Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Inflamm Bowel Dis ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38655866

ABSTRACT

BACKGROUND: Endoscopic healing is generally defined as Mayo endoscopic subscore (MES) ≤1 in ulcerative colitis (UC). However, patients with an MES of 1 are at higher relapse risk than those with an MES of 0. This study evaluated the therapeutic efficacy of proactive dose escalation of oral 5-aminosalicylic acid (5-ASA) in UC patients with an MES of 1. METHODS: An open-label, randomized controlled trial was conducted in 5 hospitals between 2018 and 2022. Ulcerative colitis patients in clinical remission under oral 5-ASA therapy and diagnosed as having an MES of 1 were enrolled. Patients receiving maintenance therapy other than 5-ASA and immunomodulator were excluded. Patients were randomly assigned in a 1:1 ratio to receive either a dose-escalated (intervention) or constant dose (control) of 5-ASA. Concomitant immunomodulator was used as the stratification factor in the randomization. The primary end point was relapse within 1 year. The subgroup analysis was stratified for the use of immunomodulators. RESULTS: The full analysis set included 79 patients (39 intervention and 40 control). Immunomodulators were used in 20 (25.3%) patients. Relapse was less in the intervention group (15.4%) than the control group (37.5%; P = .026). In the subgroup with concomitant immunomodulators, relapse was also less in the intervention group (10.0%) than the control group (70.0%; P = .020). In patients without immunomodulators, the difference was not significant between 2 groups (intervention, 17.2%; control, 26.7%; P = .53). CONCLUSIONS: Dose escalation of 5-ASA reduced relapse within 1 year in UC patients in clinical remission with an MES of 1.


Dose escalation of 5-aminosalicylic acid for ulcerative colitis reduced relapse rate in patients in clinical remission with a Mayo endoscopic subscore of 1. The therapeutic efficacy was more evident in those whom immunomodulators were used.

2.
Front Med (Lausanne) ; 9: 681316, 2022.
Article in English | MEDLINE | ID: mdl-35360717

ABSTRACT

This study was conducted to evaluate the safety and efficacy of human peripheral blood CD34 positive (CD34+) cells transplanted into a murine chronic stroke model to obtain pre-clinical proof of concept, prior to clinical testing. Granulocyte colony stimulating factor (G-CSF) mobilized human CD34+ cells [1 × 104 cells in 50 µl phosphate-buffered saline (PBS)] were intravenously (iv) or intra-carotid arterially (ia) transplanted 4 weeks after the induction of stroke (chronic stage), and neurological function was evaluated. In this study, severe combined immune deficiency (SCID) mice were used to prevent excessive immune response after cell therapy. Two weeks post cell therapy, the ia CD34+ cells group demonstrated a significant improvement in neurological functions compared to the PBS control. The therapeutic effect was maintained 8 weeks after the treatment. Even after a single administration, ia transplantation of CD34+ cells had a significant therapeutic effect on chronic stroke. Based on the result of this pre-clinical proof of concept study, a future clinical trial of autologous peripheral blood CD34+ cells administration in the intra-carotid artery for chronic stroke patients is planned.

3.
Nat Commun ; 11(1): 5627, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159072

ABSTRACT

The filamentous fungus Neurospora crassa is used as a model organism for genetics, developmental biology and molecular biology. Remarkably, it is not known to host or to be susceptible to infection with any viruses. Here, we identify diverse RNA viruses in N. crassa and other Neurospora species, and show that N. crassa supports the replication of these viruses as well as some viruses from other fungi. Several encapsidated double-stranded RNA viruses and capsid-less positive-sense single-stranded RNA viruses can be experimentally introduced into N. crassa protoplasts or spheroplasts. This allowed us to examine viral replication and RNAi-mediated antiviral responses in this organism. We show that viral infection upregulates the transcription of RNAi components, and that Dicer proteins (DCL-1, DCL-2) and an Argonaute (QDE-2) participate in suppression of viral replication. Our study thus establishes N. crassa as a model system for the study of host-virus interactions.


Subject(s)
Neurospora crassa/virology , RNA Viruses/physiology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Neurospora crassa/genetics , Neurospora crassa/metabolism , RNA Viruses/genetics , RNA Viruses/growth & development , Virus Replication
4.
EBioMedicine ; 57: 102862, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32629392

ABSTRACT

BACKGROUND: Bone marrow stem cell clonal dysfunction by somatic mutation is suspected to affect post-infarction myocardial regeneration after coronary bypass surgery (CABG). METHODS: Transcriptome and variant expression analysis was studied in the phase 3 PERFECT trial post myocardial infarction CABG and CD133+ bone marrow derived hematopoetic stem cells showing difference in left ventricular ejection fraction (∆LVEF) myocardial regeneration Responders (n=14; ∆LVEF +16% day 180/0) and Non-responders (n=9; ∆LVEF -1.1% day 180/0). Subsequently, the findings have been validated in an independent patient cohort (n=14) as well as in two preclinical mouse models investigating SH2B3/LNK antisense or knockout deficient conditions. FINDINGS: 1. Clinical: R differed from NR in a total of 161 genes in differential expression (n=23, q<0•05) and 872 genes in coexpression analysis (n=23, q<0•05). Machine Learning clustering analysis revealed distinct RvsNR preoperative gene-expression signatures in peripheral blood acorrelated to SH2B3 (p<0.05). Mutation analysis revealed increased specific variants in RvsNR. (R: 48 genes; NR: 224 genes). 2. Preclinical:SH2B3/LNK-silenced hematopoietic stem cell (HSC) clones displayed significant overgrowth of myeloid and immune cells in bone marrow, peripheral blood, and tissue at day 160 after competitive bone-marrow transplantation into mice. SH2B3/LNK-/- mice demonstrated enhanced cardiac repair through augmenting the kinetics of bone marrow-derived endothelial progenitor cells, increased capillary density in ischemic myocardium, and reduced left ventricular fibrosis with preserved cardiac function. 3. VALIDATION: Evaluation analysis in 14 additional patients revealed 85% RvsNR (12/14 patients) prediction accuracy for the identified biomarker signature. INTERPRETATION: Myocardial repair is affected by HSC gene response and somatic mutation. Machine Learning can be utilized to identify and predict pathological HSC response. FUNDING: German Ministry of Research and Education (BMBF): Reference and Translation Center for Cardiac Stem Cell Therapy - FKZ0312138A and FKZ031L0106C, German Ministry of Research and Education (BMBF): Collaborative research center - DFG:SFB738 and Center of Excellence - DFG:EC-REBIRTH), European Social Fonds: ESF/IV-WM-B34-0011/08, ESF/IV-WM-B34-0030/10, and Miltenyi Biotec GmbH, Bergisch-Gladbach, Germany. Japanese Ministry of Health : Health and Labour Sciences Research Grant (H14-trans-001, H17-trans-002) TRIAL REGISTRATION: ClinicalTrials.gov NCT00950274.


Subject(s)
AC133 Antigen/genetics , Bone Marrow Transplantation/methods , Coronary Artery Disease/therapy , Hematopoietic Stem Cell Transplantation/methods , Myocardial Ischemia/therapy , Adolescent , Adult , Aged , Bone Marrow Cells/cytology , Cellular Senescence/genetics , Coronary Artery Disease/genetics , Coronary Artery Disease/physiopathology , Female , Heart/growth & development , Heart/physiopathology , Hematopoietic Stem Cells/cytology , Humans , Male , Middle Aged , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Regeneration/genetics , Young Adult
5.
Proc Natl Acad Sci U S A ; 113(41): E6135-E6144, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27681634

ABSTRACT

DNA methylation, heterochromatin protein 1 (HP1), histone H3 lysine 9 (H3K9) methylation, histone deacetylation, and highly repeated sequences are prototypical heterochromatic features, but their interrelationships are not fully understood. Prior work showed that H3K9 methylation directs DNA methylation and histone deacetylation via HP1 in Neurospora crassa and that the histone deacetylase complex HCHC is required for proper DNA methylation. The complex consists of the chromodomain proteins HP1 and chromodomain protein 2 (CDP-2), the histone deacetylase HDA-1, and the AT-hook motif protein CDP-2/HDA-1-associated protein (CHAP). We show that the complex is required for proper chromosome segregation, dissect its function, and characterize interactions among its components. Our analyses revealed the existence of an HP1-based DNA methylation pathway independent of its chromodomain. The pathway partially depends on CHAP but not on the CDP-2 chromodomain. CDP-2 serves as a bridge between the recognition of H3K9 trimethylation (H3K9me3) by HP1 and the histone deacetylase activity of HDA-1. CHAP is also critical for HDA-1 localization to heterochromatin. Specifically, the CHAP zinc finger interacts directly with the HDA-1 argonaute-binding protein 2 (Arb2) domain, and the CHAP AT-hook motifs recognize heterochromatic regions by binding to AT-rich DNA. Our data shed light on the interrelationships among the prototypical heterochromatic features and support a model in which dual recognition by the HP1 chromodomain and the CHAP AT-hooks are required for proper heterochromatin formation.

6.
Cell Transplant ; 24(7): 1379-93, 2015.
Article in English | MEDLINE | ID: mdl-24800622

ABSTRACT

We recently demonstrated that the local transplantation of human peripheral blood (PB) CD34(+) cells, an endothelial/hematopoietic progenitor cell-rich population, contributes to fracture repair via vasculogenesis/angiogenesis and osteogenesis. Human PB mononuclear cells (MNCs) are also considered a potential cell fraction for neovascularization. We have previously shown the feasibility of human PB MNCs to enhance fracture healing. However, there is no report directly comparing the efficacy for fracture repair between CD34(+) cells and MNCs. In addition, an unhealing fracture model, which does not accurately resemble a clinical setting, was used in our previous studies. To overcome these issues, we compared the capacity of human granulocyte colony-stimulating factor-mobilized PB (GM-PB) CD34(+) cells and human GM-PB MNCs in a nonunion model, which more closely resembles a clinical setting. First, the effect of local transplantation of 1 × 10(5) GM-PB CD34(+) cells (CD34(+) group), 1 × 10(7) GM-PB MNCs (containing approximately 1 × 10(5) GM-PB CD34(+) cells) (MNC group), and phosphate-buffered saline (PBS) (PBS group) on nonunion healing was compared. Similar augmentation of blood flow recovery at perinonunion sites was observed in the CD34(+) and MNC groups. Meanwhile, a superior effect on nonunion repair was revealed by radiological, histological, and functional assessment in the CD34(+) group compared with the other groups. Moreover, through in vivo and in vitro experiments, excessive inflammation induced by GM-PB MNCs was confirmed and believed to be one of the mechanisms underlying this potency difference. These results strongly suggest that local transplantation of GM-PB CD34(+) cells is a practical and effective strategy for treatment of nonunion after fracture.


Subject(s)
Antigens, CD34/metabolism , Hematopoietic Stem Cell Transplantation/methods , Leukocytes, Mononuclear/metabolism , Transplantation Conditioning/methods , Wound Healing/drug effects , Cell Differentiation , Fractures, Bone , Humans
7.
Cell Transplant ; 21(12): 2689-709, 2012.
Article in English | MEDLINE | ID: mdl-22943882

ABSTRACT

Transplantation of bone marrow (BM) CD34(+) cells, an endothelial/hematopoietic progenitor-enriched cell population, has shown therapeutic efficiency in the treatment of ischemic diseases enhancing neovascularization. However, the number of CD34(+) cells obtained from bone marrow is not sufficient for routine clinical application. To overcome this issue, we developed a more efficient and clinically applicable CD34(+) cell expansion method. Seven-day ex vivo expansion culture of BM CD34(+) cells with a cocktail of five growth factors containing VEGF, SCF, IL-6, Flt-3 ligand, and TPO resulted in reproducible more than 20-fold increase in cell number. The favorable effect of the local transplantation of culture expanded (cEx)-BM CD34(+) cells on rat unhealing fractures was equivalent or higher than that of nonexpanded (fresh) BM CD34(+) cells exhibiting sufficient therapeutic outcome with frequent vasculogenic/osteogenic differentiation of transplanted cEx-BM CD34(+) cells and fresh BM CD34(+) cells as well as intrinsic enhancement of angiogenesis/osteogenesis at the treated fracture sites. Specifically, cEx-BM CD34(+) cell treatment demonstrated the best blood flow recovery at fracture sites compared with the nonexpanded BM CD34(+) cells. In vitro, cEx-BM CD34(+) cells showed higher colony/tube-forming capacity than nonexpanded BM CD34(+) cells. Both cells demonstrated differentiation potential into osteoblasts. Since fresh BM CD34(+) cells can be easily collected from fracture sites at the time of primary operation and stored for future use, autologous cEx-BM CD34(+) cell transplantation would be not only a simple but also a promising therapeutic strategy for unhealing fractures in the field of orthopedic trauma surgery.


Subject(s)
Antigens, CD34/metabolism , Bone Marrow Cells/cytology , Femoral Fractures/therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Animals , Blood Flow Velocity , Bone and Bones/blood supply , Cell Differentiation , Cell Proliferation/drug effects , Cells, Cultured , Female , Femoral Fractures/diagnostic imaging , Femoral Fractures/pathology , Hematopoietic Stem Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Neovascularization, Pathologic , Osteogenesis , Rats , Rats, Nude , Tomography, X-Ray Computed , Young Adult
8.
J Mol Cell Cardiol ; 51(3): 308-17, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21557947

ABSTRACT

Although endothelial progenitor cells (EPCs) differentiate from minor populations of stem cells in bone marrow (BM), the differential role of hematopoietic stem cell (HSC) subpopulations in EPC development is largely unclear. Morphological characterization of EPC colonies has revealed that c-kit+/Sca-1+/lineage (Lin)-(KSL) cells mainly develop small EPC-colony forming units (CFUs) not large EPC-CFUs. In contrast, c-kit+/Sca-1-/Lin- (KL) cells develop large EPC-CFUs not small EPC-CFUs. Neither c-kit-/Sca-1+/Lin- (SL) cells nor c-kit-/Sca-1-/Lin- (L) cells develop EPC-CFUs to an appreciable extent. Hindlimb ischemia enhances formation of large EPC-CFUs from all HSC subpopulations, suggesting an important role for ischemia in functional EPC development. Real time RT-PCR analysis shows that KSL, KL and SL cells but not L cells express various factors at high levels, maintaining a BM-EPC pool. In hindlimb ischemia, transplanted KSL, KL and SL cells efficiently differentiate into endothelial lineage cells in situ and augment capillary density. The percentage of Ki-67+ cycling cells among transplanted cells in ischemic tissue was also greater for KSL, KL and SL cells than L cells. Moreover, the frequency of VEGF- or SDF-1-expressing cells was higher transplanted KSL, KL or SL cells than L cells. Thus, KSL, KL and SL cells are not different in their angiogenic competence under ischemic conditions. In conclusion, although KSL cells are clearly the most potent contributors to EPC development, KL and SL cells may also contribute to neovascularization via both autocrine and paracrine mechanisms in response to ischemic signals.


Subject(s)
Bone Marrow Cells/metabolism , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Ischemia/metabolism , Neovascularization, Physiologic , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Hypoxia/genetics , Cell Proliferation , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Ischemia/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Physiologic/genetics
9.
Lab Invest ; 91(4): 539-52, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21135814

ABSTRACT

Human multipotent adipose-derived stem cells (hMADSCs) have recently been isolated featuring extensive expansion capacity ex vivo. We tested the hypothesis that hMADSC transplantation might contribute to cardiac functional recovery by its direct or indirect effect on myocardial infarction (MI). Nude rats were either transplanted with hMADSCs or PBS (control) in ischemic myocardium immediately following MI. Echocardiographical assessment of cardiac function after MI with hMADSCs showed significant improvement of each parameter compared to that with PBS. Histological analysis also showed significantly reduced infarct size and increased capillary density in peri-infarct myocardium by hMADSC treatment. However, remarkable transdifferentiation of hMADSCs into cardiac or vascular lineage cells was not observed. Despite the less transdifferentiation capacity, hMADSCs produced robust multiple pro-angiogenic growth factors and chemokines, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and stromal cell-derived factor-1α (SDF-1α). Specifically, hMADSC-derived SDF-1α had a crucial role for cooperative angiogenesis, with the paracrine effect of hMADSCs and Tie2-positive bone marrow (BM) progenitor recruitment in ischemic myocardium. hMADSCs exhibit a therapeutic effect on cardiac preservation following MI, with the production of VEGF, bFGF, and SDF-1α showing paracrine effects and endogenous BM stem/progenitor recruitment to ischemic myocardium rather than its direct contribution to tissue regeneration.


Subject(s)
Adipocytes/transplantation , Myocardial Ischemia/surgery , Stem Cell Transplantation , Animals , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Capillaries/pathology , Cell Differentiation , Chemokine CXCL12 , Coronary Vessels/pathology , Echocardiography , Fibroblast Growth Factors , Heart/physiopathology , Humans , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Ischemia/diagnosis , Myocardial Ischemia/etiology , Myocardial Ischemia/physiopathology , Myocardium/pathology , Neovascularization, Physiologic , Paracrine Communication , Rats , Rats, Nude , Stem Cells/pathology , Vascular Endothelial Growth Factor A/metabolism
10.
Cell Transplant ; 20(9): 1491-6, 2011.
Article in English | MEDLINE | ID: mdl-21176407

ABSTRACT

Although implantation of crude bone marrow cells has been applied in a small number of patients for fracture healing, transplantation of peripheral blood CD34(+) cells, the hematopoietic/endothelial progenitor cell-enriched population, in patients with fracture has never been reported. Here, we report the first case of tibial nonunion receiving autologous, granulocyte colony stimulating factor mobilized CD34(+) cells accompanied with autologous bone grafting. No serious adverse event occurred, and the novel therapy performed 9 months after the primary operation resulted in bone union 3 months later without any symptoms including pain and gait disturbance.


Subject(s)
Antigens, CD34/metabolism , Fractures, Ununited/therapy , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Tibial Fractures/therapy , Adult , Fractures, Ununited/diagnostic imaging , Humans , Male , Perioperative Care , Tibial Fractures/diagnostic imaging , Tomography, X-Ray Computed
11.
PLoS One ; 6(12): e28639, 2011.
Article in English | MEDLINE | ID: mdl-22216102

ABSTRACT

Numerous endothelial progenitor cell (EPC)-related investigations have been performed in mouse experiments. However, defined characteristics of mouse cultured EPC have not been examined. We focused on fast versus slow adherent cell population in bone marrow mononuclear cells (BMMNCs) in culture and examined their characteristics. After 24 h-culture of BMMNCs, attached (AT) cells and floating (FL) cells were further cultured in endothelial differentiation medium separately. Immunological and molecular analyses exhibited more endothelial-like and less monocyte/macrophage-like characteristics in FL cells compared with AT cells. FL cells formed thick/stable tube and hypoxia or shear stress overload further enhanced these endothelial-like features with increased angiogenic cytokine/growth factor mRNA expressions. Finally, FL cells exhibited therapeutic potential in a mouse myocardial infarction model showing the specific local recruitment to ischemic border zone and tissue preservation. These findings suggest that slow adherent (FL) but not fast attached (AT) BMMNCs in culture are EPC-rich population in mouse.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation , Monocytes/cytology , Stem Cells/cytology , Animals , Cell Culture Techniques , Flow Cytometry , Mice
12.
J Exp Med ; 207(10): 2207-23, 2010 Sep 27.
Article in English | MEDLINE | ID: mdl-20855498

ABSTRACT

The therapeutic potential of hematopoietic stem cells/endothelial progenitor cells (HSCs/EPCs) for fracture healing has been demonstrated with evidence for enhanced vasculogenesis/angiogenesis and osteogenesis at the site of fracture. The adaptor protein Lnk has recently been identified as an essential inhibitor of stem cell factor (SCF)-cKit signaling during stem cell self-renewal, and Lnk-deficient mice demonstrate enhanced hematopoietic reconstitution. In this study, we investigated whether the loss of Lnk signaling enhances the regenerative response during fracture healing. Radiological and histological examination showed accelerated fracture healing and remodeling in Lnk-deficient mice compared with wild-type mice. Molecular, physiological, and morphological approaches showed that vasculogenesis/angiogenesis and osteogenesis were promoted in Lnk-deficient mice by the mobilization and recruitment of HSCs/EPCs via activation of the SCF-cKit signaling pathway in the perifracture zone, which established a favorable environment for bone healing and remodeling. In addition, osteoblasts (OBs) from Lnk-deficient mice had a greater potential for terminal differentiation in response to SCF-cKit signaling in vitro. These findings suggest that inhibition of Lnk may have therapeutic potential by promoting an environment conducive to vasculogenesis/angiogenesis and osteogenesis and by facilitating OB terminal differentiation, leading to enhanced fracture healing.


Subject(s)
Fracture Healing , Osteogenesis , Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Differentiation , Fractures, Bone/metabolism , Fractures, Bone/physiopathology , Fractures, Bone/therapy , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Neovascularization, Pathologic , Neovascularization, Physiologic , Osteoblasts/metabolism , Osteoblasts/pathology , Osteogenesis/genetics , Proteins/genetics , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology
13.
Stem Cells ; 28(2): 365-75, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19859984

ABSTRACT

Lnk is an intracellular adaptor protein reported as a negative regulator of proliferation in c-Kit positive, Sca-1 positive, lineage marker-negative (KSL) bone marrow cells. The KSL fraction in mouse bone marrow is believed to represent a population of hematopoietic and endothelial progenitor cells (EPCs). We report here that, in vitro, Lnk(-/-) KSL cells form more EPC colonies than Lnk(+/+) KSL cells and show higher expression levels of endothelial marker genes, including CD105, CD144, Tie-1, and Tie2, than their wild-type counterparts. In vivo, the administration of Lnk(+/+) KSL cells to a mouse spinal cord injury model promoted angiogenesis, astrogliosis, axon growth, and functional recovery following injury, with Lnk(-/-) KSL being significantly more effective in inducing and promoting these regenerative events. At day 3 following injury, large vessels could be observed in spinal cords treated with KSL cells, and reactive astrocytes were found to have migrated along these large vessels. We could further show that the enhancement of astrogliosis appears to be caused in conjunction with the acceleration of angiogenesis. These findings suggest that Lnk deletion reinforces the commitment of KSL cells to EPCs, promoting subsequent repair of injured spinal cord through the acceleration of angiogenesis and astrogliosis.


Subject(s)
Astrocytes/physiology , Bone Marrow Cells/cytology , Hematopoietic Stem Cells/physiology , Neovascularization, Physiologic/physiology , Proteins/physiology , Spinal Cord Injuries/physiopathology , Adaptor Proteins, Signal Transducing , Animals , Astrocytes/cytology , Astrocytes/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Proteins/genetics , Spinal Cord Injuries/metabolism
14.
Circ Res ; 105(9): 860-8, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19762683

ABSTRACT

RATIONALE: Recent reports have demonstrated that signals from vascular endothelial cells are necessary for organogenesis that may precede vasculogenesis. However, the origin of these neovascular cells in regenerating tissue has not been clarified. OBJECTIVE: Here we tested the hypothesis that adult neural stem cells (NSCs) can differentiate into vascular lineage, as well as neural lineage, in the process of collaborative organogenesis. METHODS AND RESULTS: NSCs, clonally isolated from mouse brain, were shown to develop endothelial and smooth muscle phenotypes in vitro. To elucidate whether NSCs can simultaneously differentiate into vascular and neural cells in vivo, genetically labeled NSCs were administered to mice with unilateral sciatic nerve crush injury or operatively induced brain and myocardial ischemia. Two weeks later, necropsy examination disclosed recruitment of the labeled NSCs to sites of injury differentiating into vascular cells (endothelial cells and vascular smooth muscle cells) and Schwann cells in regenerating nerve. Similarly, NSC-derived vascular cells/astrocytes and endothelial cells were identified in ischemic brain tissue and capillaries in myocardium 2 weeks following transplantation, respectively. CONCLUSIONS: These findings, concurrent vasculogenesis and neurogenesis from a common stem cell, suggest that certain somatic stem cells are capable of differentiating into not only somatic cells of identity but also into vascular cells for tissue regeneration.


Subject(s)
Adult Stem Cells/metabolism , Brain Ischemia/physiopathology , Endothelial Cells/metabolism , Myocardial Infarction/physiopathology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , Neurogenesis , Neurons/metabolism , Sciatic Neuropathy/physiopathology , Adult Stem Cells/transplantation , Animals , Biomarkers/metabolism , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/surgery , Cell Communication , Cell Differentiation , Cell Lineage , Cell Movement , Disease Models, Animal , Endothelial Cells/transplantation , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocytes, Smooth Muscle/transplantation , Neurons/transplantation , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Sciatic Neuropathy/surgery , Signal Transduction , Spheroids, Cellular , Stem Cell Transplantation , Time Factors
15.
Stem Cells ; 27(11): 2857-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19711453

ABSTRACT

A number of preclinical studies have indicated the therapeutic potential of endothelial progenitor cells for vascular regeneration in ischemic diseases. A phase I/IIa clinical trial of transplantation of autologous CD34(+) cells, the endothelial and hematopoietic progenitor-enriched fraction, was performed in no-option patients with atherosclerotic peripheral artery disease or Buerger's disease with critical limb ischemia (CLI). CD34(+) cells were isolated from the G-CSF-mobilized apheresis product using a magnetic cell sorting system. CD34(+) cells (10(5)/kg, n = 6; 5 x 10(5)/kg, n = 8; or 10(6)/kg, n = 3) were injected i.m. into the leg with more severe ischemia. The Efficacy Score, representing changes in the toe brachial pressure index (TBPI), Wong-Baker FACES pain rating scale, and total walking distance 12 weeks after cell transplantation, the primary endpoint, was positive, indicating improvement in limb ischemia in all patients, although no significant dose-response relationship was observed. During the 12-week observation after cell therapy, the Wong-Baker FACES pain rating scale, TBPI, transcutaneous partial oxygen pressure, total or pain-free walking distance, and ulcer size serially improved in all patients. No death or major amputation occurred, and severe adverse events were rare, although mild to moderate events relating to G-CSF and leukapheresis were frequent during the 12-week follow-up. In conclusion, the outcomes of this prospective clinical study indicate the safety and feasibility of CD34(+) cell therapy in patients with CLI. Favorable trends in efficacy parameters encourage a randomized and controlled trial in the future.


Subject(s)
Antigens, CD34/metabolism , Cell- and Tissue-Based Therapy/methods , Granulocyte Colony-Stimulating Factor/metabolism , Ischemia/therapy , Leg/pathology , Stem Cells/cytology , Adult , Aged , Aged, 80 and over , Cell- and Tissue-Based Therapy/adverse effects , Female , Humans , Injections, Intramuscular , Male , Stem Cell Transplantation , Stem Cells/metabolism , Transplantation, Autologous , Treatment Outcome
16.
Biochem Biophys Res Commun ; 381(2): 135-8, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19032947

ABSTRACT

In the process of hematopoietic stem cell (CD133+ cell) differentiation, a drastic change in gene expression occurs which must be regulated by epigenetic mechanisms. One strategy for CD133+ cell differentiation analysis is to identify genomic DNA regions that have been modified in the process of differentiation. However, it is difficult to obtain large amounts of genomic DNA from uniform CD133+ cells. Based on this situation, we screened genomic DNA regions where modifications change during the process of differentiation in human CD133+ cells using differential methylation site scanning (DMSS), which is a method of identifying differentially methylated regions of the genome from a small number of cells. As a result, we cloned three DNA fragments which corresponded to centrosomal protein 68kDA (Cep68), TRIO and F-actin binding protein (TRIOBP), and AMP-activated protein kinase beta (AMPKb).


Subject(s)
DNA Methylation , Gene Expression , Genome, Human , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , AC133 Antigen , AMP-Activated Protein Kinases/genetics , Antigens, CD/analysis , Cloning, Molecular , Glycoproteins/analysis , Humans , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Peptides/analysis
17.
J Cell Physiol ; 215(1): 234-42, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18205179

ABSTRACT

We recently reported that systemic administration of peripheral blood (PB) CD34+ cells, an endothelial progenitor cell (EPC)-enriched population, contributed to fracture healing via vasculogenesis/angiogenesis. However, pathophysiological role of EPCs in fracture healing process has not been fully clarified. Therefore, we investigated the hypothesis whether mobilization and incorporation of bone marrow (BM)-derived EPCs may play a pivotal role in appropriate fracture healing. Serial examinations of Laser doppler perfusion imaging and histological capillary density revealed that neovascularization activity at the fracture site peaked at day 7 post-fracture, the early phase of endochondral ossifification. Fluorescence-activated cell sorting (FACS) analysis demonstrated that the frequency of BM cKit+Sca1+Lineage- (Lin-) cells and PB Sca1+Lin- cells, which are EPC-enriched fractions, significantly increased post-fracture. The Sca1+ EPC-derived vasuculogenesis at the fracture site was confirmed by double immunohistochemistry for CD31 and Sca1. BM transplantation from transgenic donors expressing LacZ transcriptionally regulated by endothelial cell-specific Tie-2 promoter into wild type also provided direct evidence that EPCs contributing to enhanced neovascularization at the fracture site were specifically derived from BM. Animal model of systemic administration of PB Sca1+Lin- Green Fluorescent Protein (GFP)+ cells further confirmed incorporation of the mobilized EPCs into the fracture site for fracture healing. These findings indicate that fracture may induce mobilization of EPCs from BM to PB and recruitment of the mobilized EPCs into fracture sites, thereby augment neovascularization during the process of bone healing. EPCs may play an essential role in fracture healing by promoting a favorable environment through neovascularization in damaged skeletal tissue.


Subject(s)
Bone Marrow Cells/pathology , Bone and Bones/pathology , Cell Movement , Endothelial Cells/pathology , Fractures, Bone/pathology , Stem Cells/pathology , Wound Healing , Animals , Ataxin-1 , Ataxins , Bone and Bones/blood supply , Fractures, Bone/chemically induced , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phenotype
18.
J Clin Exp Hematop ; 47(2): 61-72, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18040145

ABSTRACT

In order to establish the method of generating powerful gammadelta T cells for anti-tumor immunotherapy, we investigated the effects of monocyte-derived dendritic cells (mo-DCs) on anti-tumor cytotoxicity of expanded gammadelta T cells. Activation of gammadelta T cells co-cultured for 2-3 days with immature or mature mo-DCs was evaluated by CD69 expression and anti-tumor cytotoxicity using two assays : the 5- (and 6-) carboxyfluorescein diacetate, succinimidyl ester-based cytotoxicity assay and the calcein-AM-based Terascan assay. gammadelta T cells were used as effector cells and myeloma cell line (RPMI8226) or chronic myelogenous leukemia blastic crisis cell line (C2F8) were used as target cells. CD69 expression on gammadelta T cells was enhanced by co-culture with both immature and mature mo-DCs in a cell-number-dependent fashion. CD69 expression was enhanced after addition of mo-DCs of either autologous or allogeneic origin. Activation of gammadelta T cells with mo-DCs enhanced anti-tumor cytotoxicity of gammadelta T cells against RPMI8226 and C2F8 in an effector-to-target ratio-dependent manner. Activation of gammadelta T cells by mo-DCs was associated with the enhancement of anti-tumor cytotoxicity of gammadelta T cells. Potent gammadelta T cells activated by mo-DCs were considered to be applicable to an efficient gammadelta T cell-mediated immunotherapy for tumors.


Subject(s)
Dendritic Cells/metabolism , Immunotherapy, Adoptive/methods , Lymphocyte Activation/physiology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , Humans , Lectins, C-Type , Monocytes/cytology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes, Cytotoxic/immunology
19.
Int Heart J ; 48(3): 347-58, 2007 May.
Article in English | MEDLINE | ID: mdl-17592199

ABSTRACT

The electrophysiological effects of carvedilol, a beta-blocking agent with vasodilating actions, have been studied on rabbit pacemaker cells using the whole-cell patch clamp technique. Nystatin-perforated patch recordings from the sinoatrial (SA) and atrioventricular (AV) nodes demonstrated that 1-3 microM of carvedilol caused a decrease in the spontaneous firing frequency, depolarization of the maximal diastolic potential, and prolongation of the action potential duration in both species. Voltage clamp experiments were performed using SA and AV node myocytes to identify and define the carvedilol-induced changes in the Ca(2+) current, I(Ca), delayed rectifier K(+) current, I(K), and hyperpolarization-activated inward current, I(f). In the SA node cells, 1 microM of carvedilol blocked I(K), I(Ca), and I(f) by 72%, 47%, and 22%, respectively. In the AV node cells, the corresponding reductions were 64% (I(K)) and 46% (I(Ca)), respectively. In both the SA and AV nodes the decrease in I(K) appeared to be mainly due to the rapidly activating component of the delayed rectifier, I(Kr), since the high dose of carvedilol blocked I(K) in the SA and AV nodes to a submaximal degree. In conclusion, effective doses of carvedilol have classical class III antiarrhythmic actions and a negative chronotropic effect resulting from the inhibition of I(K) and I(Ca). Both actions may be efficacious for treating supraventricular tachyarrhythmias.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Atrioventricular Node/drug effects , Carbazoles/pharmacology , Cardiac Pacing, Artificial/methods , Propanolamines/pharmacology , Sinoatrial Node/drug effects , Action Potentials/drug effects , Animals , Atrioventricular Node/pathology , Atrioventricular Node/physiopathology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Carvedilol , Disease Models, Animal , Patch-Clamp Techniques/methods , Potassium Channels/drug effects , Potassium Channels/metabolism , Rabbits , Sinoatrial Node/metabolism , Sinoatrial Node/physiopathology , Tachycardia, Supraventricular/drug therapy , Tachycardia, Supraventricular/metabolism , Tachycardia, Supraventricular/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...