Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Biol Blood Marrow Transplant ; 26(9): e227-e231, 2020 09.
Article in English | MEDLINE | ID: mdl-32592856

ABSTRACT

The emotional and physical toll on caregivers of cancer patients is well documented, but research evaluating the financial burdens and time commitments of caregivers is limited. We suspected that the rural location of our cancer center would intensify these burdens for caregivers. We conducted a prospective trial to assess the out-of-pocket expenses and time commitment of caregivers of hematopoietic stem cell transplantation recipients within the first 4 weeks after discharge from the hospital from a National Cancer Institute (NCI)-designated comprehensive cancer center. These results show that caregivers of autologous recipients paid out-of-pocket expenses of $196 over 4 weeks. If lost wages were included, the expenses increased to $736 during this period. Caregivers of allogeneic recipients had out-of-pocket expenses of $110 in 4 weeks, or a total of $610 when lost wages were included. In the month after discharge from the hospital, caregivers traveled a median distance of 450 miles or 560 miles, depending on whether the patient received an autologous transplant or an allogeneic transplant, respectively. These results demonstrate a compelling need to address caregiver support, given the significant financial out-of-pocket expenses and time commitment.


Subject(s)
Hematopoietic Stem Cell Transplantation , Neoplasms , Caregivers , Health Expenditures , Humans , Neoplasms/therapy , Prospective Studies , Transplantation, Autologous
2.
Blood ; 127(11): 1384-5, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26989189

ABSTRACT

One of the major goals of hemoglobinopathy research has been to devise improved pharmacologic strategies for the induction of fetal hemoglobin (HbF) in people with sickle cell disease and ß-thalassemia. In this issue of Blood, Dulmovits and colleagues report that pomalidomide, a drug approved by the US Food and Drug Administration (FDA) for treatment of multiple myeloma, induces HbF production by decreasing levels of several key transcriptional repressors of fetal globin gene expression. In addition, they show that pomalidomide induces HbF in differentiating erythroid cells from people with sickle cell disease and in myeloma patients.


Subject(s)
Hematopoietic Stem Cells/drug effects , Thalidomide/analogs & derivatives , Transcription, Genetic/drug effects , gamma-Globins/genetics , Humans
3.
Blood ; 124(17): 2730-4, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25170120

ABSTRACT

Fetal hemoglobin (HbF) induction can ameliorate the clinical severity of sickle cell disease and ß-thalassemia. We previously reported that activation of the eukaryotic initiation factor 2α (eIF2α) stress pathway increased HbF through a posttranscriptional mechanism. In this study, we explored the underlying means by which salubrinal, an activator of eIF2α signaling, enhances HbF production in primary human erythroid cells. Initial experiments eliminated changes in globin messenger RNA (mRNA) stability or cellular location and reduction of adult hemoglobin as possible salubrinal mechanisms. We then determined that salubrinal selectively increased the number of actively translating ribosomes on γ-globin mRNA. This enhanced translation efficiency occurred in the recovery phase of the stress response as phosphorylation of eIF2α and global protein synthesis returned toward baseline. These findings highlight γ-globin mRNA translation as a novel mechanism for regulating HbF production and as a pharmacologic target for induction of HbF.


Subject(s)
Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Protein Biosynthesis , gamma-Globins/genetics , Blotting, Western , Cinnamates/pharmacology , Erythroid Cells/drug effects , Eukaryotic Initiation Factor-2/pharmacology , Fetal Hemoglobin/metabolism , Humans , K562 Cells , Phosphorylation/drug effects , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thiourea/analogs & derivatives , Thiourea/pharmacology , gamma-Globins/metabolism
4.
Blood ; 122(4): 477-85, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23690448

ABSTRACT

Strategies to increase fetal hemoglobin (HbF) levels can ameliorate symptoms and improve the lives of ß-hemoglobinopathy patients. Although most studies have focused on induction of γ-globin gene expression as an approach to induce HbF, we hypothesized that post-transcriptional regulation of HbF plays an underappreciated yet important role in controlling HbF levels. In the present study, we investigated whether increasing eukaryotic initiation factor 2α (eIF2α) phosphorylation, a key regulator of protein translation, could enhance HbF post-transcriptionally in human primary erythroid cells. Initial analysis using a known inhibitor of eIF2α dephosphorylation, salubrinal, revealed that elevated eIF2α phosphorylation enhanced HbF production without changing globin gene expression, proliferation, or cell differentiation. These results were further supported by the post-transcriptional induction of HbF by other pharmacologic activators of the eIF2α pathway and by genetic inactivation of the negative regulators, GADD34 and CReP. Additionally, we found that this novel mechanism of increasing HbF could be combined with clinically relevant transcriptional activators of γ-globin gene expression to additively enhance HbF. Taken together, these findings identify eIF2α phosphorylation as a post-transcriptional regulator of HbF induction that may be pharmacologically targeted, either alone or in combination, in ß-hemoglobinopathy patients.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Fetal Hemoglobin/metabolism , Cinnamates/pharmacology , Enzyme Inhibitors/pharmacology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Eukaryotic Initiation Factor-2/physiology , Hemoglobinopathies/genetics , Hemoglobinopathies/metabolism , Hemoglobinopathies/therapy , Humans , K562 Cells , Models, Biological , Molecular Targeted Therapy/methods , Phosphorylation/physiology , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Transcription, Genetic
5.
Blood ; 121(5): 830-9, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23223429

ABSTRACT

UNLABELLED: Although increased fetal hemoglobin (HbF) levels have proven benefit for people with ß-hemoglobinopathies, all current HbF-inducing agents have limitations. We previously reported that drugs that activate the NRF2 antioxidant response signaling pathway increase HbF in primary human erythroid cells. In an attempt to increase HbF levels achieved with NRF2 activators, in the present study, we investigated potential complementary activity between these agents and HMG-CoA reductase inhibitors (statins) based on their ability to induce KLF2 protein levels. Experiments in K562 cells showed that simvastatin increased KLF2 mRNA and protein and KLF2 binding to HS2 of the ß-globin locus control region and enhanced -globin mRNA production by the NRF2 activator Tert-butylhydroquinone (tBHQ). When tested in differentiating primary human erythroid cells, simvastatin induced HbF alone and additively with tBHQ, but it did not increase KLF2 mRNA or locus control region binding above levels seen with normal differentiation. Investigating alternative mechanisms of action, we found that both simvastatin and tBHQ suppress ß-globin mRNA and KLF1 and BCL11A mRNA and protein, similar to what is seen in people with an HPFH phenotype because of KLF1 haploinsufficiency. These findings identify statins as a potential class of HbF-inducing agents and suggest a novel mechanism of action based on pharmacologic suppression of KLF1 and BCL11A gene expression. KEY POINTS: Simvastatin and tBHQ suppress KLF1 and BCL11 gene expression and additively increase fetal hemoglobin in primary human erythroid cells. Because both drugs are FDA-approved, these findings could lead to clinical trials in the relatively near future.


Subject(s)
Carrier Proteins/biosynthesis , Erythroid Cells/metabolism , Fetal Hemoglobin/biosynthesis , Gene Expression Regulation/drug effects , Hydroquinones/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kruppel-Like Transcription Factors/biosynthesis , Nuclear Proteins/biosynthesis , Simvastatin/pharmacology , Carrier Proteins/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Erythroid Cells/cytology , Female , Fetal Hemoglobin/genetics , Gene Expression Regulation/genetics , Genetic Loci/physiology , Humans , K562 Cells , Kruppel-Like Transcription Factors/genetics , Male , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins
6.
Blood Cells Mol Dis ; 52(4): 214-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24314748

ABSTRACT

Increased fetal hemoglobin (HbF) expression is beneficial for ß-hemoglobinopathy patients; however, current inducing agents do not possess the ideal combination of efficacy, safety and ease of use. Better understanding the mechanisms involved in γ-globin gene induction is critical for designing improved therapies, as no complete mechanism for any inducing agent has been identified. Given the cytotoxic nature of most known inducing drugs, we hypothesized that γ-globin is a cell stress response gene, and that induction occurs via activation of cell stress signaling pathways. We tested this hypothesis by investigating the ability of physical stresses including heat-shock (HS), UV- and X-irradiation and osmotic shock to increase γ-globin gene expression in erythroid cells. Experiments in K562 and KU812 cells showed that each of these stresses increased steady-state γ-globin mRNA levels, but only after 3-5days of treatments. HS and UV also increased γ-globin mRNA and HbF levels in differentiating primary human erythroid cells. Mechanistic studies showed that HS affects γ-globin mRNA at multiple levels, including nascent transcription and transcript stability, and that induction is dependent on neither the master regulator of the canonical HS response, HSF1, nor p38 MAPK. Inhibitor panel testing identified PI3K inhibitor LY294002 as a novel inducing agent and revealed potential roles for NFκB and VEGFR/PDGFR/Raf kinases in HS-mediated γ-globin gene induction. These findings suggest that cell stress signaling pathways play an important role in γ-globin gene induction and may provide novel targets for the pharmacologic induction of fetal hemoglobin.


Subject(s)
Erythroid Cells/metabolism , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Gene Expression Regulation , Stress, Physiological , gamma-Globins/biosynthesis , gamma-Globins/genetics , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Erythropoiesis/genetics , Gene Expression Regulation/drug effects , Heat Shock Transcription Factors , Hemoglobinopathies/genetics , Hot Temperature , Humans , K562 Cells , Osmotic Pressure , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Ultraviolet Rays , X-Rays , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Am J Hematol ; 89(3): 339-41, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24127129

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hematological disorder that is often suspected in a patient presenting with non-immune hemolytic anemia associated with pancytopenia or venous thrombosis. This disorder is a consequence of acquired somatic mutations in the phosphatidylinositol glycan class A (PIG-A) gene in the hematopoietic stem cells (HSC) of patients. The presence of these mutations leads to production of blood cells with decreased glycosyl phosphatidylinositol-anchored cell surface proteins, making red blood cells derived from the clone more sensitive to complement mediated hemolysis. The diagnosis of PNH may be difficult in some cases due a low proportion of PNH cells in the blood and occasionally due to difficulties in selecting the most appropriate diagnostic studies. The latest generation of tests allow for detection of very small populations of PNH cells, for following the natural course and response to therapy of the disease, and for helping to decide when to initiate therapy with monoclonal antibody targeting the terminal complement protein C5 (Eculizumab), anticoagulation, and in some cases allogeneic HSC transplant. In this article, we review the different diagnostic tests available to clinicians for PNH diagnosis.


Subject(s)
Bone Marrow Examination , DNA Mutational Analysis , Hemoglobinuria, Paroxysmal/diagnosis , Immunophenotyping/methods , CD24 Antigen/blood , CD59 Antigens/blood , Flow Cytometry/methods , Glycosylphosphatidylinositols/blood , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/genetics , Humans , Lipopolysaccharide Receptors/blood , Membrane Proteins/genetics
8.
Blood ; 117(22): 5987-97, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21464371

ABSTRACT

Although hematopoietic stem cell transplantation and gene therapy have the potential to cure ß-thalassemia and sickle cell disease, they are not currently available to most people with these diseases. In the near term, pharmacologic induction of fetal hemoglobin (HbF) may offer the best possibility for safe, effective, and widely available therapy. In an effort to define new pathways for targeted drug development for HbF induction, we evaluated the nuclear factor erythroid 2-related factor 2 (NRF2) antioxidant response element signaling pathway. We found that 3 well-known activators of this pathway increased γ-globin mRNA at nontoxic doses in K562 cells. Tert-butylhydroquinone (tBHQ), the most active of these compounds, increased cellular levels and nuclear translocation of NRF2 and binding of NRF2 to the γ-globin promoter. siRNA knockdown of NRF2 inhibited γ-globin induction by tBHQ. When tested in human primary erythroid cells, tBHQ induced NRF2 binding to the γ-globin promoter, increased γ-globin mRNA and HbF, and suppressed ß-globin mRNA and HbA, resulting in a > 3-fold increase in the percentage of HbF. These results suggest that drugs that activate the NRF2/antioxidant response element signaling pathway have the potential to induce therapeutic levels of HbF in people with ß-hemoglobinopathies.


Subject(s)
Antioxidants/therapeutic use , Fetal Hemoglobin/metabolism , Hydroquinones/therapeutic use , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , beta-Thalassemia/prevention & control , gamma-Globins/genetics , Blotting, Western , Cell Differentiation , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Erythroid Cells/metabolism , Humans , K562 Cells , Luciferases/metabolism , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Promoter Regions, Genetic , Protein Transport , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Response Elements , Reverse Transcriptase Polymerase Chain Reaction , beta-Globins , beta-Thalassemia/metabolism , beta-Thalassemia/pathology , gamma-Globins/metabolism
9.
Blood Cells Mol Dis ; 47(2): 107-16, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21641240

ABSTRACT

The ß-hemoglobinopathies and thalassemias are serious genetic blood disorders affecting the ß-globin chain of hemoglobin A (α(2)ß(Α)(2)). Their clinical severity can be reduced by enhancing expression of fetal hemoglobin (γ-globin), producing HbF (α(2)γ(2,)). In studies reported here, γ-globin induction by 23 novel, structurally-unrelated compounds, which had been predicted through molecular modeling and in silico screening of a 13,000 chemical library, was evaluated in vitro in erythroid progenitors cultured from normal subjects and ß-thalassemia patients, and in vivo in transgenic mice or anemic baboons. Four predicted candidates were found to have high potency, with 4- to 8-fold induction of HbF. Two of these compounds have pharmacokinetic profiles favorable for clinical application. These studies thus effectively identified high potency γ-globin inducing candidate therapeutics and validated the utility of in silico molecular modeling.


Subject(s)
Anemia/drug therapy , Biological Products/administration & dosage , Drug Design , Erythroid Precursor Cells/drug effects , Fetal Hemoglobin/biosynthesis , Small Molecule Libraries/administration & dosage , beta-Thalassemia/drug therapy , gamma-Globins/biosynthesis , Administration, Oral , Anemia/genetics , Anemia/metabolism , Animals , Biological Products/chemistry , Biological Products/therapeutic use , Cells, Cultured , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/genetics , Gene Expression , Humans , Injections, Intravenous , Mice , Mice, Transgenic , Models, Molecular , Papio , Phlebotomy , Polymerase Chain Reaction , RNA, Messenger/analysis , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , beta-Globins/deficiency , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , gamma-Globins/genetics
10.
Mol Cell Biol ; 27(13): 5047-54, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17452448

ABSTRACT

There is little evidence addressing the role of CpG methylation in transcriptional control of genes that do not contain CpG islands. This is reflected in the ongoing debate about whether CpG methylation merely suppresses retroelements or if it also plays a role in developmental and tissue-specific gene regulation. The genes of the beta-globin locus are an important model of mammalian developmental gene regulation and do not contain CpG islands. We have analyzed the methylation status of regions in the murine beta-like globin locus in uncultured primitive and definitive erythroblasts and other cultured primary and transformed cell types. A large ( approximately 20-kb) domain is hypomethylated only in primitive erythroid cells; it extends from the region just past the locus control region to before beta-major and encompasses the embryonic genes Ey, beta h1, and beta h0. Even retrotransposons in this region are hypomethylated in primitive erythroid cells. The existence of this large developmentally regulated domain of hypomethylation supports a mechanistic role for DNA methylation in developmental regulation of globin genes.


Subject(s)
CpG Islands/genetics , Embryo, Mammalian/metabolism , Erythrocytes/metabolism , Globins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cells, Cultured , DNA Methylation , Gene Expression Regulation , Locus Control Region/genetics , Mice , Models, Genetic , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repetitive Sequences, Nucleic Acid/genetics , Transcription, Genetic
11.
Exp Hematol ; 36(9): 1057-72, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18718415

ABSTRACT

A major goal of hemoglobinopathy research is to develop treatments that correct the underlying molecular defects responsible for sickle cell disease and beta-thalassemia. One approach to achieving this goal is the pharmacologic induction of fetal hemoglobin (HbF). This strategy is capable of inhibiting the polymerization of sickle hemoglobin and correcting the globin chain imbalance of beta-thalassemia. Despite this promise, none of the currently available HbF-inducing agents exhibit the combination of efficacy, safety, and convenience of use that would make them applicable to most patients. The recent success of targeted drug therapies for malignant diseases suggests that this approach could be effective for developing optimal HbF-inducing agents. A first step in applying this approach is the identification of specific molecular targets. However, while >70 HbF-inducing agents have been described, neither molecular mechanisms nor target molecules have been definitively verified for any of these compounds. To help focus investigation in this area, we have reviewed known HbF-inducing agents and their proposed mechanisms of action. We find that in many cases, current models inadequately explain key experimental results. By integrating features of the erythropoietic stress model of HbF induction with data from recent intracellular signaling experiments, we have developed a new model that has the potential to explain several findings that are inconsistent with previous models and to unify most HbF-inducing agents under a common mechanism: cell stress signaling. If correct, this or related models could lead to new opportunities for development of targeted therapies for the beta-hemoglobinopathies.


Subject(s)
Erythrocytes/metabolism , Erythropoiesis/physiology , Fetal Hemoglobin/biosynthesis , Gene Expression Regulation/physiology , Globins/biosynthesis , Hemoglobinopathies/drug therapy , Models, Genetic , Stress, Physiological/genetics , Adolescent , Animals , Butyrates/pharmacology , Butyrates/therapeutic use , Clinical Trials as Topic/statistics & numerical data , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Drug Delivery Systems , Drug Evaluation, Preclinical , Enzyme Inhibitors/therapeutic use , Erythrocytes/pathology , Erythropoiesis/drug effects , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , Globins/genetics , Hematopoietic Stem Cell Transplantation , Hemoglobinopathies/blood , Hemoglobinopathies/genetics , Hemoglobinopathies/physiopathology , Histone Deacetylase Inhibitors , Humans , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology , Stress, Physiological/physiopathology , p38 Mitogen-Activated Protein Kinases/physiology
12.
Blood Coagul Fibrinolysis ; 30(8): 419-422, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31483321

ABSTRACT

: Primary myelofibrosis (PMF) is a clonal hematopoietic stem cell disorder characterized by fibrosis of the marrow cavity, marked megakaryocyte atypia and progressive cytopenias. Although thrombosis predominates, bleeding is the primary manifestation in up to 20% of patients and may be life-threatening. In this report, we document restoration of megakaryocyte and platelet structure and function in PMF after allogeneic hematopoietic cell transplantation (HCT). A 59-year-old man presented with recurrent episodes of postoperative bleeding preceding a diagnosis of primary myelofibrosis (PMF). Platelet aggregation and secretion studies showed abnormal responses to all agonists tested (epinephrine, ADP, arachidonic acid, U46619, collagen, ristocetin) despite the presence of thrombocytosis. After an allogeneic HCT, platelet morphology and function studies were all normal. The pathophysiology of platelet dysfunction in myeloid neoplasia is not well understood but, as highlighted in our report, restoration of platelet function by HCT supports a clonal process involving an early hematopoietic progenitor cell.


Subject(s)
Blood Platelet Disorders/etiology , Hematopoietic Stem Cell Transplantation , Primary Myelofibrosis/etiology , Recovery of Function , Blood Platelet Disorders/therapy , Blood Platelets/drug effects , Blood Platelets/metabolism , Hemorrhage/therapy , Humans , Male , Middle Aged , Platelet Aggregation/drug effects , Primary Myelofibrosis/complications , Primary Myelofibrosis/therapy , Thrombocytosis , Transplantation, Homologous , Treatment Outcome
13.
Cancer Immunol Res ; 7(7): 1079-1090, 2019 07.
Article in English | MEDLINE | ID: mdl-31088847

ABSTRACT

Tumor hypoxia is a negative prognostic factor that is implicated in oncogenic signal activation, immune escape, and resistance to treatment. Identifying the mechanistic role of hypoxia in immune escape and resistance to immune-checkpoint inhibitors may aid the identification of therapeutic targets. We and others have shown that V-domain Ig suppressor of T-cell activation (VISTA), a negative checkpoint regulator in the B7 family, is highly expressed in the tumor microenvironment in tumor models and primary human cancers. In this study, we show that VISTA and HIF1α activity are correlated in a cohort of colorectal cancer patients. High VISTA expression was associated with worse overall survival. We used the CT26 colon cancer model to investigate the regulation of VISTA by hypoxia. Compared with less hypoxic tumor regions or draining lymph nodes, regions of profound hypoxia in the tumor microenvironment were associated with increased VISTA expression on tumor-infiltrating myeloid-derived suppressor cells (MDSC). Using chromatin immunoprecipitation and genetic silencing, we show that hypoxia-inducible factor (HIF)-1α binding to a conserved hypoxia response element in the VISTA promoter upregulated VISTA on myeloid cells. Further, antibody targeting or genetic ablation of VISTA under hypoxia relieved MDSC-mediated T-cell suppression, revealing VISTA as a mediator of MDSC function. Collectively, these data suggest that targeting VISTA may mitigate the deleterious effects of hypoxia on antitumor immunity.


Subject(s)
Adenocarcinoma/immunology , B7 Antigens/metabolism , Colorectal Neoplasms/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/physiopathology , Myeloid-Derived Suppressor Cells/immunology , Tumor Microenvironment/immunology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis , B7 Antigens/genetics , Case-Control Studies , Cell Proliferation , Cohort Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Prognosis , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured
14.
J Mol Biol ; 366(3): 737-44, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17196618

ABSTRACT

GATA-1 is a hematopoietic transcription factor expressed in erythroid, megakaryocytic, mast cell and eosinophil lineages. It is required for normal erythroid differentiation, the expression of erythroid-specific genes and for the establishment of an active chromatin structure throughout the beta-globin gene locus. GATA-1 is also necessary for the formation and function of the locus control region DNase I hypersensitive site (HS) core elements. To determine whether GATA-1 was sufficient to direct formation of the locus control region (LCR) and an erythroid pattern of gene expression, we expressed GATA-1 in the non-hematopoietic HeLa cell line that does not express other hematopoietic transcription factors but does express GATA-2, GATA-3, and GATA-6. We found that production of the GATA-1 protein resulted in the formation of LCR DNase I HSs 1-4 in their normal locations, and that histones became hyperacetylated within these regulatory elements. Transcription of several erythroid-specific genes was activated in HeLa cells expressing GATA-1, including those coding for alpha-globin, beta-globin, the erythropoietin receptor, the erythroid krüpple-like factor and p45 NF-E2. Despite increased expression of these genes at the mRNA level, their protein products were not detected. These results imply that GATA-1 is sufficient to direct chromatin structure reorganization within the beta-globin LCR and an erythroid pattern of gene expression in the absence of other hematopoietic transcription factors.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Erythroid Cells/metabolism , GATA1 Transcription Factor/genetics , Globins/genetics , Hematopoiesis/genetics , Locus Control Region/genetics , Acetylation , Animals , Cell Line , Deoxyribonucleases/metabolism , GATA1 Transcription Factor/metabolism , Globins/metabolism , HeLa Cells , Histones/metabolism , Humans , K562 Cells , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , NF-E2 Transcription Factor, p45 Subunit/genetics , NF-E2 Transcription Factor, p45 Subunit/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
J Community Support Oncol ; 13(9): 316-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26859751

ABSTRACT

BACKGROUND: About 1 in 7 of all hospitalized patients is readmitted within 30 days of discharge. The cost of readmissions is significant, with Medicare readmissions alone costing the health care system an estimated $28 billion a year. OBJECTIVE: To identify the rates of and causes for readmission within 100 days of patients receiving a hematopoietic stem cell transplant. METHODS: We performed a retrospective review of 235 consecutive transplant recipients (autologous, n = 144; allogeneic, n = 91) to determine rates and causes for readmission within 100 days of patients receiving a transplant. Medical records and hospital readmissions were reviewed for each patient. RESULTS: 36 allogeneic patients accounted for 56 readmissions. 23 autologous patients accounted for 26 readmissions. Autologous transplant recipients were most commonly readmitted for the development of a fever (n = 15 patients) or cardiopulmonary issues (n = 4). The most prevalent reasons for readmission in the allogeneic recipients included a fever (n = 21) or the development or exacerbation of graft-versus-host disease (n = 5). The readmission length of stay was 6 days (median range, 1-91 days) for allogeneic patients and 4 days (median range, 1-22 days) for autologous patients. There was no difference in survival between the readmitted and the non-readmitted cohorts (𝑃 = .55 for allogeneic patients; 𝑃 = .24 for autologous patients). Although allogeneic graft recipients demonstrated a higher readmission rate (39.6%) compared with autologous recipients (16%), none of the variables examined, including age, gender, performance status, diagnosis, remission status at the time of transplant, comorbidities, type of preparative chemotherapy regimen or donor type, identified patients at increased risk for readmission. LIMITATIONS: Variations in clinical care, physician practices, and patient characteristics need to be considered when examining readmission rates. Most of the allogeneic patient population included unrelated donor recipients (65%) who received nonmyeloablative conditioning regimens (81% of allogeneic recipients). These features may not be characteristic of other centers. CONCLUSIONS: In these high-risk patients, readmissions following a transplant are common. Enhanced predischarge education by nurses and pharmacists, along with ongoing outpatient education and rigorous outpatient follow-up through phone calls or social media may decrease readmission rates.

16.
Expert Opin Biol Ther ; 3(8): 1279-89, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14640954

ABSTRACT

Gene transfer involves the introduction of an engineered gene into a person's cells with the expectation that the protein expressed from the gene will produce a therapeutic benefit. Strategies based on this principle have led to the approval of > 600 clinical trials and enrollment of approximately 3500 subjects worldwide in attempts to treat diseases ranging from cancer to AIDS to cystic fibrosis. While gene therapy has met with limited success and still has many hurdles to overcome before it sees wide application, it may be useful as a defensive strategy against bioterrorism agents including infectious microbes and toxins. Although many defensive strategies are possible, immunological strategies are currently the most developed and are being actively applied to the development of strategies against several of the most virulent potential bio-weapons. While most of these strategies are not yet ready for human application, DNA-based vaccines appear to be among the most promising in the fight against bioterrorism.


Subject(s)
Bioterrorism/prevention & control , Gene Transfer Techniques , Immunotherapy/methods , Vaccines, DNA , Animals , Anthrax/prevention & control , Hemorrhagic Fevers, Viral/prevention & control , Humans , Plague/prevention & control , Smallpox/prevention & control , Tularemia/prevention & control
17.
Mol Cancer Ther ; 12(8): 1504-14, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23723123

ABSTRACT

Proteins of the BCL2 family provide a survival mechanism in many human malignancies, including chronic lymphocytic leukemia (CLL). The BCL2 inhibitor ABT-263 (navitoclax) is active in clinical trials for lymphoid malignancies, yet resistance is expected on the basis of preclinical models. We recently showed that vinblastine can dramatically sensitize several leukemia cell lines to ABT-737 (the experimental congener of ABT-263). The goal of these experiments was to determine the impact of vinblastine on ABT-737 sensitivity in CLL cells isolated from peripheral blood and to define the underlying mechanism. Freshly isolated CLL cells from 35 patients, as well as normal lymphocytes and platelets, were incubated with various microtubule-disrupting agents plus ABT-737 to assess sensitivity to the single agents and the combination. ABT-737 and vinblastine displayed a range of sensitivity as single agents, and vinblastine markedly sensitized all CLL samples to ABT-737 within six hours. Vinblastine potently induced the proapoptotic protein PMAIP1 (NOXA) in both time- and dose-dependent manner and this was required for the observed apoptosis. Combretastatin A4, which dissociates microtubules by binding to a different site, had the same effect, confirming that interaction of these agents with microtubules is the initial target. Similarly, vincristine and vinorelbine induced NOXA and enhanced CLL sensitivity to ABT-737. Furthermore, vinblastine plus ABT-737 overcame stroma-mediated resistance to ABT-737 alone. Apoptosis was induced with clinically achievable concentrations with no additional toxicity to normal lymphocytes or platelets. These results suggest that vinca alkaloids may improve the clinical efficacy of ABT-263 in patients with CLL.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biphenyl Compounds/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Sulfonamides/pharmacology , Vinblastine/pharmacology , Antineoplastic Agents, Phytogenic/toxicity , Apoptosis/drug effects , Apoptosis/genetics , Biphenyl Compounds/toxicity , Blood Platelets/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphocytes/drug effects , Nitrophenols/toxicity , Piperazines/pharmacology , Piperazines/toxicity , Prognosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Stromal Cells/metabolism , Sulfonamides/toxicity , Tubulin Modulators/pharmacology , Tubulin Modulators/toxicity , Vinblastine/toxicity
18.
Cancer Biol Ther ; 12(4): 314-25, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21768777

ABSTRACT

The efficacy of many chemotherapeutic agents can be attenuated by expression of the anti-apoptotic proteins Bcl-2, Bcl-X(L) and Mcl-1. Flavopiridol and dinaciclib are cyclin-dependent kinase 7 and 9 inhibitors that transcriptionally inhibit expression of Mcl-1. We have investigated the ability of flavopiridol and dinaciclib to sensitize a panel of leukemia cell lines to vinblastine and paclitaxel. Both drugs acutely sensitized most of the leukemia lines to vinblastine, with 100% apoptosis in 4 h. Furthermore, dinaciclib sensitized freshly isolated chronic lymphocytic leukemia cells to vinblastine. This rapid induction of apoptosis was attributed to vinblastine-mediated activation of JNK because (a) flavopiridol and dinaciclib failed to induce apoptosis when combined with non-JNK activating concentrations of vinblastine; (b) JNK inhibitors suppressed JNK activity and prevented apoptosis; (c) flavopiridol did not potentiate apoptosis induced by paclitaxel which does not activate JNK in these cells; and (d) Jurkat cells failed to activate JNK in response to vinblastine and were not sensitive to combinations of vinblastine and flavopiridol or dinaciclib. The rapid induction of apoptosis by this combination in multiple cell systems but not in normal lymphocytes provides justification for performing a clinical trial to assess the efficacy in patients.


Subject(s)
Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Flavonoids/pharmacology , Piperidines/pharmacology , Pyridinium Compounds/pharmacology , Vinblastine/pharmacology , Anthracenes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclic N-Oxides , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Drug Synergism , HL-60 Cells , Humans , Immunoblotting , Indolizines , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Jurkat Cells , Myeloid Cell Leukemia Sequence 1 Protein , Paclitaxel/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , U937 Cells , Cyclin-Dependent Kinase-Activating Kinase
19.
Mol Cancer Ther ; 9(4): 791-802, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20371726

ABSTRACT

Chemotherapeutic agents modify intracellular signaling that culminates in the inhibition of Bcl-2 family members and initiates apoptosis. Inhibition of the extracellular signal-regulated kinase by PD98059 dramatically accelerates vinblastine-mediated apoptosis in ML-1 leukemia with cells dying in 4 hours from all phases of the cell cycle. Inhibition of protein synthesis by cycloheximide also markedly accelerated vinblastine-induced apoptosis, showing that the proteins required for this acute apoptosis are constitutively expressed. Vinblastine induced the rapid induction of Mcl-1 that was inhibited by PD98059 and cycloheximide. No change in Bcl-2 or Bcl-X was observed. We hypothesize that ML-1 cells use Mcl-1 for protection from the rapid vinblastine-induced apoptosis. This was confirmed by targeting Mcl-1 with short hairpin RNA. We also investigated the response of 13 other leukemia and lymphoma cell lines and cells from seven chronic lymphocytic leukemia patients. Four cell lines and all chronic lymphocytic leukemia cells were killed in 6 hours by vinblastine alone. Two additional cell lines were sensitized to vinblastine by PD98059, which suppressed Mcl-1. This acute apoptosis either alone or in combination with PD98059 required vinblastine-mediated activation of c-Jun-NH(2)-terminal kinase. PD98059 did not suppress Mcl-1 in other cell lines whereas sorafenib did, but this did not sensitize the cells to vinblastine, suggesting that the acute apoptosis varies depending on which Bcl-2 protein mediates protection. Most of the cell lines were sensitized to vinblastine by cycloheximide, suggesting that inhibition of a short-lived protein in addition to Mcl-1 can acutely sensitize cells. These results suggest several clinical strategies that might provide an effective therapy for selected patients. Mol Cancer Ther; 9(4); 791-802. (c)2010 AACR.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Leukemia/pathology , Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Vinblastine/pharmacology , Cell Line, Tumor , Cycloheximide/pharmacology , Cytoprotection/drug effects , Drug Screening Assays, Antitumor , Flavonoids/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Leukemia/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma/enzymology , MAP Kinase Signaling System/drug effects , Myeloid Cell Leukemia Sequence 1 Protein
20.
Exp Hematol ; 37(7): 799-806.e4, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19460472

ABSTRACT

OBJECTIVE: The regulation of the beta-globin switch remains undetermined, and understanding this mechanism has important benefits for clinical and basic science. Histone modifications regulate gene expression and this study determines the presence of three important histone modifications across the beta-globin locus in erythroblasts with different beta-like globin-expression profiles. Understanding the chromatin associated with weak gamma gene expression in bone marrow cells is an important objective, with the goal of ultimately inducing postnatal expression of weak gamma-globin to cure beta-hemoglobinopathies. MATERIALS AND METHODS: These studies use uncultured primary fetal and bone marrow erythroblasts and human embryonic stem cell-derived primitive-like erythroblasts. Chromatin immunoprecipitation with antibodies against modified histones reveals DNA associated with such histones. Precipitated DNA is quantitated by real-time polymerase chain reaction for 40 sites across the locus. RESULTS: Distribution of histone modifications differs at each developmental stage. The most highly expressed genes at each stage are embedded within large domains of modifications associated with expression (acetylated histone H3 [H3ac] and dimethyl lysine 4 of histone H3 [H3K4me2]). Moderately expressed genes have H3ac and H3K4me2 in the immediate area around the gene. Dimethyl lysine 9 of histone H3 (H3K9me2), a mark associated with gene suppression, is present at the epsilon and gamma genes in bone marrow cells, suggesting active suppression of these genes. CONCLUSION: This study reveals complex patterns of histone modifications associated with highly expressed, moderately expressed, and unexpressed genes. Activation of gamma postnatally will likely require extensive modification of the histones in a large domain around the gamma genes.


Subject(s)
Histones/metabolism , beta-Globins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Humans , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL