inosine/genetics , RNA Editing/genetics , RNA-Binding Proteins
11.
Eur J Cell Biol ; 87(5): 267-78, 2008 May.
Article in English | MEDLINE | ID: mdl-18403046

ABSTRACT

Proteins of the nuclear envelope have been implicated as participating in gene silencing. BAF, a DNA- and LEM domain-binding protein, has been suggested to link chromatin to the nuclear envelope. We have previously shown that LAP2beta, a LEM-domain inner nuclear membrane protein, represses transcription through binding to HDAC3 and induction of histone H4 deacetylation. We now show that LAP2zeta, the smallest LAP2 family member, is also involved in regulation of transcription. We show that similar to other LEM-domain proteins LAP2zeta interacts with BAF. LAP2zeta-YFP and BAF co-localize in the cytoplasm, and overexpression of LAP2zeta leads to reduction of nucleoplasmic BAF. Mutations in the LAP2zeta-YFP LEM domain decrease its interaction with BAF retaining the nucleo-cytoplasmic distribution of BAF. Co-expression of LAP2beta and LAP2zeta results in inhibition of LAP2beta-induced gene silencing while overexpression of LAP2zeta alone leads to a small increase in transcriptional activity of various transcription factors. Our results suggest that LAP2zeta is a transcriptional regulator acting predominantly to inhibit LAP2beta-mediated repression. LAP2zeta may function by decreasing availability of BAF. These findings could have implications in the study of nuclear lamina-associated diseases and BAF-dependent retroviral integration.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Nucleus/metabolism , Cloning, Molecular , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Histone Deacetylases/metabolism , Humans , Membrane Proteins/genetics , Mice , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Alignment , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...