Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Exp Cell Res ; 427(2): 113603, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37075826

ABSTRACT

Hematopoietic toxicity due to ionizing radiation (IR) is a leading cause of death in nuclear incidents, occupational hazards, and cancer therapy. Oxymatrine (OM), an extract originating from the root of Sophora flavescens (Kushen), possesses extensive pharmacological properties. In this study, we demonstrate that OM treatment accelerates hematological recovery and increases the survival rate of mice subjected to irradiation. This outcome is accompanied by an increase in functional hematopoietic stem cells (HSCs), resulting in enhanced hematopoietic reconstitution abilities. Mechanistically, we observed significant activation of the MAPK signaling pathway, accelerated cellular proliferation, and decreased cell apoptosis. Notably, we identified marked increases in the cell cycle transcriptional regulator Cyclin D1 (Ccnd1) and the anti-apoptotic protein BCL2 in HSCs after OM treatment. Further investigation revealed that the expression of Ccnd1 transcript and BCL2 levels were reversed upon specific inhibition of ERK1/2 phosphorylation, effectively negating the rescuing effect of OM. Moreover, we determined that targeted inhibition of ERK1/2 activation significantly counteracted the regenerative effect of OM on human HSCs. Taken together, our results suggest a crucial role for OM in hematopoietic reconstitution following IR via MAPK signaling pathway-mediated mechanisms, providing theoretical support for innovative therapeutic applications of OM in addressing IR-induced injuries in humans.


Subject(s)
Alkaloids , Mice , Humans , Animals , Phosphorylation , Alkaloids/pharmacology , Signal Transduction , Apoptosis , Proto-Oncogene Proteins c-bcl-2/genetics
2.
Appl Microbiol Biotechnol ; 108(1): 31, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38175233

ABSTRACT

A complete catalase-encoding gene, designated soiCat1, was obtained from soil samples via metagenomic sequencing, assembly, and gene prediction. soiCat1 showed 73% identity to a catalase-encoding gene of Mucilaginibacter rubeus strain P1, and the amino acid sequence of soiCAT1 showed 99% similarity to the catalase of a psychrophilic bacterium, Pedobacter cryoconitis. soiCAT1 was identified as a psychrophilic enzyme due to the low optimum temperature predicted by the deep learning model Preoptem, which was subsequently validated through analysis of enzymatic properties. Experimental results showed that soiCAT1 has a very narrow range of optimum temperature, with maximal specific activity occurring at the lowest test temperature (4 °C) and decreasing with increasing reaction temperature from 4 to 50 °C. To rationally design soiCAT1 with an improved temperature range, soiCAT1 was engineered through site-directed mutagenesis based on molecular evolution data analyzed through position-specific amino acid possibility calculation. Compared with the wild type, one mutant, soiCAT1S205K, exhibited an extended range of optimum temperature ranging from 4 to 20 °C. The strategies used in this study may shed light on the mining of genes of interest and rational design of desirable proteins. KEY POINTS: • Numerous putative catalases were mined from soil samples via metagenomics. • A complete sequence encoding a psychrophilic catalase was obtained. • A mutant psychrophilic catalase with an extended range of optimum temperature was engineered through site-directed mutagenesis.


Subject(s)
Deep Learning , Catalase/genetics , Amino Acid Sequence , Amino Acids , Soil
3.
Exp Cell Res ; 409(2): 112934, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34801561

ABSTRACT

Hematopoietic stem cells (HSCs) are sensitive to ionizing radiation (IR) damage, and its injury is the primary cause of bone marrow (BM) hematopoietic failure and even death after exposure to a certain dose of IR. However, the underlying mechanisms remain incompletely understood. Here we show that mitochondrial oxidative damage, which is characterized by mitochondrial reactive oxygen species overproduction, mitochondrial membrane potential reduction and mitochondrial permeability transition pore opening, is rapidly induced in both human and mouse HSCs and directly accelerates HSC apoptosis after IR exposure. Mechanistically, 5-lipoxygenase (5-LOX) is induced by IR exposure and contributes to IR-induced mitochondrial oxidative damage through inducing lipid peroxidation. Intriguingly, a natural antioxidant, caffeic acid (CA), can attenuate IR-induced HSC apoptosis through suppressing 5-LOX-mediated mitochondrial oxidative damage, thus protecting against BM hematopoietic failure after IR exposure. These findings uncover a critical role for mitochondria in IR-induced HSC injury and highlight the therapeutic potential of CA in BM hematopoietic failure induced by IR.


Subject(s)
Antioxidants/pharmacology , Arachidonate 5-Lipoxygenase/chemistry , Caffeic Acids/pharmacology , Cobalt Radioisotopes/toxicity , Hematopoietic Stem Cells/drug effects , Mitochondria/drug effects , Oxidative Stress , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , DNA Damage , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/radiation effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/radiation effects
4.
Haematologica ; 106(2): 412-423, 2021 02 01.
Article in English | MEDLINE | ID: mdl-31974197

ABSTRACT

Long-term hematopoietic output is dependent on hematopoietic stem cell (HSC) homeostasis which is maintained by a complex molecular network. Among these, microRNAs play crucial roles, while the underlying molecular basis has not been fully elucidated. Here, we show that miR-21 is enriched in murine HSCs, and mice with conditional knockout of miR-21 exhibit an obvious perturbation in normal hematopoiesis. Moreover, significant loss of HSC quiescence and long-term reconstituting ability are observed in the absence of miR-21. Further studies reveal that miR-21 deficiency markedly decreases the NF-κB pathway, accompanied by increased expression of PDCD4, a direct target of miR-21, in HSCs. Interestingly, overexpression of PDCD4 in wild-type HSCs generates similar phenotypes as those of miR-21-deficient HSCs. More importantly, knockdown of PDCD4 can significantly rescue the attenuation of NF-κB activity, thereby improving the defects in miR-21-null HSCs. On the other hand, we find that miR-21 is capable of preventing HSCs from ionizing radiation-induced DNA damage via activation of the NF-κB pathway. Collectively, our data demonstrate that miR-21 is involved in maintaining HSC homeostasis and function, at least in part, by regulating the PDCD4-mediated NF-κB pathway and provide a new insight into the radioprotection of HSCs.


Subject(s)
MicroRNAs , NF-kappa B , Animals , Hematopoietic Stem Cells/metabolism , Homeostasis , Mice , Mice, Knockout , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction
5.
Blood ; 132(2): 210-222, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29802222

ABSTRACT

It is known that insulin-like growth factor-1 (IGF-1) also functions as a hematopoietic factor, although its direct effect on thrombopoiesis remains unclear. In this study, we show that IGF-1 is able to promote CD34+ cell differentiation toward megakaryocytes (MKs), as well as the facilitation of proplatelet formation (PPF) and platelet production from cultured MKs. The in vivo study demonstrates that IGF-1 administration accelerates platelet recovery in mice after 6.0 Gy of irradiation and in mice that received bone marrow transplantation following 10.0 Gy of lethal irradiation. Subsequent investigations reveal that extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt activation mediate the effect of IGF-1 on thrombopoiesis. Notably, Akt activation induced by IGF-1 is more apparent than that of ERK1/2, compared with that of thrombopoietin (TPO) treatment. Moreover, the effect of IGF-1 on thrombopoiesis is independent of TPO signaling because IGF-1 treatment can also lead to a significant increase of platelet counts in homozygous TPO receptor mutant mice. Further analysis indicates that the activation of Akt triggered by IGF-1 requires the assistance of steroid receptor coactivator-3 (SRC-3). Therefore, our data reveal a distinct role of IGF-1 in regulating thrombopoiesis, providing new insights into TPO-independent regulation of platelet generation.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thrombopoiesis , Animals , Biomarkers , Blood Platelets/metabolism , Cell Differentiation , Cells, Cultured , Cytoskeleton , Gene Knockdown Techniques , Humans , Insulin-Like Growth Factor I/genetics , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Knockout , Models, Biological , Platelet Activation , Platelet Count , Ploidies , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Thrombopoiesis/genetics , Thrombopoietin/metabolism
6.
Blood ; 132(9): 911-923, 2018 08 30.
Article in English | MEDLINE | ID: mdl-29959189

ABSTRACT

Quiescence maintenance is an important property of hematopoietic stem cells (HSCs), whereas the regulatory factors and underlying mechanisms involved in HSC quiescence maintenance are not fully uncovered. Here, we show that steroid receptor coactivator 3 (SRC-3) is highly expressed in HSCs, and SRC-3-deficient HSCs are less quiescent and more proliferative, resulting in increased sensitivity to chemotherapy and irradiation. Moreover, the long-term reconstituting ability of HSCs is markedly impaired in the absence of SRC-3, and SRC-3 knockout (SRC-3-/-) mice exhibit a significant disruption of hematopoietic stem and progenitor cell homeostasis. Further investigations show that SRC-3 deficiency leads to enhanced mitochondrial metabolism, accompanied by overproduction of reactive oxygen species (ROS) in HSCs. Notably, the downstream target genes of peroxisome proliferator-activated receptor-coactivators 1α (PGC-1α) involved in the regulation of mitochondrial metabolism are significantly upregulated in SRC-3-deficient HSCs. Meanwhile, a significant decrease in the expression of histone acetyltransferase GCN5 accompanied by downregulation of PGC-1α acetylation is observed in SRC-3-null HSCs. Conversely, overexpression of GCN5 can inhibit SRC-3 deficiency-induced mitochondrial metabolism enhancement and ROS overproduction, thereby evidently rescuing the impairment of HSCs in SRC-3-/- mice. Collectively, our findings demonstrate that SRC-3 plays an important role in HSC quiescence maintenance by regulating mitochondrial metabolism.


Subject(s)
Hematopoietic Stem Cells/metabolism , Homeostasis/physiology , Mitochondria/metabolism , Nuclear Receptor Coactivator 3/metabolism , Reactive Oxygen Species/metabolism , Animals , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , Mitochondria/genetics , Nuclear Receptor Coactivator 3/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
7.
FASEB J ; 33(7): 8138-8147, 2019 07.
Article in English | MEDLINE | ID: mdl-30922079

ABSTRACT

Hematopoietic stem cells (HSCs) establish the entire hematopoietic system and maintain lifelong hematopoiesis. Previous studies have reported the significance of microRNAs (miRNAs) in the regulation of self-renewal and differentiation of HSCs. In this study, we show that the expression of miRNA 34a (miR-34a) is markedly up-regulated in HSCs from mice subjected to ionizing radiation (IR). Reduced numbers and DNA damage repair, as well as increased apoptosis, are observed in HSCs from miR-34a-deficient mice induced by irradiation, although miR-34a is dispensable for steady-state hematopoiesis. Further investigations show that HSCs deficient in miR-34a exhibit decreased expressions of DNA repair-associated genes involved in homologous recombination and nonhomologous end joining. Competitive transplantation confirms that loss of miR-34a leads to more severe impairment of the long-term hematopoietic function of HSCs after irradiation exposure. Consistently, treating mice with an miR-34a agomir can significantly alleviate irradiation-induced DNA damage in HSCs. Our findings demonstrate that miR-34a contributes to promoting HSCs' survival after irradiation, which provides a promising approach for protecting HSCs from IR.-Zeng, H., Hu, M., Lu, Y., Zhang, Z., Xu, Y., Wang, S., Chen, M., Shen, M., Wang, C., Chen, F., Du, C., Tang, Y., Su,Y., Chen, S., Wang, J. MicroRNA 34a promotes ionizing radiation-induced DNA damage repair in murine hematopoietic stem cells.


Subject(s)
DNA Damage , DNA Repair , Gamma Rays/adverse effects , Hematopoietic Stem Cells/metabolism , MicroRNAs/biosynthesis , Animals , Hematopoietic Stem Cells/pathology , Mice , Mice, Knockout , MicroRNAs/genetics
8.
J Transl Med ; 17(1): 161, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31101050

ABSTRACT

BACKGROUND: The roles played by cholesterol in cancer development and progression represent a popular field in the cancer community. High cholesterol levels are positively correlated with the risk of various types of cancer. APOA-I binding protein (AIBP) promotes the reverse cholesterol transport pathway (RCT) in cooperation with Apolipoprotein A-I (APOA-I) or high-density lipoprotein cholesterol. However, the combined effect of AIBP and APOA-I on intestinal tumor cells is still unclear. METHODS: Immunohistochemistry, western blot and qPCR were performed to investigate the expression of AIBP and APOA-I in intestinal tumor tissues and cell lines. The anti-tumor activity of AIBP and APOA-I was evaluated by overexpression or recombinant protein treatment. Cholesterol efflux and localization of lipid raft-related proteins were analyzed by a cholesterol efflux assay and lipid raft fraction assay, respectively. RESULTS: Here, we reported that both AIBP expression and APOA-I expression were associated with the degree of malignancy in intestinal tumors. Co-overexpression of AIBP and APOA-I more potently inhibited colon cancer cell-mediated tumor growth and metastasis compared to overexpression of each protein individually. Additionally, the recombinant fusion proteins of AIBP and APOA-I exhibited a significant therapeutic effect on tumor growth in Apcmin/+ mice as an inherited intestinal tumor model. The synergistic effect of the two proteins inhibited colon cancer cell migration, invasion and tumor-induced angiogenesis by promoting cholesterol efflux, reducing the membrane raft content, and eventually disrupting the proper localization of migration- and invasion-related proteins on the membrane raft. Moreover, cyclosporine A, a cholesterol efflux inhibitor, rescued the inhibitory effect induced by the combination of AIBP and APOA-I. CONCLUSIONS: These results indicate that the combination of APOA-I and AIBP has an obvious anticancer effect on colorectal cancer by promoting cholesterol efflux.


Subject(s)
Apolipoprotein A-I/metabolism , Cholesterol/metabolism , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Phosphoproteins/metabolism , Racemases and Epimerases/metabolism , Animals , Biological Transport , Cell Line , Cell Movement , Cell Proliferation , Humans , Mice, Inbred C57BL , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
9.
Blood ; 129(19): 2667-2679, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28264799

ABSTRACT

Thrombosis is a common complication of chronic kidney disease (CKD), but the causes and mechanisms of CKD-associated thrombosis are not well clarified. Here, we show that platelet activity is remarkably enhanced in CKD mice, with increase of serum indoxyl sulfate (IS), a typical uremic toxin, which cannot be effectively cleared by routine dialysis. Ex vivo and in vitro experiments reveal that IS displays a distinct ability to enhance platelet activities, including elevated response to collagen and thrombin, increases in platelet-derived microparticles, and platelet-monocyte aggregates. The flow chamber assay and carotid artery thrombosis model demonstrate that IS-induced platelet hyperactivity contributes to thrombus formation. Further investigations disclose that reactive oxygen species (ROS)-mediated p38MAPK signaling plays a key role in IS-induced platelet hyperactivity. Moreover, we show that Klotho, which is expressed dominantly in the kidneys, has the capacity to counteract IS-induced platelet hyperactivity by inhibiting ROS/p38MAPK signaling, whereas Klotho reduction may aggravate the effect of IS on platelet activation in CKD and klotho+/- mice. Finally, we demonstrate that Klotho protein treatment can protect against IS-induced thrombosis and atherosclerosis in apoE-/- mice. Our findings uncover the mechanism of platelet hyperactivity induced by IS and provide new insights into the pathogenesis and treatment of CKD-associated thrombosis.


Subject(s)
Blood Platelets/drug effects , Indican/adverse effects , Platelet Activation/drug effects , Renal Insufficiency, Chronic/chemically induced , Thrombosis/chemically induced , Animals , Blood Platelets/pathology , Glucuronidase/administration & dosage , Glucuronidase/metabolism , Glucuronidase/therapeutic use , Klotho Proteins , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/metabolism , Thrombosis/drug therapy , Thrombosis/metabolism
10.
Exp Cell Res ; 370(2): 409-416, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30146064

ABSTRACT

Ionizing radiation (IR) triggers the generation of reactive oxygen species (ROS), which shows potential roles in damaging the DNA and proteins at the nucleus, and eventually results in apoptosis and even cell death. Antioxidant agents can inhibit the generation of ROS after IR exposure. Tannic acid (TA), has an antioxidant activity involving in preventing cardiovascular and cerebrovascular diseases. However, little is known about the effects of TA on irradiation-induced apoptosis in megakaryocytes. Here, we evaluated the anti-radiation activity of TA in megakaryocytes. Our results showed that TA protected megakaryocytes from apoptosis induced by IR, attenuated IR-induced increases in the production of ROS, and inhibited the changes of mitochondrial membrane potential (MMP). Moreover, TA down-regulated NAPDH oxidase 1 (Nox1) expression, and decreased the phosphorylated levels of JNK and p38. Furthermore, JNK inhibitor could reduce apoptosis induced by X-irradiation in M07e cells. In vivo experiments confirmed that TA could promote the platelet recovery, reduce the percentage of apoptosis CD41+ megakaryocytes in bone marrow and raise survival during 30 days in mice by total body irradiation. In conclusion, TA can protecte the megakaryocytes from apoptosis caused by IR through inhibiting Nox1 expression to reduce ROS generation and repressing JNK/p38 MAPK pathway activation.


Subject(s)
Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Megakaryocytes/drug effects , Tannins/pharmacology , Cell Line, Tumor , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Megakaryocytes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Article in English | MEDLINE | ID: mdl-29158275

ABSTRACT

The increasing incidence of multidrug-resistant Acinetobacter baumannii (MDRAb) infections worldwide has necessitated the development of novel antibiotics. Human defensin 5 (HD5) is an endogenous peptide with a complex architecture and antibacterial activity against MDRAb In the present study, we attempted to simplify the structure of HD5 by removing disulfide bonds. We found that the Cys2-4 bond was most indispensable for HD5 to inactivate MDRAb, although the antibacterial activity of the derivative was significantly attenuated. We then replaced the noncationic and nonhydrophobic residues with electropositive Arg to increase the antibacterial activity of HD5 derivative that contains a Cys2-4 bond, obtaining another derivative termed HD5d5. The in vitro antibacterial assay and irradiation-wound-infection animal experiment both showed that HD5d5 was much more effective than HD5 at eliminating MDRAb Further investigations revealed that HD5d5 efficiently bound to outer membrane lipid A and penetrated membranes, leading to bacterial collapse and peptide translocation. Compared to HD5, more HD5d5 molecules were located in the cytoplasm of MDRAb, and HD5d5 was more efficient at reducing the activities of superoxide dismutase and catalase, causing the accumulation of reactive oxygen species that are detrimental to microbes. In addition, HD5 failed to suppress the pathogenic outer membrane protein A of Acinetobacter baumannii (AbOmpA) at concentrations up to 50 µg/ml, whereas HD5d5 strongly bound to AbOmpA and exhibited a dramatic toxin-neutralizing ability, thus expanding the repertoire of drugs that is available to treat MDRAb infections.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Gene Expression Regulation, Bacterial , Wound Infection/drug therapy , alpha-Defensins/pharmacology , Acinetobacter Infections/microbiology , Acinetobacter Infections/mortality , Acinetobacter Infections/pathology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/growth & development , Acinetobacter baumannii/metabolism , Animals , Anti-Bacterial Agents/chemical synthesis , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalase/antagonists & inhibitors , Catalase/genetics , Catalase/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Lipid A/metabolism , Mice , Mice, Inbred BALB C , Protein Binding , Protein Engineering/methods , Protein Isoforms/chemical synthesis , Protein Isoforms/pharmacology , Protein Transport , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Survival Analysis , Whole-Body Irradiation , Wound Infection/microbiology , Wound Infection/mortality , Wound Infection/pathology , alpha-Defensins/chemical synthesis
12.
J Gene Med ; 20(6): e3022, 2018 06.
Article in English | MEDLINE | ID: mdl-29656516

ABSTRACT

BACKGROUND: The cellular and molecular mechanisms responsible for the age-associated delay of cutaneous wound healing are still not well understood. Previous studies have shown that miR-21 plays key roles during skin wound healing. We presumed that dysregulation of miR-21 may be involved in age-associated defects in wound healing and that miR-21 may be one potential therapeutic target by which to ameliorate wound defects in elderly subjects. METHODS: Circular full thickness excisional wounds were made on the dorsal skin of young (2-month-old) and aged (12-month-old) female mice. The wound healing rates were quantified and compared between wild-type and miR-21 knock-in mice. Both histologic and morphometric analyses of the wounds were evaluated. Furthermore, the expression patterns of miR-21 during wound healing in both young and aged mice were assessed by in situ hybridization. The effects of topical miR-21 overexpression on wound healing in aged mice were estimated by both wound closure quantification and histological analyses. RESULTS: Aged miR-21 knock-in female mice showed significantly improved wound healing compared to their wild-type counterparts with respect to mature granulation tissue, smaller wound width and thinner epidermis. The expression patterns of miR-21 showed that miR-21 levels were insufficient for repairing granulation tissue in aged mice. Intradermal injection of miR-21 plasmid around wounds could upregulate miR-21 levels during wound healing and ameliorate age-associated skin wound defects. CONCLUSIONS: The results of the present study reveal that the upregulation of miR-21 levels could improve wound repair in aged mice, which suggests that a therapeutic strategy targeting miR-21 expression in age-associated wound healing may be feasible.


Subject(s)
Aging/physiology , MicroRNAs/metabolism , Skin/injuries , Wound Healing/physiology , Animals , Female , Gene Expression Regulation , Gene Knock-In Techniques , Mice, Inbred C57BL , Mice, Mutant Strains , MicroRNAs/genetics , Skin/pathology
13.
Biochem Biophys Res Commun ; 506(4): 1052-1058, 2018 12 02.
Article in English | MEDLINE | ID: mdl-30409423

ABSTRACT

As compared with 2D cell line cultures, 3D intestinal organoids are better at maximally recapitulating the physiological features of stem cells in vivo. However, the complex 3D structure is an obstacle which must be objectively and automatically evaluated to assess colony growth and regeneration. Meanwhile, no internal standard currently exists for evaluating the size of heterogeneities in organoids or defining those regenerating colonies. Herein, we developed a simple morphometry system to image MTT-stained organoids. The growth curve of organoids can be automatically generated based upon analyzing the integrated optical density using software. Referencing the definition standards of in vivo regenerating crypts, the perimeters of crypts cultured 24 h after seeding were selected as an "Organoid Unit" to further evaluate colony survival rate and colony size heterogeneities after exposure to varying doses of irradiation. Moreover, the morphometry-based quantification data collected confirmed other findings associated with radiation sensitizing effects of ataxia telangiectasia mutated (ATM) and ataxia telangiectasia and Rad3-related protein (ATR) inhibitor and the radiation protective effect of IL-22. In summary, the novel organoid morphometry system combined with a new internal reference is a practical means for standardizing assessment of growth, survival and regeneration of intestinal organoid colonies. This method has promise to facilitate drug screens in intestinal and other organoid systems.


Subject(s)
Imaging, Three-Dimensional , Intestines/growth & development , Organoids/growth & development , Regeneration , Animals , Automation , Cell Survival/drug effects , Intestines/drug effects , Mice , Organ Size , Organoids/drug effects , Radiation-Protective Agents/pharmacology , Regeneration/drug effects , Stem Cells/cytology , Stem Cells/drug effects
14.
Blood ; 127(8): 1024-35, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26644453

ABSTRACT

The effect of sympathetic stimulation on thrombopoiesis is not well understood. Here, we demonstrate that both continual noise and exhaustive exercise elevate peripheral platelet levels in normal and splenectomized mice, but not in dopamine ß-hydroxylase-deficient (Dbh(-/-)) mice that lack norepinephrine (NE) and epinephrine (EPI). Further investigation demonstrates that sympathetic stimulation via NE or EPI injection markedly promotes platelet recovery in mice with thrombocytopenia induced by 6.0 Gy of total-body irradiation and in mice that received bone marrow transplants after 10.0 Gy of lethal irradiation. Unfavorably, sympathetic stress-stimulated thrombopoiesis may also contribute to the pathogenesis of atherosclerosis by increasing both the amount and activity of platelets in apolipoprotein E-deficient (ApoE(-/-)) mice. In vitro studies reveal that both NE and EPI promote megakaryocyte adhesion, migration, and proplatelet formation (PPF) in addition to the expansion of CD34(+) cells, thereby facilitating platelet production. It is found that α2-adrenoceptor-mediated extracellular signal-regulated kinase 1/2 (ERK1/2) activation is involved in NE- and EPI-induced megakaryocyte adhesion and migration, and PPF is regulated by ERK1/2 activation-mediated RhoA GTPase signaling. Our data deeply characterize the role of sympathetic stimulation in the regulation of thrombopoiesis and reevaluate its physiopathological implications.


Subject(s)
Blood Platelets/cytology , Cell Movement , Megakaryocytes/cytology , Thrombopoiesis , Animals , Blotting, Western , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/drug effects , Cell Movement/physiology , Disease Models, Animal , Epinephrine/metabolism , Epinephrine/pharmacology , MAP Kinase Signaling System/physiology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Norepinephrine/metabolism , Norepinephrine/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Stress, Physiological/physiology , Sympathetic Nervous System/physiology
15.
Platelets ; 29(7): 702-708, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29119850

ABSTRACT

Dopamine (DA), a catecholamine neurotransmitter, is known to for its diverse roles on hematopoiesis, yet its function in thrombopoiesis remains poorly understood. This study shows that DA stimulation can directly induce platelet production from megakaryocytes (MKs) in the final stages of thrombopoiesis via a reactive oxygen species (ROS)-dependent pathway. The mechanism was suggested by the results that DA treatment could significantly elevate the ROS levels in MKs, and time-dependently activate oxidative stress-mediated signaling, including p38 mitogen-activated protein kinase, c-Jun NH2-terminal kinase, and caspase-3 signaling pathways, while the antioxidants N-acetylcysteine and L-glutathione could effectively inhibit the activation of these signaling pathways, as well as the ROS increase and platelet production triggered by DA. Therefore, our data revealed that the direct role and mechanism of DA in thrombopoiesis, which provides new insights into the function recognition of DA in hematopoiesis.


Subject(s)
Blood Platelets/metabolism , Dopamine/metabolism , Megakaryocytes/metabolism , Oxidative Stress , Signal Transduction , Thrombopoiesis , Animals , Apoptosis , Caspase 3/metabolism , Dopamine/pharmacology , Flow Cytometry , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Megakaryocytes/cytology , Mice , Reactive Oxygen Species/metabolism , Thrombopoiesis/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Biochim Biophys Acta ; 1862(8): 1443-52, 2016 08.
Article in English | MEDLINE | ID: mdl-27169579

ABSTRACT

Keratinocyte migration is essential for re-epithelialization during skin wound healing, but the molecular mechanisms regulating this cellular response remain to be completely clarified. Here we show that keratinocyte-specific miR-205 is significantly downregulated in the leading edge of the migrating epithelial tongue after skin injury in mice. In HaCaT keratinocytes, miR-205 could be downregulated by TGF-ß1 stimulation. And similar to the effect of TGF-ß1, miR-205 knockdown could promote keratinocyte migration in wound scratch model in vitro. Furthermore, topical inhibition of miR-205 by administrating Pluronic gel containing antagomir-205 could accelerate re-epithelialization in mouse skin wound model in vivo. Moreover, we identified integrin alpha 5 (ITGA5) as one key functional miR-205 target in the re-epithelialization process and epidermal downregulation of miR-205 may desilence ITGA5 to promote keratinocyte migration. And knockdown of ITGA5 would abolish the pro-migratory effects of miR-205 inhibition in vitro. What's more, we found dysregulation of miR-205 and its target ITGA5 in epidermis of clinical chronic wound samples with persistence of high level miR-205 and absence of ITGA5. Our findings indicate that downregulation of miR-205 in the leading migrating keratinocytes is critical for re-epithelialization and miR-205 may be a potential therapeutic target for chronic wounds.


Subject(s)
Integrin alpha5/metabolism , Keratinocytes/metabolism , MicroRNAs/metabolism , Mouth Mucosa/injuries , Tongue/injuries , Wound Healing , Animals , Cell Movement , Disease Models, Animal , Humans , Integrin alpha5/genetics , Keratinocytes/pathology , Male , Mice , MicroRNAs/genetics , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Tongue/metabolism , Tongue/pathology
17.
Blood ; 123(14): 2250-60, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24553176

ABSTRACT

Human growth hormone (hGH) is known to play a functional role in regulating hematopoiesis, although its direct effect on thrombopoiesis is unclear. In this study, we show for the first time that hGH has a distinct capacity to promote the differentiation of human primary megakaryocytes derived from umbilical cord blood CD34(+) cells. In particular, hGH is potent in facilitating proplatelet formation and platelet production from cultured megakaryocytes. The stage- and time-specific activations of extracellular signal-regulated kinase 1/2 and protein kinase B signaling pathways are involved in the action of hGH. Fusion with hGH enhances the effect of a tandem dimer of thrombopoietin mimetic peptide (dTMP) on thrombopoiesis, manifested by a significant acceleration and increase of platelet production, indicating that hGH may exert a complementary and synergistic effect with c-Mpl ligands on thrombopoiesis. Accordingly, the administration of dTMP-growth hormone fusion protein led to a rapid platelet recovery in mice with severe thrombocytopenia induced by 6.5 Gy total body irradiation, thereby markedly abridging the duration of thrombocytopenia crisis (platelets <150 × 10(9)/L), in comparison with high doses of dTMP. These findings demonstrate the functional role of growth hormone in promoting thrombopoiesis and provide a promising avenue for the treatment of severe thrombocytopenia.


Subject(s)
Human Growth Hormone/pharmacology , Megakaryocytes/drug effects , Peptides/pharmacology , Receptors, Thrombopoietin/agonists , Thrombopoiesis/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Drug Synergism , Humans , Ligands , Male , Megakaryocytes/physiology , Mice , Mice, Inbred BALB C , Peptides/chemistry
18.
Int J Mol Sci ; 17(1)2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26703592

ABSTRACT

Programmed cell death 4 (PDCD4) is one multi-functional tumor suppressor inhibiting neoplastic transformation and tumor invasion. The role of PDCD4 in tumorigenesis has attracted more attention and has been systematically elucidated in cutaneous tumors. However, the normal biological function of PDCD4 in skin is still unclear. In this study, for the first time, we find that tumor suppressor PDCD4 is uniquely induced in a cell density-dependent manner in keratinocytes. To determine the potential role of PDCD4 in keratinocyte cell biology, we show that knockdown of PDCD4 by siRNAs can promote cell proliferation in lower cell density and partially impair contact inhibition in confluent HaCaT cells, indicating that PDCD4 serves as an important regulator of keratinocytes proliferation and contact inhibition in vitro. Further, knockdown of PDCD4 can induce upregulation of cyclin D1, one key regulator of the cell cycle. Furthermore, the expression patterns of PDCD4 in normal skin, different hair cycles and the process of wound healing are described in detail in vivo, which suggest a steady-state regulatory role of PDCD4 in epidermal homeostasis and wound healing. These findings provide a novel molecular mechanism for keratinocytes' biology and indicate that PDCD4 plays a role in epidermal homeostasis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Keratinocytes/metabolism , RNA-Binding Proteins/metabolism , Up-Regulation , Wound Healing , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Hair Follicle/cytology , Hair Follicle/metabolism , Hair Follicle/physiology , HeLa Cells , Homeostasis , Humans , Keratinocytes/physiology , Male , Mice , Mice, Inbred BALB C , RNA-Binding Proteins/genetics
19.
J Virol ; 87(5): 2835-45, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23269800

ABSTRACT

Herpes simplex virus 2 (HSV-2) infection is still one of the common causes of sexually transmitted diseases worldwide. The prevalence of HSV strains resistant to traditional nucleoside antiviral agents has led to the development of novel antiviral drugs. Human alpha-defensin 5 (HD5), a kind of endogenous antimicrobial peptide expressed in the epithelia of the small intestine and urogenital tract, displays natural antiviral activity. Based on arginine-rich features and adaptive evolution characteristics of vertebrate defensins, we conducted a screen for HD5 derivatives with enhanced anti-HSV-2 activity by a single arginine substitution at the adaptive evolution sites. Cell protection assay and temporal antiviral studies showed that HD5 and its mutants displayed affirmatory but differential anti-HSV-2 effects in vitro by inhibiting viral adhesion and entry. Inspiringly, the E21R-HD5 mutant had significantly higher antiviral activity than natural HD5, which is possibly attributed to the stronger binding affinity of the E21R-HD5 mutant with HSV-2 capsid protein gD, indicating that E21R mutation can increase the anti-HSV-2 potency of HD5. In a mouse model of lethal HSV-2 infection, prophylactic and/or therapeutic treatment with E21R-HD5 via intravaginal instillation remarkably alleviated the symptoms and delayed disease progress and resulted in about a 1.5-fold-higher survival rate than in the HD5 group. Furthermore, the E21R variant exhibited a 2-fold-higher antiviral potency against HIV-1 over parental HD5 in vitro. This study demonstrates that arginine mutagenesis at appropriate evolution sites may significantly enhance the antiviral activity of HD5, which also paves a facile way to search for potent antiviral drugs based on natural antimicrobial peptides.


Subject(s)
Antiviral Agents/pharmacology , HIV Infections/drug therapy , Herpes Simplex/drug therapy , Herpesvirus 2, Human/drug effects , Virus Attachment/drug effects , alpha-Defensins , Amino Acid Sequence , Animals , Anti-HIV Agents/pharmacology , Capsid Proteins/drug effects , Capsid Proteins/metabolism , Chlorocebus aethiops , Evolution, Molecular , Female , HIV Infections/prevention & control , HIV-1/drug effects , Herpes Simplex/prevention & control , Herpesvirus 2, Human/physiology , Humans , Mice , Mice, Inbred BALB C , Mutagenesis , Mutation , Sequence Alignment , Vero Cells , Viral Load , alpha-Defensins/chemistry , alpha-Defensins/genetics , alpha-Defensins/metabolism , alpha-Defensins/pharmacology
20.
Arch Biochem Biophys ; 542: 21-7, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24315958

ABSTRACT

AIM: Cells respond differently to DNA damaging agents, which may related to cell context and differentiation status. The aim of present study was to observe the cellular and molecular responses of cells in different differentiation status to ionizing irradiation (IR). METHODS: Crypt-villus unit of murine small intestine was adopted as a cell differentiation model. DNA damage responses (DDRs) of crypt and villus were observed 1-24 h after 12 Gy IR using gene expression microarray analysis, immunohistochemical staining, Western blotting and Electrophoretic Mobility Shift Assay. RESULTS: Microarray analysis revealed that most differentially expressed genes were related to p53 signaling pathway in crypt 4h after IR and in both crypt and villus 24h after IR. In crypt stem cells/progenitor cells, H2AX was phosphorylated and dephosphorylated quickly, Ki67 attenuated, cell apoptosis enhanced, phosphorylated P53 increased and translocated into nuclear with the ability to bind p53-specific sequence. In upper crypt (transit amplifying cells) and crypt-villus junction, cells kept survive and proliferate as indicated by retained Ki67 expression, suppressed p53 activation, and rare apoptosis. CONCLUSIONS: DDRs varied with cell differentiation status and cell function in small intestinal epithelium. P53 signaling pathway could be an important regulatory mechanism of DDRs.


Subject(s)
Cell Differentiation/radiation effects , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus/radiation effects , Adult Stem Cells/cytology , Adult Stem Cells/radiation effects , Animals , Apoptosis/radiation effects , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Cell Proliferation/radiation effects , DNA Damage , Histones/metabolism , Intestinal Mucosa/radiation effects , Male , Mice , Mice, Inbred C57BL , Phosphorylation/radiation effects , Signal Transduction/radiation effects , Whole-Body Irradiation/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL