Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biochem Cell Biol ; 101(6): 481-495, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37566901

ABSTRACT

Modulating the immune system to treat diseases, including myeloid malignancies, has resulted in the development of a multitude of novel therapeutics in recent years. Myelodysplastic syndromes or neoplasms (MDS) and acute myeloid leukemia (AML) are hematologic malignancies that arise from defects in hematopoietic stem and progenitor cells (HSPCs). Dysregulated immune responses, especially in innate immune and inflammatory pathways, are highly associated with the acquisition of HSPC defects in MDS and AML pathogenesis. In addition to utilizing the immune system in immunotherapeutic interventions such as chimeric antigen receptor T cell therapy, vaccines, and immune checkpoint inhibitors, mitigating dysregulation of innate immune and inflammatory responses in MDS and AML remains a priority in slowing the initiation and progression of these myeloid malignancies. This review provides a comprehensive summary of the current progress of diverse strategies to utilize or modulate the immune system in the treatment of MDS and AML.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/pathology , Leukemia, Myeloid, Acute/therapy , Immune System/metabolism , Immune System/pathology
2.
Haematologica ; 108(10): 2715-2729, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37102608

ABSTRACT

Inflammation is associated with the pathogenesis of myelodysplastic syndromes (MDS) and emerging evidence suggests that MDS hematopoietic stem and progenitor cells (HSPC) exhibit an altered response to inflammation. Deletion of chromosome 5 (del(5q)) is the most common chromosomal abnormality in MDS. Although this MDS subtype contains several haploinsufficient genes that impact innate immune signaling, the effects of inflammation on del(5q) MDS HSPC remains undefined. Utilizing a model of del(5q)-like MDS, inhibiting the IRAK1/4-TRAF6 axis improved cytopenias, suggesting that activation of innate immune pathways contributes to certain clinical features underlying the pathogenesis of low-risk MDS. However, low-grade inflammation in the del(5q)-like MDS model did not contribute to more severe disease but instead impaired the del(5q)-like HSPC as indicated by their diminished numbers, premature attrition and increased p53 expression. Del(5q)-like HSPC exposed to inflammation became less quiescent, but without affecting cell viability. Unexpectedly, the reduced cellular quiescence of del(5q) HSPC exposed to inflammation was restored by p53 deletion. These findings uncovered that inflammation confers a competitive advantage of functionally defective del(5q) HSPC upon loss of p53. Since TP53 mutations are enriched in del(5q) AML following an MDS diagnosis, increased p53 activation in del(5q) MDS HSPC due to inflammation may create a selective pressure for genetic inactivation of p53 or expansion of a pre-existing TP53-mutant clone.


Subject(s)
Myelodysplastic Syndromes , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Chromosome Deletion , Myelodysplastic Syndromes/pathology , Hematopoietic Stem Cells/metabolism , Signal Transduction , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 5/metabolism
3.
J Antibiot (Tokyo) ; 75(3): 146-154, 2022 03.
Article in English | MEDLINE | ID: mdl-35058577

ABSTRACT

Disulfide analogs of the alcohol sobriety medication disulfiram (Antabuse®) were evaluated for antimicrobial activity. Structure-activity relationship analyses of MIC data obtained for methicillin-resistant Staphylococcus aureus (MRSA) and other pathogenic organisms revealed correlations between the lipophilicity and bulkiness of the substituents. Analogs conferring optimal anti-MRSA activity contained S-octyl disulfides and either N,N-dimethyl- or N-pyrrolidine dithiocarbamate substituents. Additional testing revealed that both disulfiram and its S-octyl derivative are capable of sensitizing S. aureus to the bactericidal effects of fosfomycin. Mechanistic studies established that the compounds decrease intracellular levels of the fosB cofactor bacillithiol through a thiol-disulfide exchange reaction. The increased fosfomycin susceptibility in S. aureus was thereby attributed to a depleted cellular bacillithiol pool available for inactivation by fosB.


Subject(s)
Anti-Infective Agents/pharmacology , Disulfiram/pharmacology , Drug Resistance, Bacterial/drug effects , Fosfomycin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Bacterial Proteins/metabolism , Cysteine/analogs & derivatives , Cysteine/metabolism , Glucosamine/analogs & derivatives , Glucosamine/metabolism , Humans , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests/methods , Staphylococcal Infections/drug therapy , Sulfhydryl Compounds/pharmacology
4.
Front Immunol ; 12: 720635, 2021.
Article in English | MEDLINE | ID: mdl-35087510

ABSTRACT

Objective: Prenatal cadmium (Cd) exposure leads to immunotoxic phenotypes in the offspring affecting coding and non-coding genes. Recent studies have shown that long non-coding RNAs (lncRNAs) are integral to T cell regulation. Here, we investigated the role of long non-coding RNA small nucleolar RNA host gene 7 (lncSnhg7) in T cell proliferation. Methods: RNA sequencing was used to analyze the expression of lncRNAs in splenic CD4+ T cells with and without CD3/CD28 stimulation. Next, T cells isolated from offspring exposed to control or Cd water throughout mating and gestation were analyzed with and without stimulation with anti-CD3/CD28 beads. Quantitative qPCR and western blotting were used to detect RNA and protein levels of specific genes. Overexpression of a miR-34a mimic was achieved using nucleofection. Apoptosis was measured using flow cytometry and luminescence assays. Flow cytometry was also used to measure T cell proliferation in culture. Finally, lncSnhg7 was knocked down in splenic CD4+ T cells with lentivirus to assess its effect on proliferation. Results: We identified 23 lncRNAs that were differentially expressed in stimulated versus unstimulated T cells, including lncSnhg7. LncSnhg7 and a downstream protein, GALNT7, are upregulated in T cells from offspring exposed to Cd during gestation. Overexpression of miR-34a, a regulator of lncSnhg7 and GALNT7, suppresses GALNT7 protein levels in primary T cells, but not in a mouse T lymphocyte cell line. The T cells isolated from Cd-exposed offspring exhibit increased proliferation after activation in vitro, but Treg suppression and CD4+ T cell apoptosis are not affected by prenatal Cd exposure. Knockdown on lncSnhg7 inhibits proliferation of CD4+ T cells. Conclusion: Prenatal Cd exposure alters the expression of lncRNAs during T cell activation. The induction of lncSnhg7 is enhanced in splenic T cells from Cd offspring resulting in the upregulation of GALNT7 protein and increased proliferation following activation. miR-34a overexpression decreased GALNT7 expression and knockdown of lncSnhg7 inhibited proliferation suggesting that the lncSnhg7/miR-34a/GALNT7 is an important pathway in primary CD4+ T cells. These data highlight the need to understand the consequences of environmental exposures on lncRNA functions in non-cancerous cells as well as the effects in utero.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cadmium/adverse effects , Cell Proliferation/drug effects , Prenatal Exposure Delayed Effects/chemically induced , RNA, Long Noncoding/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , CD28 Antigens/genetics , CD3 Complex/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/genetics , Female , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
5.
Vaccines (Basel) ; 8(4)2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33153066

ABSTRACT

Whole cell vaccines are frequently the first generation of vaccines tested for pathogens and can inform the design of subsequent acellular or subunit vaccines. For respiratory pathogens, administration of vaccines at the mucosal surface can facilitate the generation of a localized mucosal immune response. Here, we examined the innate and vaccine-induced immune responses to infection by two respiratory pathogens: Bordetella pertussis and Pseudomonas aeruginosa. In a model of intranasal administration of whole cell vaccines (WCVs) with the adjuvant curdlan, we examined local and systemic immune responses following infection. These studies showed that intranasal vaccination with a WCV led to a reduction of the bacterial burden in the airways of animals infected with the respective pathogen. However, there were unique changes in the cytokines produced, cells recruited, and inflammation at the site of infection. Both mucosal vaccinations induced antibodies that bind the target pathogen, but linear regression and principal component analysis revealed that protection from these pathogens is not solely related to antibody titer. Protection from P. aeruginosa correlated to a reduction in lung weight, blood lymphocytes and neutrophils, and the cytokines IL-6, TNF-α, KC/GRO, and IL-10, and promotion of serum IgG antibodies and the cytokine IFN-γ in the lung. Protection from B. pertussis infection correlated strongly with increased anti-B-pertussis serum IgG antibodies. These findings reveal valuable correlates of protection for mucosal vaccination that can be used for further development of both B. pertussis and P. aeruginosa vaccines.

6.
Cell Rep ; 30(8): 2776-2790.e6, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32101751

ABSTRACT

TRAF-interacting protein with a forkhead-associated domain B (TIFAB) is implicated in myeloid malignancies with deletion of chromosome 5q. Employing a combination of proteomic and genetic approaches, we find that TIFAB regulates ubiquitin-specific peptidase 15 (USP15) ubiquitin hydrolase activity. Expression of TIFAB in hematopoietic stem/progenitor cells (HSPCs) permits USP15 signaling to substrates, including MDM2 and KEAP1, and mitigates p53 expression. Consequently, TIFAB-deficient HSPCs exhibit compromised USP15 signaling and are sensitized to hematopoietic stress by derepression of p53. In MLL-AF9 leukemia, deletion of TIFAB increases p53 signaling and correspondingly decreases leukemic cell function and development of leukemia. Restoring USP15 expression partially rescues the function of TIFAB-deficient MLL-AF9 cells. Conversely, elevated TIFAB represses p53, increases leukemic progenitor function, and correlates with MLL gene expression programs in leukemia patients. Our studies uncover a function of TIFAB as an effector of USP15 activity and rheostat of p53 signaling in stressed and malignant HSPCs.


Subject(s)
Hematopoiesis , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Catalytic Domain , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Binding , Stress, Physiological , Ubiquitin/metabolism , Ubiquitin-Specific Proteases/chemistry , Ubiquitination
7.
Front Immunol ; 10: 2497, 2019.
Article in English | MEDLINE | ID: mdl-31708925

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen causing acute and chronic respiratory infections associated with morbidity and mortality, especially in patients with cystic fibrosis. Vaccination against P. aeruginosa before colonization may be a solution against these infections and improve the quality of life of at-risk patients. To develop a vaccine against P. aeruginosa, we formulated a novel peptide-based P. aeruginosa subunit vaccine based on the extracellular regions of one of its major siderophore receptors, FpvA. We evaluated the effectiveness and immunogenicity of the FpvA peptides conjugated to keyhole limpet hemocyanin (KLH) with the adjuvant curdlan in a murine vaccination and challenge model. Immunization with the FpvA-KLH vaccine decreased the bacterial burden and lung edema after P. aeruginosa challenge. Vaccination with FpvA-KLH lead to antigen-specific IgG and IgM antibodies in sera, and IgA antibodies in lung supernatant. FpvA-KLH immunized mice had an increase in recruitment of CD11b+ dendritic cells as well as resident memory CD4+ T cells in the lungs compared to non-vaccinated challenged mice. Splenocytes isolated from vaccinated animals showed that the FpvA-KLH vaccine with the adjuvant curdlan induces antigen-specific IL-17 production and leads to a Th17 type of immune response. These results indicate that the intranasal FpvA-KLH conjugate vaccine can elicit both mucosal and systemic immune responses. These observations suggest that the intranasal peptide-based FpvA-KLH conjugate vaccine with curdlan is a potential vaccine candidate against P. aeruginosa pneumonia.


Subject(s)
Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/immunology , Vaccines, Conjugate/immunology , Vaccines, Subunit/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Hemocyanins/chemistry , Hemocyanins/immunology , Humans , Immunity, Mucosal , Immunization , Memory, Short-Term , Mice , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Pseudomonas Vaccines/administration & dosage , Recombinant Proteins , Vaccines, Conjugate/administration & dosage , Vaccines, Subunit/administration & dosage
8.
NPJ Vaccines ; 4: 40, 2019.
Article in English | MEDLINE | ID: mdl-31602318

ABSTRACT

Current acellular pertussis vaccines fall short of optimal protection against the human respiratory pathogen Bordetella pertussis resulting in increased incidence of a previously controlled vaccine- preventable disease. Natural infection is known to induce a protective mucosal immunity. Therefore, in this study, we aimed to use acellular pertussis vaccines to recapitulate these mucosal immune responses. We utilized a murine immunization and challenge model to characterize the efficacy of intranasal immunization (IN) with DTaP vaccine or DTaP vaccine supplemented with curdlan, a known Th1/Th17 promoting adjuvant. Protection from IN delivered DTaP was compared to protection mediated by intraperitoneal injection of DTaP and whole-cell pertussis vaccines. We tracked fluorescently labeled DTaP after immunization and detected that DTaP localized preferentially in the lungs while DTaP with curdlan was predominantly in the nasal turbinates. IN immunization with DTaP, with or without curdlan adjuvant, resulted in anti-B. pertussis and anti-pertussis toxin IgG titers at the same level as intraperitoneally administered DTaP. IN immunization was able to protect against B. pertussis challenge and we observed decreased pulmonary pro-inflammatory cytokines, neutrophil infiltrates in the lung, and bacterial burden in the upper and lower respiratory tract at day 3 post challenge. Furthermore, IN immunization with DTaP triggered mucosal immune responses such as production of B. pertussis-specific IgA, and increased IL-17A. Together, the induction of a mucosal immune response and humoral antibody-mediated protection associated with an IN administered DTaP and curdlan adjuvant warrant further exploration as a pertussis vaccine candidate formulation.

9.
Front Immunol ; 9: 2376, 2018.
Article in English | MEDLINE | ID: mdl-30405604

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) compartments are altered to direct immune responses to infection. Their roles during immunization are not well-described. To elucidate mechanisms for waning immunity following immunization with acellular vaccines (ACVs) against Bordetella pertussis (Bp), we tested the hypothesis that immunization with Bp ACVs and whole cell vaccines (WCVs) differ in directing the HSPC characteristics and immune cell development patterns that ultimately contribute to the types and quantities of cells produced to fight infection. Our data demonstrate that compared to control and ACV-immunized CD-1 mice, immunization with an efficacious WCV drives expansion of hematopoietic multipotent progenitor cells (MPPs), increases circulating white blood cells (WBCs), and alters the size and composition of lymphoid organs. In addition to MPPs, common lymphoid progenitor (CLP) proportions increase in the bone marrow of WCV-immunized mice, while B220+ cell proportions decrease. Upon subsequent infection, increases in maturing B cell populations are striking in WCV-immunized mice. RNAseq analyses of HSPCs revealed that WCV and ACV-immunized mice vastly differ in developing VDJ gene segment diversity. Moreover, gene set enrichment analyses demonstrate WCV-immunized mice exhibit unique gene signatures that suggest roles for interferon (IFN) induced gene expression. Also observed in naïve infection, these IFN stimulated gene (ISG) signatures point toward roles in cell survival, cell cycle, autophagy, and antigen processing and presentation. Taken together, these findings underscore the impact of vaccine antigen and adjuvant content on skewing and/or priming HSPC populations for immune response.


Subject(s)
Bordetella pertussis/immunology , Hematopoietic Stem Cells/metabolism , Whooping Cough/immunology , Whooping Cough/microbiology , Animals , Bacterial Vaccines/immunology , Biomarkers , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Culture Techniques , Computational Biology/methods , Cytokines/metabolism , Female , Gene Expression Profiling , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , High-Throughput Nucleotide Sequencing , Immunization , Mice , V(D)J Recombination , Whooping Cough/metabolism
10.
Infect Immun ; 86(10)2018 10.
Article in English | MEDLINE | ID: mdl-30012638

ABSTRACT

Bordetella pertussis is the primary causative agent of pertussis (whooping cough), which is a respiratory infection that leads to a violent cough and can be fatal in infants. There is a need to develop more effective vaccines because of the resurgence of cases of pertussis in the United States since the switch from the whole-cell pertussis vaccines (wP) to the acellular pertussis vaccines (aP; diphtheria-tetanus-acellular-pertussis vaccine/tetanus-diphtheria-pertussis vaccine). Adenylate cyclase toxin (ACT) is a major virulence factor of B. pertussis that is (i) required for establishment of infection, (ii) an effective immunogen, and (iii) a protective antigen. The C-terminal repeats-in-toxin domain (RTX) of ACT is sufficient to induce production of toxin-neutralizing antibodies. In this study, we characterized the effectiveness of vaccines containing the RTX antigen against experimental murine infection with B. pertussis RTX was not protective as a single-antigen vaccine against B. pertussis challenge, and adding RTX to 1/5 human dose of aP did not enhance protection. Since the doses of aP used in murine studies are not proportionate to mouse/human body masses, we titrated the aP from 1/20 to 1/160 of the human dose. Mice receiving 1/80 human aP dose had bacterial burden comparable to those of naive controls. Adding RTX antigen to the 1/80 aP base resulted in enhanced bacterial clearance. Inclusion of RTX induced production of antibodies recognizing RTX, enhanced production of anti-pertussis toxin, decreased secretion of proinflammatory cytokines, such as interleukin-6, and decreased recruitment of total macrophages in the lung. This study shows that adding RTX antigen to an appropriate dose of aP can enhance protection against B. pertussis challenge in mice.


Subject(s)
Adenylyl Cyclases/immunology , Bordetella pertussis/immunology , Pertussis Vaccine/immunology , Toxoids/immunology , Whooping Cough/immunology , Adenylyl Cyclases/administration & dosage , Adenylyl Cyclases/genetics , Animals , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Bordetella pertussis/genetics , Drug Evaluation, Preclinical , Humans , Mice , Pertussis Vaccine/administration & dosage , Pertussis Vaccine/genetics , Toxoids/administration & dosage , Toxoids/genetics , Whooping Cough/microbiology
11.
J Exp Med ; 212(11): 1967-85, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26458771

ABSTRACT

TRAF-interacting protein with forkhead-associated domain B (TIFAB) is a haploinsufficient gene in del(5q) myelodysplastic syndrome (MDS). Deletion of Tifab results in progressive bone marrow (BM) and blood defects, including skewed hematopoietic stem/progenitor cell (HSPC) proportions and altered myeloid differentiation. A subset of mice transplanted with Tifab knockout (KO) HSPCs develop a BM failure with neutrophil dysplasia and cytopenia. In competitive transplants, Tifab KO HSPCs are out-competed by wild-type (WT) cells, suggesting a cell-intrinsic defect. Gene expression analysis of Tifab KO HSPCs identified dysregulation of immune-related signatures, and hypersensitivity to TLR4 stimulation. TIFAB forms a complex with TRAF6, a mediator of immune signaling, and reduces TRAF6 protein stability by a lysosome-dependent mechanism. In contrast, TIFAB loss increases TRAF6 protein and the dynamic range of TLR4 signaling, contributing to ineffective hematopoiesis. Moreover, combined deletion of TIFAB and miR-146a, two genes associated with del(5q) MDS/AML, results in a cooperative increase in TRAF6 expression and hematopoietic dysfunction. Re-expression of TIFAB in del(5q) MDS/AML cells results in attenuated TLR4 signaling and reduced viability. These findings underscore the importance of efficient regulation of innate immune/TRAF6 signaling within HSPCs by TIFAB, and its cooperation with miR-146a as it relates to the pathogenesis of hematopoietic malignancies, such as del(5q) MDS/AML.


Subject(s)
Hematopoiesis , Proteins/physiology , Signal Transduction/physiology , TNF Receptor-Associated Factor 6/physiology , Toll-Like Receptors/physiology , Animals , Apoptosis , Bone Marrow Transplantation , Cell Differentiation , Chromosomes, Human, Pair 5 , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , MicroRNAs/physiology , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Proteins/genetics
12.
Exp Hematol ; 43(8): 587-598, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26143580

ABSTRACT

Overexpression of immune-related genes is widely reported in myelodysplastic syndromes (MDSs), and chronic immune stimulation increases the risk for developing MDS. Aberrant innate immune activation, such as that caused by increased toll-like receptor (TLR) signaling, in MDS can contribute to systemic effects on hematopoiesis, in addition to cell-intrinsic defects on hematopoietic stem/progenitor cell (HSPC) function. This review will deconstruct aberrant function of TLR signaling mediators within MDS HSPCs that may contribute to cell-intrinsic consequences on hematopoiesis and disease pathogenesis. We will discuss the contribution of chronic TLR signaling to the pathogenesis of MDS based on evidence from patients and mouse genetic models.


Subject(s)
Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Immunity, Innate , Myelodysplastic Syndromes/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Animals , Disease Models, Animal , Hematopoiesis/genetics , Hematopoietic Stem Cells/pathology , Humans , Mice , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Signal Transduction/genetics , Toll-Like Receptors/genetics
13.
Haematologica ; 98(9): 1353-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23716557

ABSTRACT

mTOR integrates signals from nutrients and growth factors to control protein synthesis, cell growth, and survival. Although mTOR has been established as a therapeutic target in hematologic malignancies, its physiological role in regulating hematopoiesis remains unclear. Here we show that conditional gene targeting of mTOR causes bone marrow failure and defects in multi-lineage hematopoiesis including myelopoiesis, erythropoiesis, thrombopoiesis, and lymphopoiesis. mTOR deficiency results in loss of quiescence of hematopoietic stem cells, leading to a transient increase but long-term exhaustion and defective engraftment of hematopoietic stem cells in lethally irradiated recipient mice. Furthermore, ablation of mTOR causes increased apoptosis in lineage-committed blood cells but not hematopoietic stem cells, indicating a differentiation stage-specific function. These results demonstrate that mTOR is essential for hematopoietic stem cell engraftment and multi-lineage hematopoiesis.


Subject(s)
Gene Targeting/methods , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation/methods , TOR Serine-Threonine Kinases/physiology , Animals , Cell Survival/physiology , Cells, Cultured , Mice , Mice, Knockout , Mice, SCID
14.
Blood ; 120(4): 858-67, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22685174

ABSTRACT

Bortezomib (Velcade) is used widely for the treatment of various human cancers; however, its mechanisms of action are not fully understood, particularly in myeloid malignancies. Bortezomib is a selective and reversible inhibitor of the proteasome. Paradoxically, we find that bortezomib induces proteasome-independent degradation of the TRAF6 protein, but not mRNA, in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) cell lines and primary cells. The reduction in TRAF6 protein coincides with bortezomib-induced autophagy, and subsequently with apoptosis in MDS/AML cells. RNAi-mediated knockdown of TRAF6 sensitized bortezomib-sensitive and -resistant cell lines, underscoring the importance of TRAF6 in bortezomib-induced cytotoxicity. Bortezomib-resistant cells expressing an shRNA targeting TRAF6 were resensitized to the cytotoxic effects of bortezomib due to down-regulation of the proteasomal subunit α-1 (PSMA1). To determine the molecular consequences of loss of TRAF6 in MDS/AML cells, in the present study, we applied gene-expression profiling and identified an apoptosis gene signature. Knockdown of TRAF6 in MDS/AML cell lines or patient samples resulted in rapid apoptosis and impaired malignant hematopoietic stem/progenitor function. In summary, we describe herein novel mechanisms by which TRAF6 is regulated through bortezomib/autophagy-mediated degradation and by which it alters MDS/AML sensitivity to bortezomib by controlling PSMA1 expression.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Boronic Acids/pharmacology , Leukemia, Myeloid, Acute/pathology , Lysosomes/metabolism , Myelodysplastic Syndromes/pathology , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , TNF Receptor-Associated Factor 6/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Bortezomib , Cell Proliferation/drug effects , Clinical Trials, Phase II as Topic , Gene Expression Profiling , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Multicenter Studies as Topic , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Oligonucleotide Array Sequence Analysis , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors , TNF Receptor-Associated Factor 6/genetics
15.
Curr Pharm Des ; 18(22): 3170-9, 2012.
Article in English | MEDLINE | ID: mdl-22571695

ABSTRACT

MicroRNAs (miRNAs) are significant regulators of human hematopoietic stem cells (HSC), and their deregulation contributes to hematological malignancies. Myelodysplastic syndromes (MDS) represent a spectrum of hematological disorders characterized by dysfunctional HSC, ineffective blood cell production, progressive marrow failure, and an increased risk of developing acute myeloid leukemia (AML). Although miRNAs have been primarily studied in AML, only recently have similar studies been performed on MDS. In this review, we describe the normal function and expression of miRNAs in human HSC, and describe mounting evidence that deregulation of miRNAs contributes to the pathogenesis of MDS.


Subject(s)
Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Myelodysplastic Syndromes/genetics , Animals , Disease Progression , Down-Regulation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/etiology , MicroRNAs/metabolism , Myelodysplastic Syndromes/pathology , Risk
16.
Lipids ; 46(1): 47-57, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21038084

ABSTRACT

The effects of the polyunsaturated omega-3 (n-3) and omega-6 (n-6) fatty acids (FA) on hematopoiesis are complex in that both FA forms are processed into leukotrienes, eicosanoids, and prostaglandins, which can have independent effects. These FA have antagonistic effects in that n-6 FA prostaglandins tend to be pro-proliferative and pro-inflammatory, while the effects of n-3 FA prostaglandins are the opposite. We have previously shown that diets high in n-3 FA reduce the size of the middle to later stage myeloid progenitor compartment in FVB X sv129 F(1)hybrid mice. To assay the effects of high n-3 FA diets on earlier stages of myelopoiesis, we fed C57BL/6J mice diets high in n-3 FA or levels of n-3/n-6 FA similar to western diets and assayed the effects on myelopoiesis with flow cytometry and colony forming cell assays. Results indicate an expansion of the common myeloid progenitor cell compartment in high n-3 FA diets, which does not persist into later stages where the number of progenitor cells is actually lower in high n-3 FA fed animals. Investigations in vitro with the hematopoietic stem cell line EML-clone 1 indicate that cells cultured with eicosapentaenoic acid (n-3 FA) or arachidonic acid (n-6 FA) have no differences in cell viability but that arachidonic acid more rapidly produces progenitors with low levels of the macrophage developmental marker, F4/80.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Myeloid Progenitor Cells/drug effects , Animals , Antigens, Differentiation/metabolism , Arachidonic Acid/metabolism , Cell Differentiation , Diet , Eicosapentaenoic Acid/metabolism , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6/metabolism , Flow Cytometry , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Myelopoiesis/physiology , Phenotype
17.
Pigment Cell Melanoma Res ; 23(3): 419-29, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20374520

ABSTRACT

Many melanoma cells are resistant to the anti-proliferative effect of all trans retinoic acid (ATRA). Retinoic Acid Receptor-beta2 (RAR-beta2) mediates the ATRA growth inhibition. We found a correlation between the anti-proliferative activity of ATRA and expression of RAR-beta2. There was not a strict correlation between DNA methylation of RAR-beta gene and its expression. There was no difference in global and RARbeta specific nucleosome repeat length (NRL) in melanoma and melanocytes or between control and ATRA treated cells. Pan-acetylation of H3 and H4 within the RAR-beta gene promoter was higher in cells expressing RAR-beta2. All trans retinoic acid treatment of responsive cells did not change pan-acetylation of H3/H4, but addition of ATRA to non-responsive cells increased H4 pan-acetylation. Phytochemicals or the histone deacetylase inhibitor Trichostatin A did not restore expression of RAR-beta2. Treatment of WM1366 melanoma cells with 5-aza 2'-deoxycytidine reactivated RAR-beta2 gene expression and restored the ability of ATRA to further induce the expression of this gene. Therefore, promoter methylation is responsible for silencing of RAR-beta2 in some melanoma cells and pan-acetylation of H3 likely plays a permissive role in expression of RAR-beta2.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Silencing , Melanoma/genetics , Receptors, Retinoic Acid/genetics , Skin Neoplasms/genetics , Acetylation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , DNA Methylation/drug effects , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Histones/metabolism , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/pathology , Nucleosomes/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Retinoic Acid/metabolism , Skin Neoplasms/pathology , Tretinoin/pharmacology
18.
Lipids Health Dis ; 8: 9, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19296839

ABSTRACT

BACKGROUND: Omega 3 fatty acids have been found to inhibit proliferation, induce apoptosis, and promote differentiation in various cell types. The processes of cell survival, expansion, and differentiation are of key importance in the regulation of hematopoiesis. We investigated the role of omega 3 fatty acids in controlling the frequency of various myeloid progenitor cells in the bone marrow of mice. Increased progenitor cell frequency and blocked differentiation are characteristics of hematopoietic disorders of the myeloid lineage, such as myeloproliferative diseases and myeloid leukemias. RESULTS: We found that increasing the proportion of omega 3 fatty acids relative to the proportion of omega 6 fatty acids in the diet caused increased differentiation and reduced the frequency of myeloid progenitor cells in the bone marrow of mice. Furthermore, this had no adverse effect on peripheral white blood cell counts. CONCLUSION: Our results indicate that omega 3 fatty acids impact hematopoietic differentiation by reducing myeloid progenitor cell frequency in the bone marrow and promoting progenitor cell differentiation. Further exploration of this discovery could lead to the use of omega 3 fatty acids as a therapeutic option for patients that have various disorders of hematopoiesis.


Subject(s)
Cell Differentiation/drug effects , Fatty Acids, Omega-3/pharmacology , Myeloid Progenitor Cells/drug effects , Animals , Bone Marrow , Bone Marrow Cells , Cell Count , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-6 , Hematopoiesis , Mice , Myeloid Progenitor Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...