Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Sci Adv ; 10(5): eadj9479, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38295180

ABSTRACT

Folate, an essential vitamin, is a one-carbon acceptor and donor in key metabolic reactions. Erythroid cells harbor a unique sensitivity to folate deprivation, as revealed by the primary pathological manifestation of nutritional folate deprivation: megaloblastic anemia. To study this metabolic sensitivity, we applied mild folate depletion to human and mouse erythroid cell lines and primary murine erythroid progenitors. We show that folate depletion induces early blockade of purine synthesis and accumulation of the purine synthesis intermediate and signaling molecule, 5'-phosphoribosyl-5-aminoimidazole-4-carboxamide (AICAR), followed by enhanced heme metabolism, hemoglobin synthesis, and erythroid differentiation. This is phenocopied by inhibition of folate metabolism using the inhibitor SHIN1, and by AICAR supplementation. Mechanistically, the metabolically driven differentiation is independent of mechanistic target of rapamycin complex 1 (mTORC1) and adenosine 5'-monophosphate-activated protein kinase (AMPK) and is instead mediated by protein kinase C. Our findings suggest that folate deprivation-induced premature differentiation of erythroid progenitor cells is a molecular etiology to folate deficiency-induced anemia.


Subject(s)
Folic Acid , Purines , Mice , Humans , Animals , Folic Acid/metabolism , Cell Differentiation , Cell Line , Mechanistic Target of Rapamycin Complex 1
2.
Blood Adv ; 7(16): 4647-4657, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37603347

ABSTRACT

The majority of patients with chronic graft-versus-host disease (cGVHD) are steroid refractory (SR), creating a need for safe and effective therapies. Subcutaneous low-dose interleukin-2 (LD IL-2), which preferentially expands CD4+ regulatory T cells (Tregs), has been evaluated in 5 clinical trials at our center with partial responses (PR) in ∼50% of adults and 82% of children by week 8. We now report additional real-world experience with LD IL-2 in 15 children and young adults. We conducted a retrospective chart review of patients with SR-cGVHD at our center who received LD IL-2 from August 2016 to July 2022 not on a research trial. The median age at start of LD IL-2 was 10.4 years (range, 1.2-23.2 years) at a median of 234 days from cGVHD diagnosis (range, 11-542 days). Patients had a median of 2.5 (range, 1-3) active organs at LD IL-2 start and received a median of 3 (range, 1-5) prior therapies. The median duration of LD IL-2 therapy was 462 days (range, 8-1489 days). Most patients received 1 × 106 IU/m2 per day. There were no serious adverse effects. The overall response rate in 13 patients who received >4 weeks of therapy was 85% (complete response, n = 5; PR, n = 6) with responses in diverse organs. Most patients significantly weaned corticosteroids. Tregs preferentially expanded with a median peak fold increase of 2.8 in the ratio of Tregs to CD4+ conventional T cells (range, 2.0-19.8) by 8 weeks on therapy. LD IL-2 is a well-tolerated, steroid-sparing agent with a high response rate in children and young adults with SR-cGVHD.


Subject(s)
Graft vs Host Disease , Interleukin-2 , Child , Humans , Young Adult , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Immunotherapy , Interleukin-2/administration & dosage , Retrospective Studies , Infant , Child, Preschool , Adolescent
3.
Cell Rep Methods ; 2(8): 100273, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35942328

ABSTRACT

Neutralizing antibody (NAb) titer is a key biomarker of protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, but point-of-care methods for assessing NAb titer are not widely available. Here, we present a lateral flow assay that captures SARS-CoV-2 receptor-binding domain (RBD) that has been neutralized from binding angiotensin-converting enzyme 2 (ACE2). Quantification of neutralized RBD in this assay correlates with NAb titer from vaccinated and convalescent patients. This methodology demonstrated superior performance in assessing NAb titer compared with either measurement of total anti-spike immunoglobulin G titer or quantification of the absolute reduction in binding between ACE2 and RBD. Our testing platform has the potential for mass deployment to aid in determining at population scale the degree of protective immunity individuals may have following SARS-CoV-2 vaccination or infection and can enable simple at-home assessment of NAb titer.


Subject(s)
Antibodies, Neutralizing , COVID-19 , Humans , SARS-CoV-2 , Angiotensin-Converting Enzyme 2 , COVID-19 Vaccines , Point-of-Care Systems , Antibodies, Viral , COVID-19/diagnosis
4.
Science ; 377(6604): eabm5551, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35862544

ABSTRACT

To accelerate the translation of cancer nanomedicine, we used an integrated genomic approach to improve our understanding of the cellular processes that govern nanoparticle trafficking. We developed a massively parallel screen that leverages barcoded, pooled cancer cell lines annotated with multiomic data to investigate cell association patterns across a nanoparticle library spanning a range of formulations with clinical potential. We identified both materials properties and cell-intrinsic features that mediate nanoparticle-cell association. Using machine learning algorithms, we constructed genomic nanoparticle trafficking networks and identified nanoparticle-specific biomarkers. We validated one such biomarker: gene expression of SLC46A3, which inversely predicts lipid-based nanoparticle uptake in vitro and in vivo. Our work establishes the power of integrated screens for nanoparticle delivery and enables the identification and utilization of biomarkers to rationally design nanoformulations.


Subject(s)
Antineoplastic Agents , Biomarkers, Pharmacological , Copper Transport Proteins , Drug Compounding , Nanoparticle Drug Delivery System , Nanoparticles , Neoplasms , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Cell Line, Tumor , Copper Transport Proteins/genetics , Gene Expression , Genomics , Humans , Liposomes , Mice , Nanomedicine , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism
5.
STAR Protoc ; 3(1): 101070, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35024628

ABSTRACT

Early erythroid progenitors are transit-amplifying cells with high proliferative capacity committed to undergoing red cell differentiation. CD71/CD24low progenitors are less mature and have greater proliferative capacity than CD71/CD24high. We present protocols for isolation of CD71/CD24low progenitors from mouse fetal liver using both fluorescence-activated cell sorting (FACS) and immunomagnetic enrichment. CD71/CD24low progenitors isolated with both approaches show similar transcriptomes at single-cell resolution and exhibit characteristic proliferative responses to glucocorticoids. For complete details on the use and execution of this protocol, please refer to Li et al. (2019).


Subject(s)
Erythroid Precursor Cells , Erythropoiesis , Animals , Erythropoiesis/physiology , Flow Cytometry/methods , Liver , Mice , Transcriptome
6.
Neurol Neuroimmunol Neuroinflamm ; 6(3): e560, 2019 05.
Article in English | MEDLINE | ID: mdl-31044148

ABSTRACT

Objective: To highlight a novel, treatable syndrome, we report 4 patients with CNS-isolated inflammation associated with familial hemophagocytic lymphohistiocytosis (FHL) gene mutations (CNS-FHL). Methods: Retrospective chart review. Results: Patients with CNS-FHL are characterized by chronic inflammation restricted to the CNS that is not attributable to any previously described neuroinflammatory etiology and have germline mutations in known FHL-associated genes with no signs of systemic inflammation. Hematopoietic stem cell transplantation (HCT) can be well tolerated and effective in achieving or maintaining disease remission in patients with CNS-FHL. Conclusions: Early and accurate diagnosis followed by treatment with HCT can reduce morbidity and mortality in CNS-FHL, a novel, treatable syndrome. Classification of evidence: This study provides Class IV evidence that HCT is well tolerated and effective in treating CNS-FHL.


Subject(s)
Central Nervous System Diseases/diagnosis , Lymphohistiocytosis, Hemophagocytic/diagnosis , Central Nervous System Diseases/genetics , Child , Child, Preschool , Female , Germ-Line Mutation , Humans , Lymphohistiocytosis, Hemophagocytic/genetics
7.
Dev Cell ; 49(1): 118-129.e7, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30827895

ABSTRACT

The nature of cell-state transitions during the transit-amplifying phases of many developmental processes-hematopoiesis in particular-is unclear. Here, we use single-cell RNA sequencing to demonstrate a continuum of transcriptomic states in committed transit-amplifying erythropoietic progenitors, which correlates with a continuum of proliferative potentials in these cells. We show that glucocorticoids enhance erythrocyte production by slowing the rate of progression through this developmental continuum of transit-amplifying progenitors, permitting more cell divisions prior to terminal erythroid differentiation. Mechanistically, glucocorticoids prolong expression of genes that antagonize and slow induction of genes that drive terminal erythroid differentiation. Erythroid progenitor daughter cell pairs have similar transcriptomes with or without glucocorticoid stimulation, indicating largely symmetric cell division. Thus, the rate of progression along a developmental continuum dictates the absolute number of erythroid cells generated from each transit-amplifying progenitor, suggesting a paradigm for regulating the total output of differentiated cells in numerous other developmental processes.


Subject(s)
Blood Cells/metabolism , Cell Proliferation/genetics , Erythroid Precursor Cells/metabolism , Hematopoiesis/genetics , Animals , Blood Cells/cytology , Cell Differentiation/genetics , Cell Division/genetics , Cells, Cultured , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythroid Precursor Cells/cytology , Erythropoiesis/genetics , Glucocorticoids/genetics , High-Throughput Nucleotide Sequencing/methods , Mice , Single-Cell Analysis/methods , Transcriptome/genetics
9.
J Biol Chem ; 293(51): 19797-19811, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30366982

ABSTRACT

Erythropoietin (EPO) signaling is critical to many processes essential to terminal erythropoiesis. Despite the centrality of iron metabolism to erythropoiesis, the mechanisms by which EPO regulates iron status are not well-understood. To this end, here we profiled gene expression in EPO-treated 32D pro-B cells and developing fetal liver erythroid cells to identify additional iron regulatory genes. We determined that FAM210B, a mitochondrial inner-membrane protein, is essential for hemoglobinization, proliferation, and enucleation during terminal erythroid maturation. Fam210b deficiency led to defects in mitochondrial iron uptake, heme synthesis, and iron-sulfur cluster formation. These defects were corrected with a lipid-soluble, small-molecule iron transporter, hinokitiol, in Fam210b-deficient murine erythroid cells and zebrafish morphants. Genetic complementation experiments revealed that FAM210B is not a mitochondrial iron transporter but is required for adequate mitochondrial iron import to sustain heme synthesis and iron-sulfur cluster formation during erythroid differentiation. FAM210B was also required for maximal ferrochelatase activity in differentiating erythroid cells. We propose that FAM210B functions as an adaptor protein that facilitates the formation of an oligomeric mitochondrial iron transport complex, required for the increase in iron acquisition for heme synthesis during terminal erythropoiesis. Collectively, our results reveal a critical mechanism by which EPO signaling regulates terminal erythropoiesis and iron metabolism.


Subject(s)
Erythroid Cells/metabolism , Erythropoietin/metabolism , Ferrochelatase/metabolism , Heme/biosynthesis , Iron/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Erythroid Cells/cytology , Erythropoiesis , HEK293 Cells , Humans , Membrane Proteins/chemistry , Mice , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/chemistry , Protein Transport
10.
J Neuropathol Exp Neurol ; 77(12): 1079-1084, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30295794

ABSTRACT

Familial hemophagocytic lymphohistiocytosis (HLH) is an immune hyperactivation syndrome caused by mutations in genes associated with cytotoxic T-cell and NK-cell function. While neurological manifestations frequently accompany systemic inflammation at initial presentation, isolated central nervous system (CNS) involvement is rare, and the histological correlates are not well described. We present 3 patients (ages 5, 6, and 7 years) with CNS-isolated familial HLH, who presented with a variety of neurological symptoms and underwent brain biopsies for multifocal enhancing supratentorial and infratentorial lesions. Biopsy slides from all 3 patients revealed similar findings: perivascular lymphocytes, predominantly CD3+ T-cells (CD4>CD8) with occasional intramural infiltration of small vessels; scattered histiocytes without hemophagocytosis; parenchymal and leptomeningeal inflammation varying from mild and focal to severe and sheet-like with associated destructive lesions. There was no evidence of demyelination, neoplasia, or infection. Genetic testing identified compound heterozygous mutations in PRF1 (Patients 1 and 2) and UNC13D (Patient 3), with no evidence of systemic disease except decreased NK-cell function. All 3 patients were treated with hematopoietic stem cell transplantation with marked improvement of symptoms. These findings combined with the poor outcomes associated with delayed diagnosis and lack of aggressive treatment highlight the need to consider HLH in the differential diagnosis of inflammatory brain lesions.


Subject(s)
Brain/pathology , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/pathology , Central Nervous System/pathology , Child , Child, Preschool , Diagnosis, Differential , Female , Humans
11.
Hematol Oncol Clin North Am ; 32(4): 701-712, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30047421

ABSTRACT

Diamond-Blackfan anemia (DBA) is a severe congenital hypoplastic anemia caused by mutation in a ribosomal protein gene. Major clinical issues concern the optimal management of patients resistant to steroids, the first-line therapy. Hematopoietic stem cell transplantation is indicated in young patients with an HLA-matched unaffected sibling donor, and recent results with matched unrelated donor transplants indicate that these patients also do well. When neither steroids nor a transplant is possible red cell transfusions are required, and iron loading is rapid in some DBA patients, so effective chelation is vital. Also discussed are novel treatments under investigation for DBA.


Subject(s)
Anemia, Diamond-Blackfan , Erythrocyte Transfusion , Hematopoietic Stem Cell Transplantation , Mutation , Tissue Donors , Allografts , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/metabolism , Anemia, Diamond-Blackfan/pathology , Anemia, Diamond-Blackfan/therapy , Humans , Siblings
12.
Proc Natl Acad Sci U S A ; 114(12): 3157-3162, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28270614

ABSTRACT

Current therapies for autoimmune diseases rely on traditional immunosuppressive medications that expose patients to an increased risk of opportunistic infections and other complications. Immunoregulatory interventions that act prophylactically or therapeutically to induce antigen-specific tolerance might overcome these obstacles. Here we use the transpeptidase sortase to covalently attach disease-associated autoantigens to genetically engineered and to unmodified red blood cells as a means of inducing antigen-specific tolerance. This approach blunts the contribution to immunity of major subsets of immune effector cells (B cells, CD4+ and CD8+ T cells) in an antigen-specific manner. Transfusion of red blood cells expressing self-antigen epitopes can alleviate and even prevent signs of disease in experimental autoimmune encephalomyelitis, as well as maintain normoglycemia in a mouse model of type 1 diabetes.

13.
Nat Rev Mol Cell Biol ; 18(4): 214, 2017 04.
Article in English | MEDLINE | ID: mdl-28248321
16.
Blood ; 122(19): 3283-7, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24085764

ABSTRACT

Monogenic diseases, including hemophilia, represent ideal targets for genome-editing approaches aimed at correcting a defective gene. Here we report that systemic adeno-associated virus (AAV) vector delivery of zinc finger nucleases (ZFNs) and corrective donor template to the predominantly quiescent livers of adult mice enables production of high levels of human factor IX in a murine model of hemophilia B. Further, we show that off-target cleavage can be substantially reduced while maintaining robust editing by using obligate heterodimeric ZFNs engineered to minimize unwanted cleavage attributable to homodimerization of the ZFNs. These results broaden the therapeutic potential of AAV/ZFN-mediated genome editing in the liver and could expand this strategy to other nonreplicating cell types.


Subject(s)
Endonucleases/genetics , Factor IX/biosynthesis , Genetic Therapy/methods , Genetic Vectors , Genome , Hemophilia B/therapy , Zinc Fingers/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Endonucleases/metabolism , Factor IX/genetics , Factor IX/metabolism , Hemophilia B/genetics , Hemophilia B/pathology , Liver/metabolism , Male , Mice , Mice, Transgenic , Protein Multimerization
17.
Nature ; 475(7355): 217-21, 2011 Jun 26.
Article in English | MEDLINE | ID: mdl-21706032

ABSTRACT

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


Subject(s)
DNA Repair/genetics , Disease Models, Animal , Gene Targeting/methods , Genetic Therapy/methods , Genome/genetics , Hemophilia B/genetics , Hemostasis , Animals , Base Sequence , Cell Line, Tumor , DNA Breaks, Double-Stranded , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism , Exons/genetics , Factor IX/analysis , Factor IX/genetics , Genetic Vectors/genetics , HEK293 Cells , Hemophilia B/physiopathology , Humans , Introns/genetics , Liver/metabolism , Liver Regeneration , Mice , Mice, Inbred C57BL , Mutation/genetics , Phenotype , Sequence Homology , Zinc Fingers
18.
Blood ; 117(12): 3311-9, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21106988

ABSTRACT

Gene transfer using adeno-associated virus (AAV) vectors has great potential for treating human disease. Recently, questions have arisen about the safety of AAV vectors, specifically, whether integration of vector DNA in transduced cell genomes promotes tumor formation. This study addresses these questions with high-dose liver-directed AAV-mediated gene transfer in the adult mouse as a model (80 AAV-injected mice and 52 controls). After 18 months of follow-up, AAV-injected mice did not show a significantly higher rate of hepatocellular carcinoma compared with controls. Tumors in mice treated with AAV vectors did not have significantly different amounts of vector DNA compared with adjacent normal tissue. A novel high-throughput method for identifying AAV vector integration sites was developed and used to clone 1029 integrants. Integration patterns in tumor tissue and adjacent normal tissue were similar to each other, showing preferences for active genes, cytosine-phosphate-guanosine islands, and guanosine/cytosine-rich regions. [corrected] Gene expression data showed that genes near integration sites did not show significant changes in expression patterns compared with genes more distal to integration sites. No integration events were identified as causing increased oncogene expression. Thus, we did not find evidence that AAV vectors cause insertional activation of oncogenes and subsequent tumor formation.


Subject(s)
Dependovirus/genetics , Genetic Therapy/adverse effects , Genetic Vectors/adverse effects , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Dependovirus/physiology , Disease Models, Animal , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/physiology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional/physiology , Mutagenicity Tests
19.
J Med Virol ; 81(1): 65-74, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19031458

ABSTRACT

Humoral immune responses occur following exposure to Adeno-associated virus (AAV) or AAV vectors. Many studies characterized antibody responses to AAV, but human IgG subclass responses to AAV have not been previously described. In this study, IgG subclass responses were examined in serum samples of normal human subjects exposed to wild-type AAV, subjects injected intramuscularly with AAV vectors and subjects injected intravascularly with AAV vectors. A diversity of IgG subclass responses to AAV capsid were found in different subjects. IgG1 was found to be the dominant response. IgG2, IgG3, and IgG4 responses were also observed in most normal human subjects; IgG2 and IgG3 each represented the major fraction of total anti-AAV capsid IgG in a subset of normal donors. Subjects exposed to AAV vectors showed IgG responses to AAV capsid of all four IgG subclasses. IgG responses to AAV capsid in clinical trial subjects were inversely proportional to the level of pre-existing anti-AAV antibody and independent of the vector dose. The high levels of anti-AAV capsid IgG1 can mask differences in IgG2, IgG3, and IgG4 responses that were observed in this study. Analysis of IgG subclass distribution of anti-AAV capsid antibodies indicates a complex, non-uniform pattern of responses to this viral antigen. J. Med. Virol. 81:65-74, 2009. (c) 2008 Wiley-Liss, Inc.


Subject(s)
Antibodies, Viral/blood , Dependovirus/immunology , Genetic Vectors/immunology , Immunoglobulin G/blood , Parvoviridae Infections/immunology , Adolescent , Adult , Aged , Capsid/immunology , Humans , Immunoglobulin M/blood , Middle Aged , Neutralization Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...