Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Nat Commun ; 15(1): 615, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38242888

ABSTRACT

Anti-multiple myeloma B cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T-cell therapies represent a promising treatment strategy with high response rates in myeloma. However, durable cures following anti-BCMA CAR-T cell treatment of myeloma are rare. One potential reason is that a small subset of minimal residual myeloma cells seeds relapse. Residual myeloma cells following BCMA-CAR-T-mediated treatment show less-differentiated features and express stem-like genes, including CD24. CD24-positive myeloma cells represent a large fraction of residual myeloma cells after BCMA-CAR-T therapy. In this work, we develop CD24-CAR-T cells and test their ability to eliminate myeloma cells. We find that CD24-CAR-T cells block the CD24-Siglec-10 pathway, thereby enhancing macrophage phagocytic clearance of myeloma cells. Additionally, CD24-CAR-T cells polarize macrophages to a M1-like phenotype. A dual-targeted BCMA-CD24-CAR-T exhibits improved efficacy compared to monospecific BCMA-CAR-T-cell therapy. This work presents an immunotherapeutic approach that targets myeloma cells and promotes tumor cell clearance by macrophages.


Subject(s)
Multiple Myeloma , Receptors, Chimeric Antigen , Humans , Multiple Myeloma/pathology , T-Lymphocytes , B-Cell Maturation Antigen/genetics , Neoplasm Recurrence, Local , Antibodies , CD24 Antigen
2.
Front Immunol ; 13: 997482, 2022.
Article in English | MEDLINE | ID: mdl-36172388

ABSTRACT

T-cell lymphomas are aggressive lymphomas that often resist current therapy options or present with relapsed disease, making the development of more effective treatment regimens clinically important. Previously, we have shown that CD4 CAR can effectively target T-cell malignancies in preclinical studies. As IL-15 has been shown to strengthen the anti-tumor response, we have modified CD4 CAR to secrete an IL-15/IL-15sushi complex. These CD4-IL15/IL15sushi CAR T cells and NK92 cells efficiently eliminated CD4+ leukemic cell lines in co-culture assays. Additionally, CD4-IL15/IL15sushi CAR out-performed CD4 CAR in in vivo models, demonstrating a benefit to IL-15/IL-15sushi inclusion. In a Phase I clinical trial, CD4-IL15/IL15sushi CAR T cells were tested for safety in three patients with different T-cell lymphomas. Infusion of CD4-IL15/IL15sushi CAR T cells was well-tolerated by the patients without significant adverse effects and led to the remission of their lymphomas. Additionally, infusion led to the depletion of CD4+ Treg cells and expansion of CD3+CD8+ T cells and NK cells. These results suggest that CD4-IL15/IL15sushi CAR T cells may be a safe and effective treatment for patients with relapsed or refractory T-cell lymphomas, where new treatment options are needed.


Subject(s)
Leukemia , Lymphoma, T-Cell , Clinical Trials, Phase I as Topic , Humans , Immunotherapy, Adoptive/methods , Interleukin-15 , Killer Cells, Natural
3.
Leuk Res Rep ; 17: 100327, 2022.
Article in English | MEDLINE | ID: mdl-35634195

ABSTRACT

Chronic myeloid leukemia(CML) is characterized by Philadelphia(Ph) chromosome. About 5% of cases are diagnosed in blast phase. We report a case of a 53-year-old female with no significant medical history, in B-lymphoblast crisis. Flow cytometry demonstrated B-lymphoblasts with no myeloid aberrancies, together with immature neutrophils in blood, and B-lymphoblasts in bone marrow. Cytogenetic studies identified Ph+ with complex abnormalities. Molecular analysis showed positive both for p210 and p190 transcripts in blood. ABL1 mutation analysis by Next Generation Sequencing(NGS) detected Thr315Ile mutation, which confers resistance to many tyrosine kinase inhibitors(TKIs). Eight months later she received allogeneic transplant and is doing well.

6.
Pharmacol Res ; 169: 105656, 2021 07.
Article in English | MEDLINE | ID: mdl-33964470

ABSTRACT

Non-small cell lung cancer (NSCLC) is one of the most frequently diagnosed cancers and the leading causes of cancer death worldwide. Therefore, new therapeutic agents are urgently needed to improve patient outcomes. Plumbagin (PLB), a natural sesquiterpene present in many Chinese herbal medicines, has been reported for its anti-cancer activity in various cancer cells. In this study, the effects and underlying mechanisms of PLB on the tumorigenesis of NSCLC were investigated. PLB dose-dependently inhibited the growth of NSCLC cell lines. PLB promoted ROS production, activated the endoplasmic reticulum (ER) stress pathway, and induced cell apoptosis, accompanied by the decreased expression level of ADP-ribosylation factor 1 (ARF1) in NSCLC cancer cells, and those effects of PLB could be reversed by the pretreatment with N-acetyl-L-cysteine (NAC). More importantly, the calcium chelator (BM) significantly reversed PLB-induced cell apoptosis. Furthermore, PLB significantly inhibited the growth of both H1975 xenograft and LLC1 tumors and exhibited antitumor activity by enhancing the number and the effector function of CD8+ T cells in KRASLA2 mice model and the LLC1 xenograft. Our findings suggest that PLB exerts potent antitumor activity against NSCLC in vitro and in vivo through ARF1 downregulation and induction of antitumor immune response, indicating that PLB is a new novel therapeutic candidate for the treatment of patients with NSCLC.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Naphthoquinones/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Down-Regulation/drug effects , Female , Lymphocyte Activation/drug effects , Mice, Nude , Naphthoquinones/pharmacology , Neoplasm Transplantation
7.
Cancer Lett ; 515: 36-48, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34052328

ABSTRACT

Upregulated expression of immune checkpoint molecules correlates with exhausted phenotype and impaired function of cytotoxic T cells to evade host immunity. By disrupting the interaction of PD-L1 and PD1, immune checkpoint inhibitors can restore immune system function against cancer cells. Growing evidence have demonstrated apigenin and luteolin, which are flavonoids abundant in common fruits and vegetables, can suppress growth and induce apoptosis of multiple types of cancer cells with their potent anti-inflammatory, antioxidant and anticancer properties. In this study, the effects and underlying mechanisms of luteolin, apigenin, and anti-PD-1 antibody combined with luteolin or apigenin on the PD-L1 expression and anti-tumorigenesis in KRAS-mutant lung cancer were investigated. Luteolin and apigenin significantly inhibited lung cancer cell growth, induced cell apoptosis, and down-regulated the IFN-γ-induced PD-L1 expression by suppressing the phosphorylation of STAT3. Both luteolin and apigenin showed potent anti-cancer activities in the H358 xenograft and Lewis lung carcinoma model in vivo, and the treatment with monoclonal PD1 antibody enhanced the infiltration of T cells into tumor tissues. Apigenin exhibited anti-tumor activity in Genetically engineered KRASLA2 mice. In conclusion, both apigenin and luteolin significantly suppressed lung cancer with KRAS mutant proliferation, and down-regulated the IFN-γ induced PD-L1 expression. Treatment with the combination of PD-1 blockade and apigenin/luteolin has a synergistic effect and might be a prospective therapeutic strategy for NSCLC with KRAS-mutant.


Subject(s)
Apigenin/pharmacology , B7-H1 Antigen/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Luteolin/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , A549 Cells , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Humans , Interferon-gamma/metabolism , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude
8.
Stem Cell Rev Rep ; 17(2): 652-661, 2021 04.
Article in English | MEDLINE | ID: mdl-33410096

ABSTRACT

While treatment for B-cell malignancies has been revolutionized through the advent of CAR immunotherapy, similar strategies for T-cell malignancies have been limited. Additionally, T-cell leukemias and lymphomas can commonly metastasize to the CNS, where outcomes are poor and treatment options are associated with severe side effects. Consequently, the development of safer and more effective alternatives for targeting malignant T cells that have invaded the CNS remains clinically important. CD5 CAR has previously been shown to effectively target various T-cell cancers in preclinical studies. As IL-15 strengthens the anti-tumor response, we have modified CD5 CAR to secrete an IL-15/IL-15sushi complex. In a Phase I clinical trial, these CD5-IL15/IL15sushi CAR T cells were tested for safety and efficacy in a patient with refractory T-LBL with CNS infiltration. CD5-IL15/IL15sushi CAR T cells were able to rapidly ablate the CNS lymphoblasts within a few weeks, resulting in the remission of the patient's lymphoma. Despite the presence of CD5 on normal T cells, the patient only experienced a brief, transient T-cell aplasia. These results suggest that CD5-IL15/IL15sushi CAR T cells may be a safe and useful treatment of T-cell malignancies and may be particularly beneficial for patients with CNS involvement.Graphical Abstract.


Subject(s)
Immunotherapy, Adoptive , Interleukin-15 , Precursor Cell Lymphoblastic Leukemia-Lymphoma , T-Lymphocytes , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
9.
Free Radic Biol Med ; 164: 164-174, 2021 02 20.
Article in English | MEDLINE | ID: mdl-33359909

ABSTRACT

Cytoprotective mechanisms of heme oxygenases function by derivatizing heme to generate carbon monoxide, ferrous iron, and isomeric biliverdins, followed by rapid NAD(P)H-dependent biliverdin reduction to the antioxidant bilirubin using two non-overlapping biliverdin reductases that display biliverdin isomer-restricted redox activity. Although cytoprotective functions of heme oxygenases are widely recognized, concomitant effects of downstream biliverdin reductases remain incomplete. A computational model predicated on murine hematopoietic single-cell transcriptomic data identified Blvrb as a biological driver linked to the tumor necrosis factor stress pathway as a predominant source of variation defining hematopoietic cell heterogeneity. In vivo studies using Blvrb-deficient mice established the dispensable role of Blvrb in steady-state hematopoiesis, although model validation using aged Blvrb-deficient mice established an important cytoprotective function in stress hematopoiesis with dichotomous megakaryocyte-biased hematopoietic recovery. Defective stress erythropoiesis was evident in Blvrb-/- spleens and in bone marrow erythroid development, occurring in conjunction with defective lipid peroxidation as a marker of oxidant mishandling. Cell autonomous effects on megakaryocyte lineage bias were documented using multipotential progenitor assays. These data provide the first physiological function of murine Blvrb in a non-redundant pathway of stress cytoprotection. Divergent effects on erythroid/megakaryocyte lineage speciation impute a novel redox-regulated mechanism for lineage partitioning.


Subject(s)
Hematopoiesis , Megakaryocytes , Oxidoreductases Acting on CH-CH Group Donors/genetics , Animals , Biliverdine , Cell Lineage , Hematopoiesis/genetics , Heme , Mice , Mice, Knockout
10.
Stem Cell Rev Rep ; 16(2): 369-384, 2020 04.
Article in English | MEDLINE | ID: mdl-32008159

ABSTRACT

T-cell malignancies often result in poor prognosis and outcome for patients. Immunotherapy has recently emerged as a revolutionary treatment against cancer, and the success seen in CD19 CAR clinical trials may extend to T cell diseases. However, a shared antigen pool coupled with the impact of T-cell depletion incurred by targeting T cell disease remain concepts to be clinically explored with caution. Here we report on the ability of T cells transduced with a CD5CAR to specifically and potently lyse malignant T-cell lines and primary tumors in vitro in addition to significantly improving in vivo control and survival of xenograft models of T-ALL. To extensively explore and investigate the biological properties of a CD5 CAR, we evaluated multiple CD5 CAR constructs and constructed 3 murine models to characterize the properties of CD5 down-regulation, the efficacy and specificity produced by different CD5 CAR construct designs, and the impact of incorporating a CD52 safety switch using CAMPATH to modulate the persistency and function of CAR cells. These data support the potential use of CD5CAR T cells in the treatment of T cell malignancies or refractory disease in clinical settings.


Subject(s)
CD5 Antigens/metabolism , Immunotherapy, Adoptive , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Alemtuzumab/pharmacology , Alemtuzumab/therapeutic use , Animals , Cell Line , Down-Regulation/drug effects , Humans , Male , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recombinant Proteins/metabolism , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
11.
Stem Cell Rev Rep ; 16(2): 385-396, 2020 04.
Article in English | MEDLINE | ID: mdl-31970687

ABSTRACT

The recent FDA approval of the first CAR immunotherapy marks a watershed moment in the advancement toward a cure for cancer. CD19 CAR treatment for B cell acute lymphocytic leukemia has achieved unprecedented remission rates. However, despite success in treating previously relapsed and refractory patients, CD19 CAR faces similar challenges as traditional chemotherapy, in that malignancy can adapt and overcome treatment. The emergence of both antigen positive and negative blasts after CAR treatment represents a need to bolster current CAR approaches. Here, we report on the anti-tumor activity of a CAR T cell possessing 2 discrete scFv domains against the leukemic antigens CD19 and CD123. We determined that the resulting compound CAR (cCAR) T cell possesses consistent, potent, and directed cytotoxicity against each target antigen population both in vitro and in vivo. Our findings indicate that targeting CD19 and CD123 on B-ALL cells may be an effective strategy for augmenting the response against leukemic blasts and reducing rates of relapse.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, B-Cell/immunology , Leukemia, B-Cell/therapy , Alemtuzumab/pharmacology , Alemtuzumab/therapeutic use , Animals , Epitopes/immunology , Humans , K562 Cells , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Male , Mice
12.
J Cancer ; 10(18): 4408-4419, 2019.
Article in English | MEDLINE | ID: mdl-31413761

ABSTRACT

Acute myeloid leukemia (AML) is an aggressive malignancy lacking targeted therapy due to shared molecular and transcriptional circuits as well as phenotypic markers with normal hematopoietic stem cells (HSCs). Identifying leukemia specific markers expressed on AML or AML subtypes for therapeutic targeting is of exquisite clinical value. Here we show that CD4, a T lymphocytes membrane glycoprotein that interacts with major histocompatibility complex class II antigens and is also expressed in certain AML subsets but not on HSCs is a proper target for genetically engineered chimeric antigen receptor T cells (CAR-T cells). Treatment with CD4 redirected CAR-T cell (CD4CAR) specifically eliminated CD4-expressing AML cell lines in vitro and exhibited a potent anti-leukemic effect in a systemic AML murine model in vivo. We also utilized natural killers as another vehicle for CAR engineered cells and this strategy similarly and robustly eliminated CD4- expressing AML cells in vitro and had a potent in vivo anti-leukemic effect and was noted to have shorter in vivo persistence. Our data offer a proof of concept for immunotherapeutic targeting of CD4 as a strategy to treat CD4 expressing refractory AML as a bridge to stem cell transplant (SCT) in a first in human clinical trial.

13.
Stem Cell Res Ther ; 10(1): 173, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31196160

ABSTRACT

BACKGROUND: Hematopoietic stem cell (HSC) transplantation has been widely applied to the treatment of malignant blood diseases. However, limited number of functional HSCs hinders successful transplantation. The purpose of our current study is to develop a new and cost-efficient medium formulation that could greatly enhance the expansion of HSCs while retaining their long-term repopulation and hematopoietic properties for effective clinical transplantation. METHODS: Enriched human CD34+ cells and mobilized nonhuman primate peripheral blood CD34+ cells were expanded with a new, cost-efficient expansion medium formulation, named hematopoietic expansion medium (HEM), consisting of various cytokines and nutritional supplements. The long-term repopulation potential and hematologic-lineage differentiation ability of expanded human cells were studied in the non-obese diabetic/severe combined immunodeficiency mouse model. Furthermore, the efficacy and safety studies were performed by autologous transplantation of expanded primate cells in the nonhuman primate model. RESULTS: HEM could effectively expand human CD34+ cells by up to 129 fold within 9 days. Expanded HSCs retained long-term repopulation potential and hematologic-lineage differentiation ability, as indicated by (1) maintenance (over unexpanded HSCs) of immunophenotypes of CD38-CD90+CD45RA-CD49f+ in CD34+ cells after expansion; (2) significant presence of multiple human hematopoietic lineages in mouse peripheral blood and bone marrow following primary transplantation; (3) enrichment (over unexpanded HSCs) in SCID-repopulating cell frequency measured by limiting dilution analysis; and (4) preservation of both myeloid and lymphoid potential among human leukocytes from mouse bone marrow in week 24 after primary transplantation or secondary transplantation. Moreover, the results of autologous transplantation in nonhuman primates demonstrated that HEM-expanded CD34+ cells could enhance hematological recovery after myelo-suppression. All primates transplanted with the expanded autologous CD34+ cells survived for over 18 months without any noticeable abnormalities. CONCLUSIONS: Together, these findings demonstrate promising potential for the utility of HEM to improve expansion of HSCs for clinical application.


Subject(s)
Antigens, CD34/metabolism , Hematopoietic Stem Cells/metabolism , ADP-ribosyl Cyclase 1/metabolism , Animals , Cells, Cultured , Female , Flow Cytometry , Hematopoietic Stem Cells/cytology , Humans , Immunophenotyping , Integrin alpha6/metabolism , Leukocyte Common Antigens/metabolism , Mice , Mice, SCID , Primates , Thy-1 Antigens/metabolism
14.
Leuk Res Rep ; 11: 34-37, 2019.
Article in English | MEDLINE | ID: mdl-31061791

ABSTRACT

Near-haploid acute lymphoblastic leukemia is seen in <1% of cases and carries an unfavorable prognosis. We report a case in an 18-year old male. He presented with two abnormal clones, one with 27-28 and one with 54-56 chromosomes. Near-haploidy (27-28) carries a poor prognosis and hyperdiploidy (>50) has a good prognosis. The correct diagnosis was critical for this patient's prognosis and treatment. The patient achieved remission after a bone marrow transplant from a 10/10 HLA matched sibling donor. He relapsed six months later and expired seven months later. This case illustrates the need for careful standard and molecular cytogenetic analysis for accurate diagnosis and treatment for this rare type of ALL.

16.
Leukemia ; 32(6): 1317-1326, 2018 06.
Article in English | MEDLINE | ID: mdl-29515236

ABSTRACT

Acute myeloid leukemia (AML) bears heterogeneous cells that can consequently offset killing by single-CAR-based therapy, which results in disease relapse. Leukemic stem cells (LSCs) associated with CD123 expression comprise a rare population that also plays an important role in disease progression and relapse. Here, we report on the robust anti-tumor activity of a compound CAR (cCAR) T-cell possessing discrete scFv domains targeting two different AML antigens, CD123, and CD33, simultaneously. We determined that the resulting cCAR T-cells possessed consistent, potent, and directed cytotoxicity against each target antigen population. Using four leukemia mouse models, we found superior in vivo survival after cCAR treatment. We also designed an alemtuzumab safety-switch that allowed for rapid cCAR therapy termination in vivo. These findings indicate that targeting both CD123 and CD33 on AML cells may be an effective strategy for eliminating both AML bulk disease and LSCs, and potentially prevent relapse due to antigen escape or LSC persistence.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/therapy , Receptors, Antigen, T-Cell/immunology , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Alemtuzumab/therapeutic use , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Male , Mice
17.
Oncotarget ; 9(7): 7442-7452, 2018 Jan 26.
Article in English | MEDLINE | ID: mdl-29484122

ABSTRACT

Similar signaling pathways could operate in both normal hematopoietic stem and progenitor cells (HSPCs) and leukemia stem cells (LSCs). Thus, targeting LSCs signaling without substantial toxicities to normal HSPCs remains challenging. SALL1, is a member of the transcriptional network that regulates stem cell pluripotency, and lacks significant expression in most adult tissues, including normal bone marrow (NBM). We examined the expression and functional characterization of SALL1 in NBM and in acute myeloid leukemia (AML) using in vitro and in vivo assays. We showed that SALL1 is expressed preferentially in LSCs- enriched CD34+CD38- cell subpopulation but not in NBM. SALL1 inhibition resulted in decreased cellular proliferation and in inferior AML engraftment in NSG mice and it was also associated with upregulation of PTEN and downregulation of m-TOR, ß-catenin, and NF-қB expression. These findings suggest that SALL1 inhibition interrupts leukemogenesis. Further studies to validate SALL1 as a potential biomarker for minimal residual disease (MRD) and to determine SALL1's role in prognostication are ongoing. Additionally, pre-clinical evaluation of SALL1 as a therapeutic target in AML is warranted.

18.
Stem Cell Res Ther ; 9(1): 14, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29357928

ABSTRACT

BACKGROUND: Autologous transplantation of endothelial progenitor cells (EPCs) is a promising therapeutic approach in the treatment of various vascular diseases. We previously reported a two-step culture system for scalable generation of human EPCs derived from cord blood CD34+ cells ex vivo. Here, we now apply this culture system to expand and differentiate human and nonhuman primate EPCs from mobilized peripheral blood (PB) CD34+ cells for the therapeutic potential of autologous transplantation. METHODS: The human and nonhuman primate EPCs from mobilized PB CD34+ cells were cultured according to our previously reported system. The generated adherent cells were then characterized by the morphology, surface markers, nitric oxide (NO)/endothelial NO synthase (eNOS) levels and Dil-acetylated low-density lipoprotein (Dil-Ac-LDL) uptake/fluorescein isothiocyanate (FITC)-lectin binding actives. Furthermore, the efficacy and safety studies were performed by autologous transplantation via hepatic portal vein injection in a nonhuman primate model with acute liver sinusoidal endothelial cell injury. RESULTS: The mobilized PB CD34+ cells from both human and nonhuman primate were efficiently expanded and differentiated. Over 2 × 108 adherent cells were generated from 20 mL mobilized primate PB (1.51 × 106 ± 3.39 × 105 CD34+ cells) by 36-day culture and more than 80% of the produced cells were identified as EPCs/endothelial cells (ECs). In the autologous transplant model, the injected EPC/ECs from nonhuman primate PB were scattered in the intercellular spaces of hepatocytes at the hepatic tissues 14 days post-transplantation, indicating successful migration and reconstitution in the liver structure as the functional EPCs/ECs. CONCLUSIONS: We successfully applied our previous two-step culture system for the generation of primate EPCs from mobilized PB CD34+ cells, evaluated the phenotypes ex vivo, and transplanted autologous EPCs/ECs in a nonhuman primate model. Our study indicates that it may be possible for these ex-vivo high-efficient expanded EPCs to be used in clinical cell therapy.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/transplantation , Vascular Diseases/therapy , Adult , Animals , Antigens, CD34/metabolism , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Fetal Blood/cytology , Humans , Lipoproteins, LDL/metabolism , Liver/metabolism , Macaca fascicularis , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Transplantation, Autologous
19.
Chin Med Sci J ; 32(3): 135-144, 2017 Sep 27.
Article in English | MEDLINE | ID: mdl-28956740

ABSTRACT

Objective To genetically correct a disease-causing point mutation in human induced pluripotent stem cells (iPSCs) derived from a hemophilia B patient. Methods First, the disease-causing mutation was detected by sequencing the encoding area of human coagulation factor IX (F IX) gene. Genomic DNA was extracted from the iPSCs, and the primers were designed to amplify the eight exons of F IX. Next, the point mutation in those iPSCs was genetically corrected using CRISPR/Cas9 technology in the presence of a 129-nucleotide homologous repair template that contained two synonymous mutations. Then, top 8 potential off-target sites were subsequently analyzed using Sanger sequencing. Finally, the corrected clones were differentiated into hepatocyte-like cells, and the secretion of F IX was validated by immunocytochemistry and ELISA assay. Results The cell line bore a missense mutation in the 6th coding exon (c.676 C>T) of F IX gene. Correction of the point mutation was achieved via CRISPR/Cas9 technology in situ with a high efficacy at about 22% (10/45) and no off-target effects detected in the corrected iPSC clones. F IX secretion, which was further visualized by immunocytochemistry and quantified by ELISA in vitro, reached about 6 ng/ml on day 21 of differentiation procedure. Conclusions Mutations in human disease-specific iPSCs could be precisely corrected by CRISPR/Cas9 technology, and corrected cells still maintained hepatic differentiation capability. Our findings might throw a light on iPSC-based personalized therapies in the clinical application, especially for hemophilia B.


Subject(s)
Cell Differentiation , Factor IX , Genetic Therapy , Hemophilia B , Induced Pluripotent Stem Cells/metabolism , Mutation , Factor IX/genetics , Factor IX/metabolism , Hemophilia B/genetics , Hemophilia B/metabolism , Hemophilia B/pathology , Hemophilia B/therapy , Humans , Induced Pluripotent Stem Cells/pathology
20.
Emerg Infect Dis ; 23(10): 1749-1751, 2017 10.
Article in English | MEDLINE | ID: mdl-28930016

ABSTRACT

We report a case of lymphadenitis caused by Mycobacterium orygis in an immunocompetent person in Stony Brook, New York, USA. Initial real-time PCR assay failed to provide a final subspecies identification within the M. tuberculosis complex, but whole-genome sequencing characterized the isolate as M. orygis.


Subject(s)
Genome, Bacterial , Lymphadenitis/diagnosis , Mycobacterium/genetics , Aged , Emigrants and Immigrants , Female , Humans , India , Lymph Nodes/microbiology , Lymph Nodes/pathology , Lymphadenitis/microbiology , Lymphadenitis/pathology , Mycobacterium/classification , Mycobacterium/isolation & purification , New York , Phylogeny , Whole Genome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL
...