Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Heliyon ; 10(1): e23595, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38187240

ABSTRACT

Objectives: This study aims to examine whether the parenterally administered mRNA-based COVID-19 vaccines can induce sufficient mucosal-type IgA responses to prevent SARS-CoV-2 transmission. Methods: We examined the longitudinal kinetics of SARS-CoV-2 spike RBD-specific IgA and IgG responses in sera of Japanese healthcare workers (HCWs) after receiving two doses and the third dose of BNT162b2 mRNA vaccines. During the prospective cohort study, Omicron breakthrough infections occurred in 62 participants among 370 HCWs who had received triple doses of the vaccine. Pre-breakthrough sera of infected HCWs and non-infected HCWs were examined for the levels of anti-RBD IgA and IgG titers. Results: The seropositivity of anti-RBD IgA at 1 M after the second vaccine (2D-1M) and after the third dose (3D-1M) was 65.4% and 87.4%, respectively, and wanes quickly. The boosting effect on anti-RBD Ab titers following breakthrough infections was more notable for anti-RBD IgA than for IgG. There were partial cause-relationships between the lower anti-RBD IgA or IgG at pre-breakthrough sera and the breakthrough infection. Conclusions: Parenterally administered COVID-19 vaccines do not generate sufficient mucosal-type IgA responses despite strong systemic IgG responses to SARS-CoV-2. These results demonstrate the necessity and importance of reevaluating vaccine design and scheduling to efficiently increase oral or respiratory mucosal immunity against SARS-CoV-2.

2.
Int J Infect Dis ; 139: 1-5, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38029832

ABSTRACT

OBJECTIVES: It is crucial to analyze the consequences of repeated messenger RNA (mRNA)-based COVID-19 vaccinations on SARS-CoV-2 spike receptor binding domain (RBD)-specific immunoglobulin (Ig)G subclass and the possible causal relationship with breakthrough infection. METHODS: We examined the longitudinal kinetics of RBD-specific IgG subclass antibodies in sera after receiving the second, third, and fourth doses of mRNA-based COVID-19 vaccines in Japanese healthcare workers. Anti-RBD IgG subclass in sera of patients with COVID-19-infected who had not received the COVID-19 vaccine were also examined. We compared anti-RBD IgG subclass antibody titers in the serum of pre-breakthrough-infected vaccinees and non-infected vaccinees. RESULTS: The seropositivity of anti-RBD IgG4 after the vaccination was 6.76% at 1 month after the second dose, gradually increased to 50.5% at 6 months after the second dose, and reached 97.2% at 1 month after the third dose. The seropositivity and titers of anti-RBD IgG1/IgG3 quickly reached the maximum at 1 month after the second dose and declined afterward. The elevated anti-RBD IgG4 Ab levels observed after repeated vaccinations were unlikely to increase the risk of breakthrough infection. CONCLUSIONS: Repeated vaccinations induce delayed but drastic increases in anti-RBD IgG4 responses. Further functional investigations are needed to reveal the magnitude of the high contribution of spike-specific IgG4 subclasses after repeated mRNA-based COVID-19 vaccinations.


Subject(s)
COVID-19 Vaccines , COVID-19 , Humans , COVID-19/prevention & control , Breakthrough Infections , SARS-CoV-2 , Immunization , Vaccination , Immunoglobulin G , RNA, Messenger/genetics , Antibodies, Viral
3.
J Immunol ; 210(8): 1086-1097, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36883861

ABSTRACT

Fatty acid-binding protein 4 (FABP4) is a critical immune-metabolic modulator, mainly expressed in adipocytes and macrophages, secreted from adipocytes in association with lipolysis, and plays essential pathogenic roles in cardiovascular and metabolic diseases. We previously reported Chlamydia pneumoniae infecting murine 3T3-L1 adipocytes and causing lipolysis and FABP4 secretion in vitro. However, it is still unknown whether C. pneumoniae intranasal lung infection targets white adipose tissues (WATs), induces lipolysis, and causes FABP4 secretion in vivo. In this study, we demonstrate that C. pneumoniae lung infection causes robust lipolysis in WAT. Infection-induced WAT lipolysis was diminished in FABP4-/- mice or FABP4 inhibitor-pretreated wild-type mice. Infection by C. pneumoniae in wild-type but not FABP4-/- mice induces the accumulation of TNF-α- and IL-6-producing M1-like adipose tissue macrophages in WAT. Infection-induced WAT pathology is augmented by endoplasmic reticulum (ER) stress/the unfolded protein response (UPR), which is abrogated by treatment with azoramide, a modulator of the UPR. C. pneumoniae lung infection is suggested to target WAT and induce lipolysis and FABP4 secretion in vivo via ER stress/UPR. FABP4 released from infected adipocytes may be taken up by other neighboring intact adipocytes or adipose tissue macrophages. This process can further induce ER stress activation and trigger lipolysis and inflammation, followed by FABP4 secretion, leading to WAT pathology. A better understanding of the role of FABP4 in C. pneumoniae infection-induced WAT pathology will provide the basis for rational intervention measures directed at C. pneumoniae infection and metabolic syndrome, such as atherosclerosis, for which robust epidemiologic evidence exists.


Subject(s)
Adipose Tissue, White , Chlamydophila Infections , Fatty Acid-Binding Proteins , Pneumonia, Bacterial , Animals , Mice , Adipose Tissue, White/pathology , Chlamydophila pneumoniae , Fatty Acid-Binding Proteins/metabolism , Lung/microbiology , Lung/pathology , Chlamydophila Infections/pathology , Pneumonia, Bacterial/pathology
4.
Elife ; 92020 02 03.
Article in English | MEDLINE | ID: mdl-32011235

ABSTRACT

Generation of skeletal muscle cells with human pluripotent stem cells (hPSCs) opens new avenues for deciphering essential, but poorly understood aspects of transcriptional regulation in human myogenic specification. In this study, we characterized the transcriptional landscape of distinct human myogenic stages, including OCT4::EGFP+ pluripotent stem cells, MSGN1::EGFP+ presomite cells, PAX7::EGFP+ skeletal muscle progenitor cells, MYOG::EGFP+ myoblasts, and multinucleated myotubes. We defined signature gene expression profiles from each isolated cell population with unbiased clustering analysis, which provided unique insights into the transcriptional dynamics of human myogenesis from undifferentiated hPSCs to fully differentiated myotubes. Using a knock-out strategy, we identified TWIST1 as a critical factor in maintenance of human PAX7::EGFP+ putative skeletal muscle progenitor cells. Our data revealed a new role of TWIST1 in human skeletal muscle progenitors, and we have established a foundation to identify transcriptional regulations of human myogenic ontogeny (online database can be accessed in http://www.myogenesis.net/).


Subject(s)
Gene Expression Regulation, Developmental/genetics , Muscle Development/genetics , Muscle, Skeletal/cytology , Nuclear Proteins , Pluripotent Stem Cells/metabolism , Twist-Related Protein 1 , Cell Line , Gene Knockout Techniques , Humans , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
5.
J Biol Chem ; 295(9): 2713-2723, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31992597

ABSTRACT

Fatty acid-binding protein 4 (FABP4) is predominantly expressed in adipocytes and macrophages and regulates metabolic and inflammatory pathways. FABP4 is secreted from adipocytes during lipolysis, and elevated circulating FABP4 levels are associated with obesity, metabolic disease, and cardiac dysfunction. We previously reported that the bacterial respiratory pathogen Chlamydia pneumoniae infects murine adipocytes and exploits host FABP4 to mobilize fat and replicate within adipocytes. However, whether C. pneumoniae induces FABP4 secretion from adipocytes has not been determined. Here, we show that FABP4 is actively secreted by murine adipocytes upon C. pneumoniae infection. Chemical inhibition of lipase activity and genetic deficiency of hormone-sensitive lipase blocked FABP4 secretion from C. pneumoniae-infected adipocytes. Mechanistically, C. pneumoniae infection induced endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), resulting in elevated levels of mitochondrial reactive oxygen species and cytosolic Ca2+ Of note, exposure to a mitochondrial reactive oxygen species-specific scavenger, MitoTEMPO, reduced FABP4 release from C. pneumoniae-infected adipocytes. Furthermore, treatment with azoramide, which protects cells against ER stress, decreased FABP4 release from C. pneumoniae-infected adipocytes. Using gene silencing of CHOP (C/EBP homologous protein), a central regulator of ER stress, we further validated the role of C. pneumoniae infection-induced ER stress/UPR in promoting FABP4 secretion. Overall, these results indicate that C. pneumoniae infection robustly induces FABP4 secretion from adipocytes by stimulating ER stress/UPR. Our findings shed additional light on the etiological link between C. pneumoniae infection and metabolic syndrome.


Subject(s)
Adipocytes/metabolism , Chlamydophila Infections/metabolism , Endoplasmic Reticulum Stress , Fatty Acid-Binding Proteins/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Animals , Calcium/metabolism , Lipase/antagonists & inhibitors , Metabolic Syndrome/etiology , Mice , Reactive Oxygen Species/metabolism , Unfolded Protein Response
6.
Cell Microbiol ; 21(1): e12962, 2019 01.
Article in English | MEDLINE | ID: mdl-30311994

ABSTRACT

Chlamydia trachomatis is an obligate intracellular bacterium that scavenges host metabolic products for its replication. Mitochondria are the power plants of eukaryotic cells and provide most of the cellular ATP via oxidative phosphorylation. Several intracellular pathogens target mitochondria as part of their obligatory cellular reprogramming. This study was designed to analyse the mitochondrial morphological changes in response to C. trachomatis infection in HeLa cells. Mitochondrial elongation and fragmentation were found at the early stages and late stages of C. trachomatis infection, respectively. C. trachomatis infection-induced mitochondrial elongation was associated with the increase of mitochondrial respiratory activity, ATP production, and intracellular growth of C. trachomatis. Silencing mitochondrial fusion mediator proteins abrogated the C. trachomatis infection-induced elevation in the oxygen consumption rate and attenuated chlamydial proliferation. Mechanistically, C. trachomatis induced the elevation of intracellular cAMP at the early phase of infection, followed by the phosphorylation of fission-inactive serine residue 637 (S637) of Drp1, resulting in mitochondrial elongation. Accordingly, treatment with adenylate cyclase inhibitor diminished mitochondrial elongation and bacterial growth in infected cells. Collectively, these results strongly indicate that C. trachomatis promotes its intracellular growth by targeting mitochondrial dynamics to regulate ATP synthesis via inhibition of the fission mediator Drp1.


Subject(s)
Chlamydia Infections/pathology , Chlamydia trachomatis/growth & development , Epithelial Cells/microbiology , Host-Pathogen Interactions , Microbial Viability , Mitochondria/pathology , Mitochondrial Dynamics , Adenosine Triphosphate/metabolism , HeLa Cells , Humans , Mitochondria/metabolism , Models, Theoretical
7.
Biochem Biophys Res Commun ; 495(1): 353-359, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29108997

ABSTRACT

Fatty acid-binding protein 4 (FABP4), a cytosolic lipid chaperone predominantly expressed in adipocytes and macrophages, modulates lipid fluxes, trafficking, signaling, and metabolism. Recent studies have demonstrated that FABP4 regulates metabolic and inflammatory pathways, and in mouse models its inhibition can improve type 2 diabetes mellitus and atherosclerosis. However, the role of FABP4 in bacterial infection, metabolic crosstalk between host and pathogen, and bacterial pathogenesis have not been studied. As an obligate intracellular pathogen, Chlamydia pneumoniae needs to obtain nutrients such as ATP and lipids from host cells. Here, we show that C. pneumoniae successfully infects and proliferates in murine adipocytes by inducing hormone sensitive lipase (HSL)-mediated lipolysis. Chemical inhibition or genetic manipulation of HSL significantly abrogated the intracellular growth of C. pneumoniae in adipocytes. Liberated free fatty acids were utilized to generate ATP via ß-oxidation, which C. pneumoniae usurped for its replication. Strikingly, chemical inhibition or genetic silencing of FABP4 significantly abrogated C. pneumoniae infection-induced lipolysis and mobilization of liberated FFAs, resulting in reduced bacterial growth in adipocytes. Collectively, these results demonstrate that C. pneumoniae exploits host FABP4 to facilitate fat mobilization and intracellular replication in adipocytes. This work uncovers a novel strategy used by intracellular pathogens for acquiring energy via hijacking of the host lipid metabolism pathway.


Subject(s)
Adipocytes/microbiology , Adipocytes/physiology , Chlamydophila pneumoniae/physiology , Fatty Acid-Binding Proteins/metabolism , Lipid Mobilization/physiology , Sterol Esterase/metabolism , 3T3-L1 Cells , Animals , Cell Proliferation/physiology , Chlamydophila pneumoniae/cytology , Mice
8.
Cell ; 172(1-2): 191-204.e10, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29224778

ABSTRACT

Hematopoietic stem cell transplantation is a potential curative therapy for malignant and nonmalignant diseases. Improving the efficiency of stem cell collection and the quality of the cells acquired can broaden the donor pool and improve patient outcomes. We developed a rapid stem cell mobilization regimen utilizing a unique CXCR2 agonist, GROß, and the CXCR4 antagonist AMD3100. A single injection of both agents resulted in stem cell mobilization peaking within 15 min that was equivalent in magnitude to a standard multi-day regimen of granulocyte colony-stimulating factor (G-CSF). Mechanistic studies determined that rapid mobilization results from synergistic signaling on neutrophils, resulting in enhanced MMP-9 release, and unexpectedly revealed genetic polymorphisms in MMP-9 that alter activity. This mobilization regimen results in preferential trafficking of stem cells that demonstrate a higher engraftment efficiency than those mobilized by G-CSF. Our studies suggest a potential new strategy for the rapid collection of an improved hematopoietic graft.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Adult , Animals , Benzylamines , Chemokine CXCL2/pharmacology , Cyclams , Female , Hematopoietic Stem Cells/drug effects , Heterocyclic Compounds/pharmacology , Humans , Male , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred ICR , Polymorphism, Genetic
9.
Stem Cells Transl Med ; 4(4): 320-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25742692

ABSTRACT

Reprogramming human adult blood mononuclear cells (MNCs) cells by transient plasmid expression is becoming increasingly popular as an attractive method for generating induced pluripotent stem (iPS) cells without the genomic alteration caused by genome-inserting vectors. However, its efficiency is relatively low with adult MNCs compared with cord blood MNCs and other fetal cells and is highly variable among different adult individuals. We report highly efficient iPS cell derivation under clinically compliant conditions via three major improvements. First, we revised a combination of three EBNA1/OriP episomal vectors expressing five transgenes, which increased reprogramming efficiency by ≥10-50-fold from our previous vectors. Second, human recombinant vitronectin proteins were used as cell culture substrates, alleviating the need for feeder cells or animal-sourced proteins. Finally, we eliminated the previously critical step of manually picking individual iPS cell clones by pooling newly emerged iPS cell colonies. Pooled cultures were then purified based on the presence of the TRA-1-60 pluripotency surface antigen, resulting in the ability to rapidly expand iPS cells for subsequent applications. These new improvements permit a consistent and reliable method to generate human iPS cells with minimal clonal variations from blood MNCs, including previously difficult samples such as those from patients with paroxysmal nocturnal hemoglobinuria. In addition, this method of efficiently generating iPS cells under feeder-free and xeno-free conditions allows for the establishment of clinically compliant iPS cell lines for future therapeutic applications.


Subject(s)
Cell Culture Techniques , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Adult , Blood Cells/cytology , Culture Media, Serum-Free , Feeder Cells/cytology , Flow Cytometry , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/transplantation , Leukocytes, Mononuclear/cytology , Vitronectin/administration & dosage
10.
PLoS One ; 10(1): e0116933, 2015.
Article in English | MEDLINE | ID: mdl-25635918

ABSTRACT

Induced pluripotent stem (iPS) cells are at the forefront of research in regenerative medicine and are envisaged as a source for personalized tissue repair and cell replacement therapy. Here, we demonstrate for the first time that oligodendrocyte progenitors (OPs) can be derived from iPS cells generated using either an episomal, non-integrating plasmid approach or standard integrating retroviruses that survive and differentiate into mature oligodendrocytes after early transplantation into the injured spinal cord. The efficiency of OP differentiation in all 3 lines tested ranged from 40% to 60% of total cells, comparable to those derived from human embryonic stem cells. iPS cell lines derived using episomal vectors or retroviruses generated a similar number of early neural progenitors and glial progenitors while the episomal plasmid-derived iPS line generated more OPs expressing late markers O1 and RIP. Moreover, we discovered that iPS-derived OPs (iPS-OPs) engrafted 24 hours following a moderate contusive spinal cord injury (SCI) in rats survived for approximately two months and that more than 70% of the transplanted cells differentiated into mature oligodendrocytes that expressed myelin associated proteins. Transplanted OPs resulted in a significant increase in the number of myelinated axons in animals that received a transplantation 24 h after injury. In addition, nearly a 5-fold reduction in cavity size and reduced glial scarring was seen in iPS-treated groups compared to the control group, which was injected with heat-killed iPS-OPs. Although further investigation is needed to understand the mechanisms involved, these results provide evidence that patient-specific, iPS-derived OPs can survive for three months and improve behavioral assessment (BBB) after acute transplantation into SCI. This is significant as determining the time in which stem cells are injected after SCI may influence their survival and differentiation capacity.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Animals , Axons/physiology , Cell Differentiation , Cell Survival , Cells, Cultured , Early Medical Intervention , Female , Humans , Motor Activity , Myelin Sheath/physiology , Nerve Regeneration , Oligodendroglia/physiology , Rats, Inbred Lew , Recovery of Function , Spinal Cord/pathology , Spinal Cord/physiopathology , Treatment Outcome
11.
Biochem Biophys Res Commun ; 452(3): 689-94, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25193701

ABSTRACT

Chlamydia pneumoniae is an obligate intracellular pathogen that replicates within a vacuole and acquires host cell nutrients. We show that C. pneumoniae utilizes host innate immune signaling NLRP3/ASC/caspase-1 inflammasome for intracellular growth. Bone marrow-derived macrophages (BMMs) secreted mature interleukin-1ß upon infection with C. pneumoniae depending on the NLRP3 inflammasome activation. Intracellular growth of C. pneumoniae was severely impaired in BMMs from Nlrp3(-/-), Asc(-/-), and Casp1(-/-) mice but not wild type or Nlrc4(-/-) mice. Furthermore defective NLRP3 inflammasome components led to accumulation of lipid droplets inside the infected BMMs, suggesting that uptake and/or utilization of lipids is disturbed in the absence of NLRP3 inflammasome activation. These results suggest C. pneumoniae has evolved to harness both host innate immune response and NLRP3 inflammasome activation, for the acquisition of essential nutrients necessary for intracellular growth. This unique property of C. pneumoniae may shed a new light on how C. pneumoniae increase the risk of atherosclerosis and metabolic syndrome.


Subject(s)
Carrier Proteins/genetics , Caspase 1/genetics , Chlamydophila pneumoniae/physiology , Inflammasomes/metabolism , Macrophages/enzymology , Signal Transduction/drug effects , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Biological Transport , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Bone Marrow Cells/microbiology , CARD Signaling Adaptor Proteins , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Carrier Proteins/immunology , Caspase 1/deficiency , Caspase 1/immunology , Enzyme Activation , Gene Expression Regulation , Host-Pathogen Interactions , Immunity, Cellular , Immunity, Innate , Inflammasomes/immunology , Interleukin-1beta/metabolism , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Primary Cell Culture
12.
Stem Cells ; 32(2): 349-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23765875

ABSTRACT

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) shares much similarity to the cancer initiation process, and the molecular mechanisms underlying both processes remain to be elucidated. Here, we report that a tumor- or embryonic stem cell-specific Ras gene ERas, which encodes a constitutively active form of GTPase, and its downstream Phosphoinositide-3 kinase/Akt signaling pathway are important facilitators for the somatic reprogramming process. We found that overexpression of ERas retrovirally enhanced mouse iPSC induction while ERas knockdown repressed it. Modulation of Akt signaling by genetic or chemical means greatly impacted the reprogramming efficiency. Forced expression of a constitutively active Akt1 gene could rescue the reduced efficiency resulting from ERas knockdown, and point-mutation analyses further revealed that ERas is tightly coupled with Akt signaling to enhance reprogramming. Mechanistically, the forkhead transcription factor FoxO1 can function as a barrier to the iPSC induction, and the inactivation of FoxO1 by Akt-dependent phosphorylation largely accounts for the enhancing effect of ERas-Akt signaling on reprogramming. Collectively, these results unravel the significance of the ERas-Akt-FoxO1 signaling axis in iPSC generation, suggesting a possibly shared molecular basis for both somatic reprogramming and cancer initiation.


Subject(s)
Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/genetics , Oncogene Protein p21(ras)/genetics , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Forkhead Box Protein O1 , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics
13.
Cell Stem Cell ; 13(3): 261-2, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-24012365

ABSTRACT

While most factors used as reprogramming transgenes can be replaced by other means, Oct4 has remained essential until now. Three recent papers have now broken this barrier through the use of opposing lineage specifying transgenes and chemical modulation, thus signifying a milestone in advancing our understanding of pluripotency induction.


Subject(s)
Cell Dedifferentiation , Cell Engineering/methods , Cellular Reprogramming/drug effects , Fibroblasts/drug effects , Fibroblasts/physiology , GATA3 Transcription Factor/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/physiology , Small Molecule Libraries/pharmacology , Transcription Factors/metabolism , Animals , Humans
14.
Stem Cell Res ; 11(3): 1103-16, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23973800

ABSTRACT

Large-scale production of human induced pluripotent stem cells (hiPSCs) by robust and economic methods has been one of the major challenges for translational realization of hiPSC technology. Here we demonstrate a scalable culture system for hiPSC expansion using the E8 chemically defined and xeno-free medium under either adherent or suspension conditions. To optimize suspension conditions guided by a computational simulation, we developed a method to efficiently expand hiPSCs as undifferentiated aggregates in spinner flasks. Serial passaging of two different hiPSC lines in the spinner flasks using the E8 medium preserved their normal karyotype and expression of undifferentiated state markers of TRA-1-60, SSEA4, OCT4, and NANOG. The hiPSCs cultured in spinner flasks for more than 10 passages not only could be remained pluripotent as indicated by in vitro and in vivo assays, but also could be efficiently induced toward mesodermal and hematopoietic differentiation. Furthermore, we established a xeno-free protocol of single-cell cryopreservation and recovery for the scalable production of hiPSCs in spinner flasks. This system is the first to enable an efficient scale-up bioprocess in completely xeno-free condition for the expansion and cryopreservation of hiPSCs with the quantity and quality compliant for clinical applications.


Subject(s)
Cell Culture Techniques , Culture Media/chemistry , Induced Pluripotent Stem Cells/cytology , Antigens, Surface/metabolism , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Proteoglycans/metabolism , Stage-Specific Embryonic Antigens/metabolism
16.
J Dermatol Sci ; 70(3): 166-72, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23452544

ABSTRACT

BACKGROUND: 1α,25-Dihydroxyvitamin D3 (1,25(OH)2D3) up-regulates the production of human cathelicidin antimicrobial peptide (CAMP) from monocytes/macrophages infected with Mycobacterium tuberculosis (M. tbc). CAMP facilitates the co-localization of autophagolysosomes with M. tbc, promoting the antimicrobial activity of monocytes. Mycobacterium marinum (M. marinum) is an acid-fast bacillus that causes less severe granulomatous skin lesions compared with M. tbc. OBJECTIVE: We investigated whether autophagic antimicrobial activity is promoted by 1,25(OH)2D3 or C-terminal of cathelicidin LL-37 in human monocytes upon infection with M. marinum. METHODS: Human monocytes (THP-1) were infected with M. marinum. Effects of simultaneous treatments of 1,25(OH)2D3, exogenous LL-37 peptide, autophagolysosome inhibitors, 3-methyladenine or chloroquine, were examined. RESULTS: CAMP was strongly induced by adding 1,25(OH)2D3 to the culture of THP-1 cells. In the absence of 1,25(OH)2D3 M. marinum infection alone did not induce CAMP, however, simultaneous addition of 1,25(OH)2D3 to M. marinum infection accelerated CAMP production more than 1,25(OH)2D3 alone. Proliferation of M. marinum was markedly decreased in the presence of 1,25(OH)2D3 or exogenous LL-37 in THP-1 cells. Co-localization of CAMP with autophagolysosome was evident in 1,25(OH)2D3 and LL-37 treated THP-1 cells after M. marinum infection. Autophagolysosome inhibitors abrogated the antimicrobial effects of 1,25(OH)2D3 and exogenous LL-37 against M. marinum infection in THP-1 cells. CONCLUSIONS: Human monocytic cells, whose CAMP production is up-regulated by 1,25(OH)2D3-vitamin D receptor pathway, accelerate antimicrobial function of autophagolysosome in M. marinum infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/metabolism , Autophagy/drug effects , Calcitriol/pharmacology , Monocytes/drug effects , Mycobacterium Infections, Nontuberculous/prevention & control , Mycobacterium marinum/drug effects , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Antimicrobial Cationic Peptides/pharmacology , Autophagy-Related Protein 5 , Cell Line, Tumor , Chloroquine/pharmacology , Cytokines/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/microbiology , Humans , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Monocytes/pathology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium Infections, Nontuberculous/pathology , RNA Interference , Receptors, Calcitriol/agonists , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Time Factors , Transfection , Up-Regulation , Cathelicidins
17.
PLoS One ; 8(3): e59633, 2013.
Article in English | MEDLINE | ID: mdl-23527234

ABSTRACT

General cellular functions of proteasomes occur through protein degradation, whereas the specific function of immunoproteasomes is the optimization of antigen processing associated with MHC class I. We and others previously reported that deficiency in subunits of immunoproteasomes impaired the activation of antigen-specific CD8(+) T cells, resulting in higher susceptibility to tumor and infections. We demonstrated that CD8(+) T cells contributed to protection against malaria parasites. In this study, we evaluated the role of immunoproteasomes in the course of infection with rodent malaria parasites. Unexpectedly, Plasmodium yoelii infection of mice deficient in LMP7, a catalytic subunit of immunoproteasomes, showed lower parasite growth in the early phase of infection and lower lethality compared with control mice. The protective characteristics of LMP7-deficient mice were not associated with enhanced immune responses, as the mutant mice showed comparable or diminished activation of innate and acquired immunity. The remarkable difference was observed in erythrocytes instead of immune responses. Parasitized red blood cells (pRBCs) purified from LMP7-deficient mice were more susceptible to phagocytosis by macrophages compared with those from wild-type mice. The susceptibility of pRBC to phagocytosis appeared to correlate with deformity of the membrane structures that were only observed after infection. Our results suggest that RBCs of LMP7-deficient mice were more likely to deform in response to infection with malaria parasites, presumably resulting in higher susceptibility to phagocytosis and in the partial resistance to malaria.


Subject(s)
Disease Resistance/physiology , Erythrocytes/parasitology , Malaria/physiopathology , Phagocytosis/physiology , Plasmodium yoelii , Proteasome Endopeptidase Complex/physiology , Animals , DNA Primers/genetics , Erythrocytes/ultrastructure , Flow Cytometry , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Proteasome Endopeptidase Complex/deficiency , Proteasome Endopeptidase Complex/metabolism , Real-Time Polymerase Chain Reaction
18.
Nat Protoc ; 7(11): 2013-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23080273

ABSTRACT

Several human postnatal somatic cell types have been successfully reprogrammed to induced pluripotent stem cells (iPSCs). Blood mononuclear cells (MNCs) offer several advantages compared with other cell types. They are easily isolated from umbilical cord blood (CB) or adult peripheral blood (PB), and can be used fresh or after freezing. A short culture allows for more efficient reprogramming, with iPSC colonies forming from blood MNCs in 14 d, compared with 28 d for age-matched fibroblastic cells. The advantages of briefly cultured blood MNCs may be due to favorable epigenetic profiles and gene expression patterns. Blood cells from adults, especially nonlymphoid cells that are replenished frequently from intermittently activated blood stem cells, are short-lived in vivo and may contain less somatic mutations than skin fibroblasts, which are more exposed to environmental mutagens over time. We describe here a detailed, validated protocol for effective generation of integration-free human iPSCs from blood MNCs by plasmid vectors.


Subject(s)
Cell Culture Techniques , Induced Pluripotent Stem Cells/cytology , Leukocytes, Mononuclear/cytology , Genetic Vectors , Humans , Plasmids/genetics , Transfection
19.
J Immunol ; 189(4): 1618-26, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22802414

ABSTRACT

Aquaporin-1 (AQP-1) is a water channel protein highly expressed in the vascular endothelial cells of proliferating tissues including malignant cancers. Given that in APC ubiquitinated peptides are effectively introduced into proteasomes from which CD8 epitopes are excised, we fused ubiquitin with AQP-1 (pUB-AQP-1) to produce a DNA vaccine. In C57BL/6J mice immunized with pUB-AQP-1, the growth of B16F10 melanoma was profoundly inhibited. The antitumor effect of the pUB-AQP-1 DNA vaccine was largely mediated by CD8 T cells, which secrete IFN-γ, perforin, and granzyme-B in the presence of APCs transfected with pUB-AQP-1. AQP-1-specific CD8 T cells possessed cytotoxic activity both in vivo and in vitro. After tumor challenge, the microvessel density decreased and the ratio of total blood vessel area to tumor area was significantly reduced as compared with control mice, resulting in a dramatic suppression of tumor growth. The immunization effect was completely abrogated in immunoproteasome-deficient mice. Strikingly this pUB-AQP-1 DNA vaccine was also effective against Colon 26 colon tumors (BALB/c) and MBT/2 bladder tumors (C3H/HeN). Thus, this ubiquitin-conjugated DNA immunization-targeting tumor vasculature is a valid and promising antitumor therapy. This vaccine works across the barriers of tumor species and MHC class I differences in host mice.


Subject(s)
Aquaporin 1/immunology , Cancer Vaccines/pharmacology , Neoplasms, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/pharmacology , Animals , Blotting, Western , Cancer Vaccines/immunology , Female , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/immunology , Neovascularization, Pathologic/therapy , Proteasome Endopeptidase Complex/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Ubiquitin/immunology , Vaccines, DNA/immunology
20.
Int J Hematol ; 95(6): 601-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22619021

ABSTRACT

Postnatal hematopoietic stem cells (HSCs) from umbilical cord blood and adult marrow/blood have been successfully used for treating various human diseases in the past several decades. However, the availability of optimal numbers of HSCs from autologous patients or allogeneic donors with adequate match remains a great barrier to improve and extend HSC and marrow transplantation to more needing patients. In addition, the inability to expand functional human HSCs to sufficient quantity in the laboratory has hindered our research and understanding of human HSCs and hematopoiesis. Recent development in reprogramming technology has provided patient-specific pluripotent stem cells (iPSCs) as a powerful enabling tool for modeling disease and developing therapeutics. Studies have demonstrated the potential of human iPSCs, which can be expanded exponentially and amenable for genome engineering, for using in modeling both inherited and acquired blood diseases. Proof-of-principle studies have also shown the feasibility of iPSCs in gene and cell therapy. Here, we review the recent development in iPSC-based blood disease modeling, and discuss the unsolved issues and challenges in this new and promising field.


Subject(s)
Hematologic Diseases/therapy , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Animals , Cell Differentiation , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...