Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Immunother ; 47(2): 49-53, 2024.
Article in English | MEDLINE | ID: mdl-37991241

ABSTRACT

Adoptive transfer of ex vivo expanded tumor-infiltrating lymphocytes (TILs) have produced long-term response in metastatic cancers. TILs have traditionally been expanded from surgically resected specimens. Ultrasound-guided core needle biopsy (CNB) is an alternative method that avoids the morbidity of surgery and have added benefits which may include patients not amenable to surgery as well as the potential to produce TILs from multiple lesions in the same patient. We assessed the ability to produce and expand TILs from primary triple-negative breast cancer tumors from CNB (n=7) and demonstrate comparable expansion, phenotype and cytokine secretion after phorbol myristate acetate-ionomycin stimulation to TILs expanded from surgery (n=6). T cell Receptor clonality and diversity were also comparable between the two cohorts throughout the TIL culture. CNB is a safe and feasible method to obtain tumor tissue for TIL generation in patients with triple-negative breast cancer.


Subject(s)
Immunotherapy, Adoptive , Triple Negative Breast Neoplasms , Humans , Biopsy, Large-Core Needle , Triple Negative Breast Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/pathology , Phenotype
2.
Front Immunol ; 14: 1200180, 2023.
Article in English | MEDLINE | ID: mdl-37415976

ABSTRACT

During the pandemic of severe respiratory distress syndrome coronavirus 2 (SARS-CoV2), many novel therapeutic modalities to treat Coronavirus 2019 induced disease (COVID-19) were explored. This study summarizes 195 clinical trials of advanced cell therapies targeting COVID-19 that were registered over the two years between January 2020 to December 2021. In addition, this work also analyzed the cell manufacturing and clinical delivery experience of 26 trials that published their outcomes by July 2022. Our demographic analysis found the highest number of cell therapy trials for COVID-19 was in United States, China, and Iran (N=53, 43, and 19, respectively), with the highest number per capita in Israel, Spain, Iran, Australia, and Sweden (N=0.641, 0.232, 0,223, 0.194, and 0.192 trials per million inhabitants). The leading cell types were multipotent mesenchymal stromal/stem cells (MSCs), natural killer (NK) cells, and mononuclear cells (MNCs), accounting for 72%, 9%, and 6% of the studies, respectively. There were 24 published clinical trials that reported on infusions of MSCs. A pooled analysis of these MSC studies found that MSCs provide a relative risk reduction for all-cause COVID-19 mortality of RR=0.63 (95% CI 0.46 to 0.85). This result corroborates previously published smaller meta-analyses, which suggested that MSC therapy demonstrated a clinical benefit for COVID-19 patients. The sources of the MSCs used in these studies and their manufacturing and clinical delivery methods were remarkably heterogeneous, with some predominance of perinatal tissue-derived products. Our results highlight the important role that cell therapy products may play as an adjunct therapy in the management of COVID-19 and its related complications, as well as the importance of controlling key manufacturing parameters to ensure comparability between studies. Thus, we support ongoing calls for a global registry of clinical studies with MSC products that could better link cell product manufacturing and delivery methods to clinical outcomes. Although advanced cell therapies may provide an important adjunct treatment for patients affected by COVID-19 in the near future, preventing pathology through vaccination still remains the best protection to date. We conducted a systematic review and meta-analysis of advanced cell therapy clinical trials as potential novel treatment for COVID-19 (resulting from SARS-CoV-2 coronavirus infection), including analysis of the global clinical trial landscape, published safety/efficacy outcomes (RR/OR), and details on cell product manufacturing and clinical delivery. This study had a 2-year observation interval from start of January 2020 to end of December 2021, including a follow-up period until end of July to identify published outcomes, which covers the most vivid period of clinical trial activity, and is also the longest observation period studied until today. In total, we identified 195 registered advanced cell therapy studies for COVID-19, employing 204 individual cell products. Leading registered trial activity was attributed to the USA, China, and Iran. Through the end of July 2022, 26 clinical trials were published, with 24 out of 26 articles employing intravenous infusions (IV) of mesenchymal stromal/stem cell (MSC) products. Most of the published trials were attributed to China and Iran. The cumulative results from the 24 published studies employing infusions of MSCs indicated an improved survival (RR=0.63 with 95% Confidence Interval 0.46 to 0.85). Our study is the most comprehensive systematic review and meta-analysis on cell therapy trials for COVID-19 conducted to date, clearly identifying the USA, China, and Iran as leading advanced cell therapy trial countries for COVID-19, with further strong contributions from Israel, Spain, Australia and Sweden. Although advanced cell therapies may provide an important adjunct treatment for patients affected by COVID-19 in the future, preventing pathology through vaccination remains the best protection.


Subject(s)
COVID-19 , Mesenchymal Stem Cell Transplantation , Humans , COVID-19/therapy , COVID-19/etiology , SARS-CoV-2 , RNA, Viral , Mesenchymal Stem Cell Transplantation/methods , Spain
3.
Cytotherapy ; 25(1): 20-32, 2023 01.
Article in English | MEDLINE | ID: mdl-36280438

ABSTRACT

BACKGROUND AIMS: The field of cell and gene therapy in oncology has moved rapidly since 2017 when the first cell and gene therapies, Kymriah followed by Yescarta, were approved by the Food and Drug Administration in the United States, followed by multiple other countries. Since those approvals, several new products have gone on to receive approval for additional indications. Meanwhile, efforts have been made to target different cancers, improve the logistics of delivery and reduce the cost associated with novel cell and gene therapies. Here, we highlight various cell and gene therapy-related technologies and advances that provide insight into how these new technologies will speed the translation of these therapies into the clinic. CONCLUSIONS: In this review, we provide a broad overview of the current state of cell and gene therapy-based approaches for cancer treatment - discussing various effector cell types and their sources, recent advances in both CAR and non-CAR genetic modifications, and highlighting a few promising approaches for increasing in vivo efficacy and persistence of therapeutic drug products.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Humans , Neoplasms/genetics , Neoplasms/therapy , Genetic Therapy , Gene Editing
4.
Mol Cancer ; 21(1): 219, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36514045

ABSTRACT

Adoptive cell therapy (ACT) using tumor infiltrating lymphocytes (TIL) is being studied in multiple tumor types. However, little is known about clonal cell expansion in vitro and persistence of the ACT product in vivo. We performed single-cell RNA and T-Cell Receptor (TCR) sequencing on serial blood and tumor samples from a patient undergoing ACT, who did not respond. We found that clonal expansion varied during preparation of the ACT product, and only one expanded clone was preserved in the ACT product. The TCR of the preserved clone which persisted and remained activated for five months was previously reported as specific for cytomegalovirus and had upregulation of granzyme family genes and genes associated with effector functions (HLA-DQB1, LAT, HLA-DQA1, and KLRD1). Clones that contracted during TIL preparation had features of exhaustion and apoptosis. At disease progression, all previously detected clonotypes were detected. New clonotypes appearing in blood or tumor at disease progression were enriched for genes associated with cytotoxicity or stemness (FGFBP2, GNLY, GZMH, GZMK, IL7R, SELL and KLF2), and these might be harnessed for alternative cellular therapy or cytokine therapy. In-depth single-cell analyses of serial samples from additional ACT-treated patients is warranted, and viral- versus tumor-specificity should be carefully analyzed.


Subject(s)
Melanoma , Humans , Melanoma/genetics , Lymphocytes, Tumor-Infiltrating/pathology , Receptors, Antigen, T-Cell/genetics , Single-Cell Analysis , Treatment Failure , Disease Progression , Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive
5.
Cell Metab ; 34(4): 595-614.e14, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35276062

ABSTRACT

Chimeric antigen receptor (CAR)-T cell-based immunotherapy for cancer and immunological diseases has made great strides, but it still faces multiple hurdles. Finding the right molecular targets to engineer T cells toward a desired function has broad implications for the armamentarium of T cell-centered therapies. Here, we developed a dead-guide RNA (dgRNA)-based CRISPR activation screen in primary CD8+ T cells and identified gain-of-function (GOF) targets for CAR-T engineering. Targeted knockin or overexpression of a lead target, PRODH2, enhanced CAR-T-based killing and in vivo efficacy in multiple cancer models. Transcriptomics and metabolomics in CAR-T cells revealed that augmenting PRODH2 expression reshaped broad and distinct gene expression and metabolic programs. Mitochondrial, metabolic, and immunological analyses showed that PRODH2 engineering enhances the metabolic and immune functions of CAR-T cells against cancer. Together, these findings provide a system for identification of GOF immune boosters and demonstrate PRODH2 as a target to enhance CAR-T efficacy.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , CD8-Positive T-Lymphocytes , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gain of Function Mutation , Humans , Proline , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism
7.
Cytotherapy ; 24(1): 27-31, 2022 01.
Article in English | MEDLINE | ID: mdl-34810083

ABSTRACT

There is considerable interest in the next generation of personalized medicine, especially cell and gene therapy products such as chimeric antigen receptor T cells (CAR-Ts). Unlike other small molecules or pharmacologic drugs, most existing cell or cell-based gene therapy products (CGTs) require apheresis collection of the patient or donor, subsequent manufacture of the product, and final shipment of the product to the clinical site for infusion. Whereas traditional pharmaceutical drugs have involved the drug sponsor and the clinical site and clinical pharmacy, this new manufacturing paradigm has evolved, in many cases, to include an apheresis center, a cell processing lab, the sponsor's manufacturing facility, and a clinical site with or without a pharmacy. Here we report the results of a survey of current practices handling investigational CGTs conducted by the Immuno-Gene Therapy committee of the International Society of Cell and Gene Therapy.


Subject(s)
Pharmacy , Receptors, Chimeric Antigen , Genetic Therapy , Hospitals , Humans , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/genetics
8.
Methods Mol Biol ; 2097: 329-339, 2020.
Article in English | MEDLINE | ID: mdl-31776936

ABSTRACT

Academic medicine, in general, serves a dual role in advancing the scientific field as well as providing the highest quality clinical care. In the last decade we have observed significant development of commercial cell and gene therapy products with rapid growth of the industry. Currently, hospital-based Good Manufacturing Practice (GMP) facilities, which are used to support primarily academic investigator-initiated clinical trials, are increasingly involved in interactions with industry.The purpose of this piece is to review the role of academic GMP facilities in development of cellular therapies. For the purpose of this review we will discuss manufacturing facilities located in hospitals or academic medical centers and compare them with commercial GMP facilities. Although the missions of academic and commercial GMP facilities are different, both are bound by industry standards and often engage in technology transfer with industry partners. Therefore, successful operation of an academic GMP facility requires striking a unique balance between commercial and academic priorities. Finally, we highlight some of the most challenging aspects of academic GMP facility operation and point to potential solutions.


Subject(s)
Cell- and Tissue-Based Therapy , Manufacturing and Industrial Facilities , Animals , Capacity Building , Guideline Adherence , Humans , Social Control, Formal
9.
J Immunol Methods ; 474: 112668, 2019 11.
Article in English | MEDLINE | ID: mdl-31525367

ABSTRACT

Cell-mediated cytotoxicity is a critical function of the immune system in mounting defense against pathogens and cancers. Current methods that allow direct evaluation of cell-mediated cytotoxicity suffer from a wide-range of drawbacks. Here, we present a novel strategy to measure cytotoxicity that is direct, sensitive, rapid, and highly adaptable. Moreover, it allows accurate measurement of viability of both target and effector cells. Target cells are fluorescently labeled with a non-toxic, cell-permeable dye that covalently binds to cell proteins, including nuclear proteins. The labeled target cells are incubated with effector cells to begin killing. Following the killing reaction, the cell mixture is incubated with another dye that specifically stains proteins of dead cells, including nuclear proteins. In the final step, cell nuclei are released by Triton X-100, and analyzed by flow cytometry. This results in four nuclear staining patterns that separate target and effector nuclei as well as nuclei of live and dead cells. Analyzing nuclei, instead of cells, greatly reduces flow cytometry errors caused by the presence of target-effector cell aggregates. Target killing time can often be reduced to 2 h and the assay can be done in a high throughput format. We have successfully validated this assay in a variety of cytotoxicity scenarios including those mediated by NK-92 cells, Chimeric Antigen Receptor (CAR)-T cells, and Tumor Infiltrating Lymphocytes (TIL). Therefore, this technique is broadly applicable, highly sensitive and easily administered, making it a powerful tool to assess immunotherapy-based, cell-mediated cytotoxicity.


Subject(s)
Cytotoxicity Tests, Immunologic/methods , Cytotoxicity, Immunologic , Flow Cytometry , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cell Nucleus/immunology , Cell Nucleus/pathology , High-Throughput Screening Assays , Humans , Immunotherapy, Adoptive , Male , Melanoma/immunology , Melanoma/pathology , Mice, Inbred C57BL , Predictive Value of Tests , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Reproducibility of Results , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Time Factors , Workflow
10.
Regen Med ; 14(4): 309-319, 2019 05.
Article in English | MEDLINE | ID: mdl-31070115

ABSTRACT

Aim: This is the first analysis of both clinical trials and published studies that employ umbilical cord mesenchymal stromal cells, for the decade 2007-2017. Materials & methods: Searching international databases, we found 178 registered trials and 98 publications. Results: Among the registered clinical trials, 20% have resulted in publications so far. Among the publications, 18% report safety and 74% report some form of improvement. Between 36 and 45% of the publications do not report aspects of the cell manufacturing, including isolation method, culture medium or number of culture passages. Conclusion: Analyses that link trials with publications can elucidate factors that promote study completion and publication. More full disclosure of cell manufacturing is needed to evaluate the efficacy of mesenchymal stromal cell isolated from umbilical cord tissue (UC-MSC) products.


Subject(s)
Clinical Trials as Topic , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Publications , Umbilical Cord/cytology , Cell Separation , Cells, Cultured , Geography , Humans
12.
Regen Med ; 12(8): 953-968, 2017 12.
Article in English | MEDLINE | ID: mdl-29139329

ABSTRACT

AIM: The first review of advanced cell therapy trials with perinatal cells. MATERIALS & METHODS: We compiled 281 clinical trials of advanced cell therapy with perinatal cells that were registered in 2005-2015. RESULTS: The most common cell source in these trials is cord blood, but the cell type that provides the mechanism of action in the majority of trials is mesenchymal stem/stromal cells. We analyze trends among the 15 parameters we compiled for these trials. CONCLUSION: Advanced cell therapy with perinatal cells is a new field that covers a wide range of diagnoses but where most of the trials are early Phase. Researchers in different countries tend to work with a preferred cell source and cell type.


Subject(s)
Cell- and Tissue-Based Therapy , Fetal Blood/cytology , Fetal Stem Cells/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Clinical Trials as Topic , Female , Humans , Pregnancy
13.
Transfusion ; 57(5): 1104-1106, 2017 05.
Article in English | MEDLINE | ID: mdl-28425600

Subject(s)
T-Lymphocytes , Humans
14.
Regen Med ; 7(6 Suppl): 50-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23210811

ABSTRACT

Gene therapy and cell therapy have followed similar roller coaster paths of rising public expectations and disappointment over the past two decades. There is now reason to believe that momentum in the field has reached the point where the successes will be more frequent. The use of gene-modified cells has opened new avenues for engineering desired cell properties, for the use of cells as vehicles for gene delivery, and for tracking cells and controlling cell persistence after transplantation. Some notable recent clinical developments in cellular engineering by gene transfer offer lessons on how the field has emerged, and hint at additional future clinical applications.


Subject(s)
Cell- and Tissue-Based Therapy , Genetic Therapy , Drug Resistance, Viral , Genetic Diseases, Inborn/immunology , Humans , Lymphocytes/immunology , Tropism
15.
Aging Cell ; 11(6): 949-59, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22812478

ABSTRACT

Upon aging, the number of hematopoietic stem cells (HSCs) in the bone marrow increases while their repopulation potential declines. Moreover, aged HSCs exhibit lineage bias in reconstitution experiments with an inclination toward myeloid at the expense of lymphoid potential. The adaptor protein Lnk is an important negative regulator of HSC homeostasis, as Lnk deficiency is associated with a 10-fold increase in HSC numbers in young mice. However, the age-related increase in functional HSC numbers found in wild-type HSCs was not observed in Lnk-deficient animals. Importantly, HSCs from aged Lnk null mice possess greatly enhanced self-renewal capacity and diminished exhaustion, as evidenced by serial transplant experiments. In addition, Lnk deficiency ameliorates the aging-associated lineage bias. Transcriptome analysis revealed that WT and Lnk-deficient HSCs share many aging-related changes in gene expression patterns. Nonetheless, Lnk null HSCs displayed altered expression of components in select signaling pathways with potential involvement in HSC self-renewal and aging. Taken together, these results suggest that loss of Lnk partially mitigates age-related HSC alterations.


Subject(s)
Aging/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Adaptor Proteins, Signal Transducing , Adult , Animals , Cell Count , Cell Proliferation , Flow Cytometry , Gene Expression Profiling , Genotype , Hematopoietic Stem Cells/cytology , Humans , Membrane Proteins , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Signal Transduction , Transcriptome
16.
J Clin Invest ; 122(6): 2079-91, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22546852

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) functions are governed by intricate signaling networks. The tyrosine kinase JAK2 plays an essential role in cytokine signaling during hematopoiesis. The adaptor protein LNK is a critical determinant of this process through its inhibitory interaction with JAK2, thereby limiting HSPC self-renewal. LNK deficiency promotes myeloproliferative neoplasm (MPN) development in mice, and LNK loss-of-function mutations are found in human MPNs, emphasizing its pivotal role in normal and malignant HSPCs. Here, we report the identification of 14-3-3 proteins as LNK binding partners. 14-3-3 interfered with the LNK-JAK2 interaction, thereby alleviating LNK inhibition of JAK2 signaling and cell proliferation. Binding of 14-3-3 required 2 previously unappreciated serine phosphorylation sites in LNK, and we found that their phosphorylation is mediated by glycogen synthase kinase 3 and PKA kinases. Mutations of these residues abrogated the interaction and augmented the growth inhibitory function of LNK. Conversely, forced 14-3-3 binding constrained LNK function. Furthermore, interaction with 14-3-3 sequestered LNK in the cytoplasm away from the plasma membrane-proximal JAK2. Importantly, bone marrow transplantation studies revealed an essential role for 14-3-3 in HSPC reconstitution that can be partially mitigated by LNK deficiency. We believe that, together, this work implicates 14-3-3 proteins as novel and positive HSPC regulators by impinging on the LNK/JAK2 pathway.


Subject(s)
14-3-3 Proteins/metabolism , Cell Proliferation , Hematopoietic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Janus Kinase 2/metabolism , Signal Transduction , 14-3-3 Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Transplantation , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Janus Kinase 2/genetics , Membrane Proteins , Mice , Mice, Knockout , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Phosphorylation/genetics , Transplantation, Homologous
17.
Blood ; 116(23): e128-38, 2010 Dec 02.
Article in English | MEDLINE | ID: mdl-20720187

ABSTRACT

MicroRNAs are small noncoding RNAs that regulate cellular development by interfering with mRNA stability and translation. We examined global microRNA expression during the differentiation of murine hematopoietic progenitors into megakaryocytes. Of 435 miRNAs analyzed, 13 were up-regulated and 81 were down-regulated. Many of these changes are consistent with miRNA profiling studies of human megakaryocytes and platelets, although new patterns also emerged. Among 7 conserved miRNAs that were up-regulated most strongly in murine megakaryocytes, 6 were also induced in the related erythroid lineage. MiR-146a was strongly up-regulated during mouse and human megakaryopoiesis but not erythropoiesis. However, overexpression of miR-146a in mouse bone marrow hematopoietic progenitor populations produced no detectable alterations in megakaryocyte development or platelet production in vivo or in colony assays. Our findings extend the repertoire of differentially regulated miRNAs during murine megakaryopoiesis and provide a useful new dataset for hematopoiesis research. In addition, we show that enforced hematopoietic expression of miR-146a has minimal effects on megakaryopoiesis. These results are compatible with prior studies indicating that miR-146a inhibits megakaryocyte production indirectly by suppressing inflammatory cytokine production from innate immune cells, but cast doubt on a different study, which suggests that this miRNA inhibits megakaryopoiesis cell-autonomously.


Subject(s)
Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , MicroRNAs/biosynthesis , Thrombopoiesis/genetics , Animals , Cell Differentiation/genetics , Cell Separation , Flow Cytometry , Gene Expression Profiling , Humans , Mice , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
18.
J Clin Invest ; 120(6): 2058-69, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20458146

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) expansion is regulated by intrinsic signaling pathways activated by cytokines. The intracellular kinase JAK2 plays an essential role in cytokine signaling, and activating mutations in JAK2 are found in a number of hematologic malignancies. We previously demonstrated that lymphocyte adaptor protein (Lnk, also known as Sh2b3) binds JAK2 and attenuates its activity, thereby limiting HSPC expansion. Here we show that loss of Lnk accelerates and exacerbates oncogenic JAK2-induced myeloproliferative diseases (MPDs) in mice. Specifically, Lnk deficiency enhanced cytokine-independent JAK/STAT signaling and augmented the ability of oncogenic JAK2 to expand myeloid progenitors in vitro and in vivo. An activated form of JAK2, unable to bind Lnk, caused greater myeloid expansion than activated JAK2 alone and accelerated myelofibrosis, indicating that Lnk directly inhibits oncogenic JAK2 in constraining MPD development. In addition, Lnk deficiency cooperated with the BCR/ABL oncogene, the product of which does not directly interact with or depend on JAK2 or Lnk, in chronic myeloid leukemia (CML) development, suggesting that Lnk also acts through endogenous pathways to constrain HSPCs. Consistent with this idea, aged Lnk-/- mice spontaneously developed a CML-like MPD. Taken together, our data establish Lnk as a bona fide suppressor of MPD in mice and raise the possibility that Lnk dysfunction contributes to the development of hematologic malignancies in humans.


Subject(s)
Hematopoietic Stem Cells/metabolism , Janus Kinase 2/metabolism , Myeloproliferative Disorders/genetics , Phosphotransferases/metabolism , Signal Transduction/genetics , Animals , Cytokines/genetics , Cytokines/metabolism , Janus Kinase 2/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Knockout , Mutation , Phosphotransferases/genetics
19.
Blood ; 115(11): 2156-66, 2010 Mar 18.
Article in English | MEDLINE | ID: mdl-20065294

ABSTRACT

Nuclear factors regulate the development of complex tissues by promoting the formation of one cell lineage over another. The cofactor FOG1 interacts with transcription factors GATA1 and GATA2 to control erythroid and megakaryocyte (MK) differentiation. In contrast, FOG1 antagonizes the ability of GATA factors to promote mast cell (MC) development. Normal FOG1 function in late-stage erythroid cells and MK requires interaction with the chromatin remodeling complex NuRD. Here, we report that mice in which the FOG1/NuRD interaction is disrupted (Fog(ki/ki)) produce MK-erythroid progenitors that give rise to significantly fewer and less mature MK and erythroid colonies in vitro while retaining multilineage capacity, capable of generating MCs and other myeloid lineage cells. Gene expression profiling of Fog(ki/ki) MK-erythroid progenitors revealed inappropriate expression of several MC-specific genes. Strikingly, aberrant MC gene expression persisted in mature Fog(ki/ki) MK and erythroid progeny. Using a GATA1-dependent committed erythroid cell line, select MC genes were found to be occupied by NuRD, suggesting a direct mechanism of repression. Together, these observations suggest that a simple heritable silencing mechanism is insufficient to permanently repress MC genes. Instead, the continuous presence of GATA1, FOG1, and NuRD is required to maintain lineage fidelity throughout MK-erythroid ontogeny.


Subject(s)
Cell Compartmentation , Cell Lineage , Erythroid Cells/cytology , Hematopoiesis , Megakaryocytes/cytology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Erythroid Cells/enzymology , Erythropoiesis , GATA1 Transcription Factor/metabolism , GATA2 Transcription Factor/metabolism , Gene Expression Regulation , Gene Knock-In Techniques , Mast Cells/metabolism , Megakaryocyte-Erythroid Progenitor Cells/cytology , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Megakaryocytes/enzymology , Mice , Nuclear Proteins/genetics , Organ Specificity , Repressor Proteins/metabolism , Spleen/cytology , Transcription Factors/genetics , Transcription, Genetic
20.
J Clin Invest ; 119(12): 3519-29, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19959876

ABSTRACT

Hematopoietic stem cell (HSC) homeostasis depends on the balance between self renewal and lineage commitment, but what regulates this decision is not well understood. Using loss-of-function approaches in mice, we found that glycogen synthase kinase-3 (Gsk3) plays a pivotal role in controlling the decision between self renewal and differentiation of HSCs. Disruption of Gsk3 in BM transiently expanded phenotypic HSCs in a betta-catenin-dependent manner, consistent with a role for Wnt signaling in HSC homeostasis. However, in assays of long-term HSC function, disruption of Gsk3 progressively depleted HSCs through activation of mammalian target of rapamycin (mTOR). This long-term HSC depletion was prevented by mTOR inhibition and exacerbated by betta-catenin knockout. Thus, GSK-3 regulated both Wnt and mTOR signaling in mouse HSCs, with these pathways promoting HSC self renewal and lineage commitment, respectively, such that inhibition of Gsk3 in the presence of rapamycin expanded the HSC pool in vivo. These findings identify unexpected functions for GSK-3 in mouse HSC homeostasis, suggest a therapeutic approach to expand HSCs in vivo using currently available medications that target GSK-3 and mTOR, and provide a compelling explanation for the clinically prevalent hematopoietic effects observed in individuals prescribed the GSK-3 inhibitor lithium.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Hematopoietic Stem Cells/enzymology , Animals , Bone Marrow Transplantation , Carrier Proteins/metabolism , Enzyme Inhibitors/pharmacology , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3/genetics , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Homeostasis , Humans , Lithium/pharmacology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Wnt Proteins/metabolism , beta Catenin/deficiency , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...