Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Abdom Radiol (NY) ; 49(3): 748-761, 2024 03.
Article in English | MEDLINE | ID: mdl-38236405

ABSTRACT

PURPOSE: To develop a diagnostic model for distinguishing pancreatobiliary-type and intestinal-type periampullary adenocarcinomas using preoperative contrast-enhanced computed tomography (CT) findings combined with clinical characteristics. METHODS: This retrospective study included 140 patients with periampullary adenocarcinoma who underwent preoperative enhanced CT, including pancreaticobiliary (N = 100) and intestinal (N = 40) types. They were randomly assigned to the training or internal validation set in an 8:2 ratio. Additionally, an independent external cohort of 28 patients was enrolled. Various CT features of the periampullary region were evaluated and data from clinical and laboratory tests were collected. Five machine learning classifiers were developed to identify the histologic type of periampullary adenocarcinoma, including logistic regression, random forest, multi-layer perceptron, light gradient boosting, and eXtreme gradient boosting (XGBoost). RESULTS: All machine learning classifiers except multi-layer perceptron used achieved good performance in distinguishing pancreatobiliary-type and intestinal-type adenocarcinomas, with the area under the curve (AUC) ranging from 0.75 to 0.98. The AUC values of the XGBoost classifier in the training set, internal validation set and external validation set are 0.98, 0.89 and 0.84 respectively. The enhancement degree of tumor, the growth pattern of tumor, and carbohydrate antigen 19-9 were the most important factors in the model. CONCLUSION: Machine learning models combining CT with clinical features can serve as a noninvasive tool to differentiate the histological subtypes of periampullary adenocarcinoma, in particular using the XGBoost classifier.


Subject(s)
Adenocarcinoma , Duodenal Neoplasms , Humans , Retrospective Studies , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Tomography, X-Ray Computed/methods , Machine Learning
2.
Nat Commun ; 15(1): 226, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38172511

ABSTRACT

Hematopoietic stem and progenitor cells generate all the lineages of blood cells throughout the lifespan of vertebrates. The emergence of hematopoietic stem and progenitor cells is finely tuned by a variety of signaling pathways. Previous studies have revealed the roles of pattern-recognition receptors such as Toll-like receptors and RIG-I-like receptors in hematopoiesis. In this study, we find that Nlrc3, a nucleotide-binding domain leucine-rich repeat containing family gene, is highly expressed in hematopoietic differentiation stages in vivo and vitro and is required in hematopoiesis in zebrafish. Mechanistically, nlrc3 activates the Notch pathway and the downstream gene of Notch hey1. Furthermore, NF-kB signaling acts upstream of nlrc3 to enhance its transcriptional activity. Finally, we find that Nlrc3 signaling is conserved in the regulation of murine embryonic hematopoiesis. Taken together, our findings uncover an indispensable role of Nlrc3 signaling in hematopoietic stem and progenitor cell emergence and provide insights into inflammation-related hematopoietic ontogeny and the in vitro expansion of hematopoietic stem and progenitor cells.


Subject(s)
Hematopoietic Stem Cells , Zebrafish , Animals , Mice , Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Hematopoiesis/genetics , Signal Transduction , Receptors, Notch/metabolism
3.
Stem Cell Res Ther ; 14(1): 117, 2023 04 30.
Article in English | MEDLINE | ID: mdl-37122014

ABSTRACT

BACKGROUND: In vertebrates, hematopoietic stem and progenitor cells (HSPCs) emerge from hemogenic endothelium in the floor of the dorsal aorta and subsequently migrate to secondary niches where they expand and differentiate into committed lineages. Glia maturation factor γ (gmfg) is a key regulator of actin dynamics that was shown to be highly expressed in hematopoietic tissue. Our goal is to investigate the role and mechanism of gmfg in embryonic HSPC development. METHODS: In-depth bioinformatics analysis of our published RNA-seq data identified gmfg as a cogent candidate gene implicated in HSPC development. Loss and gain-of-function strategies were applied to study the biological function of gmfg. Whole-mount in situ hybridization, confocal microscopy, flow cytometry, and western blotting were used to evaluate changes in the number of various hematopoietic cells and expression levels of cell proliferation, cell apoptosis and hematopoietic-related markers. RNA-seq was performed to screen signaling pathways responsible for gmfg deficiency-induced defects in HSPC initiation. The effect of gmfg on YAP sublocalization was assessed in vitro by utilizing HUVEC cell line. RESULTS: We took advantage of zebrafish embryos to illustrate that loss of gmfg impaired HSPC initiation and maintenance. In gmfg-deficient embryos, the number of hemogenic endothelium and HSPCs was significantly reduced, with the accompanying decreased number of erythrocytes, myelocytes and lymphocytes. We found that blood flow modulates gmfg expression and gmfg overexpression could partially rescue the reduction of HSPCs in the absence of blood flow. Assays in zebrafish and HUVEC showed that gmfg deficiency suppressed the activity of YAP, a well-established blood flow mediator, by preventing its shuttling from cytoplasm to nucleus. During HSPC initiation, loss of gmfg resulted in Notch inactivation and the induction of Notch intracellular domain could partially restore the HSPC loss in gmfg-deficient embryos. CONCLUSIONS: We conclude that gmfg mediates blood flow-induced HSPC maintenance via regulation of YAP, and contributes to HSPC initiation through the modulation of Notch signaling. Our findings reveal a brand-new aspect of gmfg function and highlight a novel mechanism for embryonic HSPC development.


Subject(s)
Glia Maturation Factor , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Glia Maturation Factor/genetics , Glia Maturation Factor/metabolism , Hematopoietic Stem Cells/metabolism , Zebrafish Proteins/genetics
4.
Ecotoxicol Environ Saf ; 244: 114020, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36049330

ABSTRACT

Occupational medicamentose-like dermatitis due to trichloroethylene (OMDT) is a key but unresolved question. OMDT patients often present multiple organ damage, including kidney damage. However, the underlying mechanism remains unknown. The purpose of our study was to explore the effect of tubule-specific C5b-9 deposition induced by TCE sensitization on renal tubular ferroptosis and its mechanism. By analyzing pathological changes of TCE-sensitization-mice kidney, we observed a significant renal tubular ferroptosis, which was alleviated by CD59, a C5b-9 inhibitory protein. Moreover, this phenomenon was also replicated in a C5b-9-attacked HK-2 cell model. Further experiments identified that C5b-9 induced cytosolic Ca2+ overload in renal tubular epithelia cells from TCE-sensitization-mice and HK-2 cells. Furthermore, in vitro experiments showed that BAPTA-AM, an intracellular Ca2+ chelator, could rescued ferroptosis induced by C5b-9 in HK-2 cells. Taken together, TCE sensitization induced renal tubular ferroptosis is mediated by C5b-9 and cytosolic Ca2+ overload may play a key role.


Subject(s)
Ferroptosis , Trichloroethylene , Animals , Chelating Agents , Complement Membrane Attack Complex/metabolism , Epithelial Cells/metabolism , Mice , Mice, Inbred BALB C , Trichloroethylene/toxicity
5.
Ecotoxicol Environ Saf ; 243: 114019, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36030685

ABSTRACT

Patients with trichloroethene-induced Trichloroethylene hypersensitivity syndrome (THS) often present kidney injury. However, the role of Wnt 5a/Ca2+ pathway in renal tubular injury in Trichloroethylene (TCE) sensitized mice remains unclear. This study aimed to investigate how Wnt 5a/Ca2+ pathway induced renal tubular epithelial cell injury in TCE sensitized mice. A total of 84 female BALB/c Specific Pathogen Free mice aged 6-8 weeks were used to establish TCE sensitized mouse models. Renal histology and serum levels of α1-MG and ß2-MG were used to assess the renal injury. The renal protein levels of Wnt 5a, ROR2, FZD5, PLC, p-CaMKII, IκB α, p-IκB α, NF-κB(p65), TNF α, IL 6 and IL 1ß were measured. The levels of serum α1-MG and ß2-MG and TNF α, IL 6 and IL 1ß levels in the kidney tissue were significantly increased in TCE sensitized positive group. However, Box5 pretreatment inhibited the expression of PLC, p-CaMKII, p65 and attenuated the injury of renal tubular epithelial cells and suppressed the upregulated expression of the above cytokines. In addition, KN93 also reduced nuclear translocation of p65 and renal injury as well as the elevated cytokines by inhibiting CaMKII. These data identify Wnt 5a binding to ROR2 and FZD5, p65 nuclear translocation, and inflammatory cytokine release as a novel mechanism for renal tubular epithelial cells injury by sensitization with TCE. Box5 or KN93 pretreatment can block the expression of inflammatory cytokines and reduce the injury of renal tubular epithelial cells.


Subject(s)
Calcium Signaling , Kidney , Wnt-5a Protein , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cytokines/metabolism , Epithelial Cells/metabolism , Female , Inflammation , Interleukin-6/metabolism , Kidney/pathology , Mice , Mice, Inbred BALB C , NF-KappaB Inhibitor alpha/metabolism , Trichloroethylene/toxicity , Tumor Necrosis Factor-alpha/metabolism , Wnt-5a Protein/metabolism
6.
Protein Cell ; 13(11): 808-824, 2022 11.
Article in English | MEDLINE | ID: mdl-35230662

ABSTRACT

Although widely applied in treating hematopoietic malignancies, transplantation of hematopoietic stem/progenitor cells (HSPCs) is impeded by HSPC shortage. Whether circulating HSPCs (cHSPCs) in steady-state blood could be used as an alternative source remains largely elusive. Here we develop a three-dimensional culture system (3DCS) including arginine, glycine, aspartate, and a series of factors. Fourteen-day culture of peripheral blood mononuclear cells (PBMNCs) in 3DCS led to 125- and 70-fold increase of the frequency and number of CD34+ cells. Further, 3DCS-expanded cHSPCs exhibited the similar reconstitution rate compared to CD34+ HSPCs in bone marrow. Mechanistically, 3DCS fabricated an immunomodulatory niche, secreting cytokines as TNF to support cHSPC survival and proliferation. Finally, 3DCS could also promote the expansion of cHSPCs in patients who failed in HSPC mobilization. Our 3DCS successfully expands rare cHSPCs, providing an alternative source for the HSPC therapy, particularly for the patients/donors who have failed in HSPC mobilization.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukocytes, Mononuclear , Antigens, CD34/metabolism , Hematopoietic Stem Cells , Humans , Leukocytes, Mononuclear/metabolism , Peptides/metabolism
7.
Front Psychiatry ; 13: 1082052, 2022.
Article in English | MEDLINE | ID: mdl-36713909

ABSTRACT

Background: Major depressive disorder (MDD) is a highly prevalent mental disease. Using magnetic resonance imaging (MRI), although numerous studies have revealed the alterations in structure and function of grey matter (GM), few studies focused on the synchronization of white matter (WM) structure and function in MDD. The aim of this study was to investigate whether functional and structural abnormalities of WM play an essential role in the neurobiological mechanisms of MDD. Methods: Gradient-echo imaging sequences at 3.0T were used to gather resting state functional MRI (rsfMRI) data, which were performed on 33 drug-naive first-episode MDD patients and 34 healthy controls (HCs). After data preprocessed, amplitude of low frequency fluctuation (ALFF) of WM was calculated. ALFF values in different frequency bands were analyzed, including typical (0.01-0.15 Hz) band, slow-4 (0.027-0.073 Hz) and slow-5 (0.01-0.027 Hz) bands. In addition, the fractional anisotropy (FA) values in WM in 23 patients and 26 HCs were examined using tract-based spatial statistics (TBSS) and tractography based on diffusion tensor imaging (DTI). Pearson correlation analysis was applied to analyze the relationships between ALFF values and Hamilton Depression Scale (HAMD) and Hamilton Anxiety Scale (HAMA). Results: Compared with the HCs, MDD patients showed decreased ALFF values in posterior thalamic radiation (PTR) and superior longitudinal fasciculus (SLF) in slow-5 frequency band, no significant differences of ALFF values were found in typical and slow-4 frequency bands. In addition, there were no significant differences in FA values with TBSS analysis as well as the number of fibers in PTR and SLF with tractography analysis between two groups. Further correlation analysis showed that the ALFF value in SLF was negatively correlated with HAMA-2 score (r = -0.548, p FDR = 0.037) in patients. Conclusion: Our results indicated that WM dysfunction may be associated with the pathophysiological mechanism of depression. Our study also suggested that the functional damage of the WM may precedes the structural damage in first-episode MDD patients. Furthermore, for mental disorders, slow-5 frequency band may be a more sensitive functional indicator for early detection of abnormal spontaneous brain activity in WM.

8.
Stem Cells Int ; 2021: 9919422, 2021.
Article in English | MEDLINE | ID: mdl-34434242

ABSTRACT

Widely known for self-renewal and multilineage differentiation, stem cells can be differentiated into all specialized tissues and cells in the body. In the past few years, a number of researchers have focused on deriving hematopoietic stem cells (HSCs) from pluripotent stem cells (PSCs) as alternative sources for clinic. Existing findings demonstrated that it is feasible to obtain HSCs and certain mature blood lineages from PSCs, except for several issues to be addressed. This short review outlines the technologies used for hematopoietic differentiation in recent years. In addition, the therapeutic value of PSCs as a potential source of various blood cells is also discussed as well as its challenges and directions in future clinical applications.

9.
Cell Mol Life Sci ; 78(16): 5881-5902, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34232331

ABSTRACT

Hematopoietic stem cells (HSCs) perceive both soluble signals and biomechanical inputs from their microenvironment and cells themselves. Emerging as critical regulators of the blood program, biomechanical cues such as extracellular matrix stiffness, fluid mechanical stress, confined adhesiveness, and cell-intrinsic forces modulate multiple capacities of HSCs through mechanotransduction. In recent years, research has furthered the scientific community's perception of mechano-based signaling networks in the regulation of several cellular processes. However, the underlying molecular details of the biomechanical regulatory paradigm in HSCs remain poorly elucidated and researchers are still lacking in the ability to produce bona fide HSCs ex vivo for clinical use. This review presents an overview of the mechanical control of both embryonic and adult HSCs, discusses some recent insights into the mechanisms of mechanosensing and mechanotransduction, and highlights the application of mechanical cues aiming at HSC expansion or differentiation.


Subject(s)
Biomechanical Phenomena/physiology , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/physiology , Cues , Extracellular Matrix/physiology , Humans , Mechanotransduction, Cellular/physiology , Signal Transduction/physiology , Stress, Mechanical
10.
Stem Cell Res ; 53: 102350, 2021 05.
Article in English | MEDLINE | ID: mdl-34087984

ABSTRACT

Human ELTD1 (Epidermal growth factor, latrophilin and seven-transmembrane domain-containing 1), an orphan G-protein-coupled receptor (GPCR) belonging to the adhesion GPCR family, has been reported as a novel regulator of angiogenesis and a potential anti-cancer therapeutic target. However, little is known about the function of ELTD1, especially its undiscovered ligands. In this experiment, an ELTD1 homozygous knockout human embryonic stem cell line, FAHZUe001-A, was generated by the iCRISPR/Cas9 system to achieve a deeper understanding of ELTD1. The FAHZUe001-A was confirmed with normal karyotype, typical undifferentiated morphology, pluripotency and trilineage differentiation potential in vitro.


Subject(s)
Human Embryonic Stem Cells , Neoplasms , CRISPR-Cas Systems/genetics , Epidermal Growth Factor , Humans , Neovascularization, Pathologic
11.
Exp Mol Med ; 51(10): 1-12, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31649245

ABSTRACT

Hematopoietic stem cells (HSCs) produce all lineages of mature blood cells for the lifetime of an organism. In vertebrates, HSCs derive from the transition of the hemogenic endothelium (HE) in the floor of the embryonic dorsal aorta. Most recently, a series of proinflammatory factors, such as tumor necrosis factor-α, interferon-γ, and Toll-like receptor 4, have been confirmed to play a key role in HSC specification. However, the full complement of necessary signaling inputs remains unknown to date. Here, we show that interleukin-6R (IL6R) via IL6 is required and sufficient for HSC generation. We found that Notch activates IL6R by regulating its expression in the HE and in HSCs. The secretion of IL6 mainly originates from HSC-independent myeloid cells, but not from HSCs and their adjacent vascular endothelial cells. In addition, blocking IL6 signaling does not affect vascular development or the production of primitive erythrocytes. Taken together, our results uncover a previously obscure relationship between IL6 signaling and HSC production and provide new insights into HSC regeneration using proinflammatory factors in vitro.


Subject(s)
Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Interleukin-6/genetics , Receptors, Interleukin-6/genetics , Animals , Cell Lineage/genetics , Erythrocytes/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Myeloid Cells/metabolism , Signal Transduction/genetics , Zebrafish/genetics
12.
Chemosphere ; 205: 662-673, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29723724

ABSTRACT

The exact toxicological mechanisms of paraquat (PQ) poisoning are not entirely clear, especially on the high-level acute exposure. To assess the health risk of PQ, especially to suicidal individuals, accidental ingestion eaters, occupational groups, and special multitude, firstly we explored the acute toxic effect and the possible mechanisms of high-level exposure of PQ using zebrafish. The mainly target organs of PQ were swim bladder which is the homolog of the mammalian lung, followed by gastrointestinal tract and liver. Morphological malformations which were further defined by histopathologic examination include smaller size, fibrosis and inflammatory cell invasion for swim bladder; irregularly arranged or dissolved epithelial folds, loss of villous architecture, and ecclasis of mucosal cells in a smaller lumen for gastrointestinal tract; as well as smaller size, degeneration, fibrous proliferation, atrophy for liver. In addition, PQ enhanced leukocyte recruitment (neutrophil migrated first, followed by macrophage) into swim bladder and induced ROS which can be scavenged by glutathione. Moreover, qRT-PCR results showed that PQ increased the expression level of genes involved in the inflammatory response, such as L-1ß, IL-6, IL-8, TNF-α, TNF-ß, IFN-1, TGF-ß, and NF-kB. For the first time, our results demonstrated that acute exposure of PQ induced pulmonary toxicity which was followed by gastrointestinal and hepatic toxicity via neutrophil-mediated ROS in zebrafish. In summary, these findings generated here will contribute to our better understanding of characteristics of PQ acute poisoning and can provide valuable information on better PQ poisoning treatments, occupational disease prevention, and providing theoretical foundation for risk management measures.


Subject(s)
Gastrointestinal Tract/pathology , Liver/pathology , Neutrophils/metabolism , Paraquat/toxicity , Reactive Oxygen Species/metabolism , Risk Assessment/methods , Urinary Bladder/pathology , Animals , Gastrointestinal Tract/metabolism , Humans , Liver/metabolism , Urinary Bladder/metabolism , Zebrafish
13.
Stem Cell Res Ther ; 8(1): 292, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29287601

ABSTRACT

BACKGROUND: Bone marrow mesenchymal stem cells (MSCs) are promising candidates for the treatment of various inflammatory disorders due to their profound immunomodulatory properties. However, the immunosuppressive capacity of MSCs needs activation by an inflammatory microenvironment, which may negatively impact the therapeutic effect because of increased immunogenicity. Here we explore the role of mammalian target of rapamycin (mTOR) signaling on the immunosuppressive capacity of MSCs, and its impact on immunogenicity in the inflammatory microenvironment. METHODS: Human bone marrow MSCs were cocultured with activated human peripheral blood mononuclear cells, CD4+ T cells, and mouse splenocytes to evaluate the immunosuppressive function. Immunosuppressive factors were assessed by quantitative real-time polymerase chain reaction (PCR), Western blot, and enzyme-linked immunosorbent assay (ELISA). The expression of major histocompatibility complex (MHC) was detected by flow cytometry. Short hairpin (sh)RNA was used to downregulate tuberous sclerosis complex (TSC)2, TSC1, and cyclooxygenase (COX)-2 in MSCs. RESULTS: Inhibition of mTOR signaling using rapamycin enhanced the immunosuppressive functions of MSCs, while prolonged exposure to rapamycin did not. The enhancement of the immunosuppressive function was independent of the inflammatory microenvironment, and occurred mainly through the upregulation of COX-2 and prostaglandin-E2 (PGE2) expression. Furthermore, mTOR inhibition did not impact the immunogenicity of MSCs. However, the upregulated expression of MHC class II molecules by interferon (IFN)-γ was attenuated by mTOR inhibition, whereas TSC2 knockdown had the opposite effect. CONCLUSIONS: These results reveal that the mTOR signaling pathway regulates MSC immunobiology, and short-term exposure to rapamycin could be a novel approach to improve the MSC-based therapeutic effect.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Immunomodulation/immunology , Mesenchymal Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Humans , Mesenchymal Stem Cells/cytology , Mice , Signal Transduction
14.
J Hematol Oncol ; 9(1): 102, 2016 Sep 29.
Article in English | MEDLINE | ID: mdl-27686241

ABSTRACT

BACKGROUND: The efficient generation of hematopoietic stem cells (HSCs) from human-induced pluripotent stem cells (iPSCs) holds great promise in personalized transplantation therapies. However, the derivation of functional and transplantable HSCs from iPSCs has had very limited success thus far. METHODS: We developed a synthetic 3D hematopoietic niche system comprising nanofibers seeded with bone marrow (BM)-derived stromal cells and growth factors to induce functional hematopoietic cells from human iPSCs in vitro. RESULTS: Approximately 70 % of human CD34+ hematopoietic cells accompanied with CD43+ progenitor cells could be derived from this 3D induction system. Colony-forming-unit (CFU) assay showed that iPSC-derived CD34+ cells formed all types of hematopoietic colonies including CFU-GEMM. TAL-1 and MIXL1, critical transcription factors associated with hematopoietic development, were expressed during the differentiation process. Furthermore, iPSC-derived hematopoietic cells gave rise to both lymphoid and myeloid lineages in the recipient NOD/SCID mice after transplantation. CONCLUSIONS: Our study underscores the importance of a synthetic 3D niche system for the derivation of transplantable hematopoietic cells from human iPSCs in vitro thereby establishing a foundation towards utilization of human iPSC-derived HSCs for transplantation therapies in the clinic.

SELECTION OF CITATIONS
SEARCH DETAIL
...