1.Deep overparameterized blood cell detection algorithm utilizing hybrid attention mechanisms.
Shuo ZHU ; Xukang ZHANG ; Zongyang WANG ; Rui JIANG ; Zhengda LIU
Journal of Biomedical Engineering 2025;42(5):936-944
To address the challenges in blood cell recognition caused by diverse morphology, dense distribution, and the abundance of small target information, this paper proposes a blood cell detection algorithm - the "You Only Look Once" model based on hybrid mixing attention and deep over-parameters (HADO-YOLO). First, a hybrid attention mechanism is introduced into the backbone network to enhance the model's sensitivity to detailed features. Second, the standard convolution layers with downsampling in the neck network are replaced with deep over-parameterized convolutions to expand the receptive field and improve feature representation. Finally, the detection head is decoupled to enhance the model's robustness for detecting abnormal cells. Experimental results on the Blood Cell Counting Dataset (BCCD) demonstrate that the HADO-YOLO algorithm achieves a mean average precision of 90.2% and a precision of 93.8%, outperforming the baseline YOLO model. Compared with existing blood cell detection methods, the proposed algorithm achieves state-of-the-art detection performance. In conclusion, HADO-YOLO offers a more efficient and accurate solution for identifying various types of blood cells, providing valuable technical support for future clinical diagnostic applications.
Algorithms
;
Humans
;
Blood Cells/cytology*
;
Blood Cell Count/methods*
;
Neural Networks, Computer
;
Deep Learning
;
Detection Algorithms
2.High mobility group protein B1(HMGB1) promotes myeloid dendritic cell maturation and increases Th17 cell/Treg cell ratio in patients with immune primary thrombocytopenia.
Qinzhi LI ; Dongsheng DUAN ; Xiujuan WANG ; Mingling SUN ; Ying LIU ; Xinyou WANG ; Lei WANG ; Wenxia FAN ; Mengting SONG ; Xinhong GUO
Chinese Journal of Cellular and Molecular Immunology 2025;41(1):45-50
Objective This study investigated the regulatory effect of high mobility group protein B1 (HMGB1) in the peripheral blood of patients with primary immune thrombocytopenia (ITP) on myeloid dendritic cells (mDC) and Th17/regulatory T cells (Treg) balance. Methods The study enrolled 30 newly diagnosed ITP patients and 30 healthy controls.Flow cytometry was used to measure the proportion of mDC, Th17, and Treg cells in the peripheral blood of ITP patients and healthy controls. ELISA was conducted to quantify the serum levels of HMGB1, interleukin 6 (IL-6), IL-23, IL-17, and transforming growth factor β(TGF-β). The mRNA levels of retinoic acid-related orphan receptor γt(RORγt) and forehead box P3(FOXP3) were detected by real-time PCR. The correlation between the abovementioned cells, cytokines, and platelet count was assessed using Pearson linear correlation analysis. Results The proportion of Th17 cells and the expression levels of HMGB1, IL-6, IL-23, IL-17 and the level of RORγt mRNA in the peripheral blood of ITP patients were higher than those in healthy controls. However, the Treg cell proportion and TGF-β level were lower in ITP patients than those in healthy controls. In patients with ITP, the proportion of mDC and the level of FOXP3 mRNA did not show significant changes. The proportion of mDC cells was significantly correlated with the expression of IL-6 and IL-23. Moreover, the expression of HMGB1 showed a significant correlation with the expression of mDC, IL-6, IL-23, RORγt mRNA, and IL-17. Notably, both the proportion of mDC cells and the expression of HMGB1 were negatively correlated with platelet count. Conclusion The high expression of HMGB1 in peripheral blood of ITP patients may induce Th17/Treg imbalance by promoting the maturation of mDC and affecting the secretion of cytokines, thereby potentially playing a role in the immunological mechanism of ITP.
Humans
;
Th17 Cells/cytology*
;
HMGB1 Protein/genetics*
;
T-Lymphocytes, Regulatory/cytology*
;
Female
;
Male
;
Dendritic Cells/metabolism*
;
Adult
;
Middle Aged
;
Purpura, Thrombocytopenic, Idiopathic/genetics*
;
Nuclear Receptor Subfamily 1, Group F, Member 3/genetics*
;
Young Adult
;
Interleukin-23/blood*
;
Interleukin-17/blood*
;
Interleukin-6/blood*
;
Forkhead Transcription Factors/genetics*
;
Myeloid Cells/cytology*
;
Aged
3.Establishment and Application of an in Vitro Cellular Model of Adipogenic Differentiation of Bone Marrow Mesenchymal Stem Cells with Serum Injury in aGVHD Mouse.
Run-Xiang XU ; Pei-Lin LI ; Jia-Yi TIAN ; Jie TANG ; Bo-Feng YIN ; Fu-Hao YU ; Fei-Yan WANG ; Xiao-Tong LI ; Xiao-Yu ZHANG ; Wen-Rong XIA ; Heng ZHU ; Li DING
Journal of Experimental Hematology 2025;33(1):255-261
OBJECTIVE:
To establish an in vitro cell model simulating acute graft-versus-host disease (aGVHD) bone marrow microenvironment injury with the advantage of mouse serum of aGVHD model and explore the effect of serum of aGVHD mouse on the adipogenic differentiation ability of mesenchymal stem cells (MSCs).
METHODS:
The 6-8-week-old C57BL/6N female mice and BALB/c female mice were used as the donor and recipient mice of the aGVHD model, respectively. Bone marrow transplantation (BMT) mouse model (n=20) was established by being injected with bone marrow cells (1×107 per mouse) from donor mice within 4-6 hours after receiving a lethal dose (8.0 Gy, 72.76 cGy/min) of γ ray general irradiation. A mouse model of aGVHD (n=20) was established by infusing a total of 0.4 ml of a mixture of donor mouse-derived bone marrow cells (1×107 per mouse) and spleen lymphocytes (2×106 per mouse). The blood was removed from the eyeballs and the mouse serum was aspirated on the 7th day after modeling. Bone marrow-derived MSCs were isolated from 1-week-old C57BL/6N male mice and incubated with 2%, 5% and 10% BMT mouse serum and aGVHD mouse serum in the medium, respectively. The effect of serum in the two groups on the in vitro adipogenic differentiation ability of mouse MSCs was detected by Oil Red O staining. The expression levels of related proteins PPARγ and CEBPα were detected by Western blot. The expression differences of key adipogenic transcription factors including PPARγ, CEBPα, FABP4 and LPL were determined by real-time quantitative PCR (RT-qPCR).
RESULTS:
An in vitro cell model simulating the damage of bone marrow microenvironment in mice with aGVHD was successfully established. Oil Red O staining showed that the number of orange-red fatty droplets was significantly reduced and the adipogenic differentiation ability of MSC was impaired at aGVHD serum concentration of 10% compared with BMT serum. Western blot experiments showed that adipogenesis-related proteins PPARγ and CEBPα expressed in MSCs were down-regulated. Further RT-qPCR assay showed that the production of PPARγ, CEBPα, FABP4 and LPL, the key transcription factors for adipogenic differentiation of MSC, were significantly reduced.
CONCLUSION
The adipogenic differentiation capacity of MSCs is inhibited by aGVHD mouse serum.
Animals
;
Mesenchymal Stem Cells/cytology*
;
Mice
;
Mice, Inbred BALB C
;
Mice, Inbred C57BL
;
Adipogenesis
;
Female
;
Cell Differentiation
;
Graft vs Host Disease/blood*
;
Bone Marrow Cells/cytology*
;
PPAR gamma/metabolism*
;
Disease Models, Animal
;
CCAAT-Enhancer-Binding Protein-alpha/metabolism*
4.Pig meniscus single-cell sequencing reveals highly active red zone chondrocyte populations involved in stemness maintenance and vascularization development.
Monika MANKOWSKA ; Monika STEFANSKA ; Anna Maria MLECZKO ; Katarzyna SARAD ; Witold KOT ; Lukasz KRYCH ; Julia Anna SEMBA ; Eric Lars-Helge LINDBERG ; Jakub Dalibor RYBKA
Journal of Zhejiang University. Science. B 2025;26(7):675-693
Meniscus injuries are widespread and the available treatments do not offer enough healing potential. Here, we provide critical support for using pigs as a biological model for meniscal degeneration and the development of cutting-edge therapies in orthopedics. We present a single-cell transcriptome atlas of the meniscus, consisting of cell clusters corresponding to four major cell types: chondrocytes, endothelial cells, smooth muscle cells, and immune cells. Five distinct chondrocyte subclusters (CH0‒CH4) were annotated, of which only one was widespread in both the red and white zones, indicating a major difference in the cellular makeup of the zones. Subclusters distinct to the white zone appear responsible for cartilage-specific matrix deposition and protection against adverse microenvironmental factors, while those in the red zone exhibit characteristics of mesenchymal stem cells and are more likely to proliferate and migrate. Additionally, they induce remodeling actions in other chondrocyte subclusters and promote the proliferation and maturation of endothelial cells, inducing healing and vascularization processes. Considering that they have substantial remodeling capabilities, these subclusters should be of great interest for tissue engineering studies. We also show that the cellular makeup of the pig meniscus is comparable to that of humans, which supports the use of pigs as a model in orthopedic therapy development.
Animals
;
Swine
;
Chondrocytes/physiology*
;
Single-Cell Analysis
;
Meniscus/blood supply*
;
Endothelial Cells/cytology*
;
Transcriptome
;
Mesenchymal Stem Cells/cytology*
;
Neovascularization, Physiologic
;
Cell Proliferation
5.S1PR5 activation or overexpression enhances barrier function of mouse brain microvascular endothelial cells against OGD/R injury by modulating oxidative stress.
Jingxian WANG ; Zijing REN ; Peiyang ZHOU
Journal of Southern Medical University 2025;45(7):1451-1459
OBJECTIVES:
To investigate the role of sphingosine-1-phosphate receptor 5 (S1PR5) in modulating barrier function of mouse brain microvascular endothelial cells with oxygen-glucose deprivation and reoxygenation (OGD/R).
METHODS:
Mouse brain microvascular endothelial cells (bEnd.3) were exposed to OGD/R to induce barrier dysfunction following treatment with S1PR5-specific agonist A971432 or lentivirus-mediated transfection with a S1PR5-specific siRNA, a S1PR5-overexpressing plasmid, or their respective negative control sequences. The changes in viability and endothelial barrier permeability of the treated cells were evaluated with CCK-8 assay and FITC-dextran permeability assay; the levels of intracellular reactive oxygen species (ROS) and localization and expression levels of the proteins related with barrier function and oxidative stress were detected using immunofluorescence staining, DCFH-DA probe and Western blotting.
RESULTS:
S1PR5 activation obviously enhanced viability of bEnd.3 cells exposed to OGD/R (P<0.0001). Both activation and overexpression of S1PR5 reduced FITC-dextran leakage, while S1PR5 knockdown significantly increased FITC-dextran leakage in the exposed bEnd.3 cells. Activation and overexpression of S1PR5 both increased the cellular expressions of the barrier proteins ZO-1 and occludin, while S1PR5 knockdown produced the opposite effect. In cells exposed to OGD/R, ROS production was significantly reduced by S1PR5 activation and overexpression but increased following S1PR5 knockdown. Overexpression of S1PR5 obviously increased the expressions of the antioxidant proteins Nrf2, HO-1 and SOD2 in the exposed cells.
CONCLUSIONS
S1PR5 activation and overexpression significantly improve cell viability and reduce permeability of a mouse brain microvascular endothelial cell model of OGD/R, the mechanism of which may involve the reduction in ROS production and upregulation of the antioxidant proteins.
Animals
;
Mice
;
Oxidative Stress
;
Endothelial Cells/cytology*
;
Brain/blood supply*
;
Reactive Oxygen Species/metabolism*
;
Receptors, Lysosphingolipid/metabolism*
;
Sphingosine-1-Phosphate Receptors
;
Blood-Brain Barrier/metabolism*
;
Glucose
;
Cell Line
;
Oxygen/metabolism*
;
NF-E2-Related Factor 2/metabolism*
6.Mechanism of human embryonic stem cell-derived mesenchymal stem cells on alleviating brain injury after cardiopulmonary resuscitation in swine with cardiac arrest.
Feng GE ; Jiefeng XU ; Jinjiang ZHU ; Guangli CAO ; Xuguang WANG ; Meiya ZHOU ; Tiejiang CHEN ; Mao ZHANG
Chinese Critical Care Medicine 2025;37(2):133-139
OBJECTIVE:
To investigate the mechanism of human embryonic stem cell-derived mesenchymal stem cells (hESC-MSC) in alleviating brain injury after resuscitation in swine with cardiac arrest (CA).
METHODS:
Twenty-nine healthy male large white swine were randomly divided into Sham group (n = 9), cardiopulmonary resuscitation (CPR) group (n = 10) and hESC-MSC group (n = 10). The Sham group only completed animal preparation. In CPR group and hESC-MSC group, the swine model of CA-CPR was established by inducing ventricular fibrillation for 10 minutes with electrical stimulation and CPR for 6 minutes. At 5 minutes after successful resuscitation, hESC-MSC 2.5×106/kg was injected via intravenous micropump within 1 hour in hESC-MSC group. Venous blood samples were collected before resuscitation and at 4, 8, 24, 48 and 72 hours of resuscitation. The levels of neuron specific enolase (NSE) and S100B protein (S100B) were detected by enzyme linked immunosorbent assay (ELISA). At 24, 48 and 72 hours of resuscitation, neurological deficit score (NDS) and cerebral performance category (CPC) were used to evaluate the neurological function of the animals. Three animals from each group were randomly selected and euthanized at 24, 48, and 72 hours of resuscitation, and the hippocampus tissues were quickly obtained. Immunofluorescence staining was used to detect the distribution of hESC-MSC in hippocampus. Immunohistochemical staining was used to detect the activation of astrocytes and microglia and the survival of neurons in the hippocampus. The degree of apoptosis was detected by TdT-mediated dUTP nick end labeling (TUNEL).
RESULTS:
The serum NSE and S100B levels of brain injury markers in CPR group and hESC-MSC group were significantly higher than those in Sham group at 24 hours of resuscitation, and then gradually increased. The levels of NSE and S100B in serum at each time of resuscitation in hESC-MSC group were significantly lower than those in CPR group [NSE (μg/L): 20.69±3.62 vs. 28.95±3.48 at 4 hours, 27.04±5.56 vs. 48.59±9.22 at 72 hours; S100B (μg/L): 2.29±0.39 vs. 3.60±0.73 at 4 hours, 2.38±0.15 vs. 3.92±0.50 at 72 hours, all P < 0.05]. In terms of neurological function, compared with the Sham group, the NDS score and CPC score in the CPR group and hESC-MSC group increased significantly at 24 hours of resuscitation, and then gradually decreased. The NDS and CPC scores of hESC-MSC group were significantly lower than those of CPR group at 24 hours of resuscitation (NDS: 111.67±20.21 vs. 170.00±21.79, CPC: 2.33±0.29 vs. 3.00±0.00, both P < 0.05). The expression of hESC-MSC positive markers CD73, CD90 and CD105 in the hippocampus of hESC-MSC group at 24, 48 and 72 hours of resuscitation was observed under fluorescence microscope, indicating that hESC-MSC could homing to the damaged hippocampus. In addition, compared with Sham group, the proportion of astrocytes, microglia and apoptotic index in hippocampus of CPR group were significantly increased, and the proportion of neurons was significantly decreased at 24, 48 and 72 hours of resuscitation. Compared with CPR group, the proportion of astrocytes, microglia and apoptotic index in hippocampus of hESC-MSC group decreased and the proportion of neurons increased significantly at 24 hours of resuscitation [proportion of astrocytes: (14.33±1.00)% vs. (30.78±2.69)%, proportion of microglia: (12.00±0.88)% vs. (27.89±5.68)%, apoptotic index: (12.89±3.86)% vs. (52.33±7.77)%, proportion of neurons: (39.44±3.72)% vs. (28.33±1.53)%, all P < 0.05].
CONCLUSIONS
Application of hESC-MSC at the early stage of resuscitation can reduce the brain injury and neurological dysfunction after resuscitation in swine with CA. The mechanism may be related to the inhibition of immune cell activation, reduction of cell apoptosis and promotion of neuronal survival.
Animals
;
Heart Arrest/therapy*
;
Cardiopulmonary Resuscitation
;
Swine
;
Humans
;
Male
;
Human Embryonic Stem Cells/cytology*
;
Mesenchymal Stem Cell Transplantation
;
Mesenchymal Stem Cells/cytology*
;
Phosphopyruvate Hydratase/blood*
;
Brain Injuries/therapy*
;
S100 Calcium Binding Protein beta Subunit
;
Apoptosis
;
Disease Models, Animal
7.Research advances in relationship between biological clock and cardiovascular diseases.
Ting-Ting JIANG ; Shuang JI ; Guang-Rui YANG ; Li-Hong CHEN
Acta Physiologica Sinica 2019;71(5):783-791
Circadian rhythms widely exist in living organisms, and they are regulated by the biological clock. Growing evidence has shown that circadian rhythms are tightly related to the physiological function of the cardiovascular system, including blood pressure, heart rate, metabolism of cardiomyocytes, function of endothelial cells, and vasoconstriction and vasodilation. In addition, disruption of circadian rhythms has been considered as one of the important risk factors for cardiovascular diseases, such as myocardial infarction. This review summarizes the recent research advances in the relationship between circadian clock and cardiovascular diseases, hoping to improve treatment strategies for patients with cardiovascular diseases according to the theory of biological clock.
Blood Pressure
;
Cardiovascular Diseases
;
physiopathology
;
Circadian Clocks
;
Circadian Rhythm
;
Endothelial Cells
;
cytology
;
Heart Rate
;
Humans
;
Myocytes, Cardiac
;
metabolism
;
Vasoconstriction
;
Vasodilation
8.Glycosylation of dentin matrix protein 1 is a novel key element for astrocyte maturation and BBB integrity.
Bo JING ; Chunxue ZHANG ; Xianjun LIU ; Liqiang ZHOU ; Jiping LIU ; Yinan YAO ; Juehua YU ; Yuteng WENG ; Min PAN ; Jie LIU ; Zuolin WANG ; Yao SUN ; Yi Eve SUN
Protein & Cell 2018;9(3):298-309
The blood-brain barrier (BBB) is a tight boundary formed between endothelial cells and astrocytes, which separates and protects brain from most pathogens as well as neural toxins in circulation. However, detailed molecular players involved in formation of BBB are not completely known. Dentin matrix protein 1 (DMP1)-proteoglycan (PG), which is known to be involved in mineralization of bones and dentin, is also expressed in soft tissues including brain with unknown functions. In the present study, we reported that DMP1-PG was expressed in brain astrocytes and enriched in BBB units. The only glycosylation site of DMP1 is serine89 (S89) in the N-terminal domain of the protein in mouse. Mutant mice with DMP1 point mutations changing S89 to glycine (S89G), which completely eradicated glycosylation of the protein, demonstrated severe BBB disruption. Another breed of DMP1 mutant mice, which lacked the C-terminal domain of DMP1, manifested normal BBB function. The polarity of S89G-DMP1 astrocytes was disrupted and cell-cell adhesion was loosened. Through a battery of analyses, we found that DMP1 glycosylation was critically required for astrocyte maturation both in vitro and in vivo. S89G-DMP1 mutant astrocytes failed to express aquaporin 4 and had reduced laminin and ZO1 expression, which resulted in disruption of BBB. Interestingly, overexpression of wild-type DMP1-PG in mouse brain driven by the nestin promoter elevated laminin and ZO1 expression beyond wild type levels and could effectively resisted intravenous mannitol-induced BBB reversible opening. Taken together, our study not only revealed a novel element, i.e., DMP1-PG, that regulated BBB formation, but also assigned a new function to DMP1-PG.
Animals
;
Astrocytes
;
cytology
;
metabolism
;
Blood-Brain Barrier
;
cytology
;
metabolism
;
Cells, Cultured
;
Extracellular Matrix Proteins
;
genetics
;
metabolism
;
Female
;
Glycosylation
;
Male
;
Mice
;
Proteoglycans
;
metabolism
;
Reverse Transcriptase Polymerase Chain Reaction
9.CRISPR/Cas9-mediated gene knockout reveals a guardian role of NF-κB/RelA in maintaining the homeostasis of human vascular cells.
Ping WANG ; Zunpeng LIU ; Xiaoqian ZHANG ; Jingyi LI ; Liang SUN ; Zhenyu JU ; Jian LI ; Piu CHAN ; Guang-Hui LIU ; Weiqi ZHANG ; Moshi SONG ; Jing QU
Protein & Cell 2018;9(11):945-965
Vascular cell functionality is critical to blood vessel homeostasis. Constitutive NF-κB activation in vascular cells results in chronic vascular inflammation, leading to various cardiovascular diseases. However, how NF-κB regulates human blood vessel homeostasis remains largely elusive. Here, using CRISPR/Cas9-mediated gene editing, we generated RelA knockout human embryonic stem cells (hESCs) and differentiated them into various vascular cell derivatives to study how NF-κB modulates human vascular cells under basal and inflammatory conditions. Multi-dimensional phenotypic assessments and transcriptomic analyses revealed that RelA deficiency affected vascular cells via modulating inflammation, survival, vasculogenesis, cell differentiation and extracellular matrix organization in a cell type-specific manner under basal condition, and that RelA protected vascular cells against apoptosis and modulated vascular inflammatory response upon tumor necrosis factor α (TNFα) stimulation. Lastly, further evaluation of gene expression patterns in IκBα knockout vascular cells demonstrated that IκBα acted largely independent of RelA signaling. Taken together, our data reveal a protective role of NF-κB/RelA in modulating human blood vessel homeostasis and map the human vascular transcriptomic landscapes for the discovery of novel therapeutic targets.
Blood Vessels
;
cytology
;
metabolism
;
CRISPR-Cas Systems
;
Embryonic Stem Cells
;
cytology
;
Gene Knockout Techniques
;
Homeostasis
;
Humans
;
NF-kappa B
;
deficiency
;
metabolism
;
Transcription Factor RelA
;
deficiency
;
metabolism
10.Cancer stem-like cell-derived exosomes promotes the proliferation and invasion of human umbilical cord blood-derived mesenchymal stem cells.
Dan ZHANG ; Dawei HE ; Dian LI ; Bo TANG ; Dong HU ; Wenhao GUO ; Zhang WANG ; Lianju SHEN ; Guanghui WEI
Journal of Southern Medical University 2018;38(12):1440-1447
OBJECTIVE:
To investigate the effect of Piwil2-induced cancer stem-like cell (Piwil2-iCSC)-derived exosomes on the proliferation,migration and invasion of human umbilical cord blood-derived mesenchymal stem cells (hucMSCs).
METHODS:
Piwil2-iCSC-derived exosomes were isolated by ultracentrifugation and identified using transmission electron microscopy,nanoparticle tracking analysis and Western blotting.Exosome uptake assay was used to identify the pathway that Piwil2-iCSCderived exosomes utilized.HucMSCs were divided into control group,PBS intervention group and exosome intervention group,and CCK-8 assay,wound healing assay,Transwell assay,Western blotting and cell karyotype analysis were used to observe the proliferation,migration,invasion,expression levels of MMP2 and MMP9 proteins,and chromosome structure of hucMSCs.
RESULTS:
The diameter of Piwil2-iCSC-derived exosomes ranged from 50 nm to 100 nm,and most of them were oval or spherical capsules rich in CD9,CD63 and Piwil2 proteins.Exosomal uptake assay showed that the exosomes executed theirs functions after entering the cells.Compared with the control cells and PBS-treated cells,hucMSCs treated with the exosomes showed significantly increased number of proliferating cells (<0.05) with accelerated healing rate (<0.05 at 24 h;<0.01 at 48 h),increased invasive cells (<0.01),enhanced protein expressions of MMP2(<0.05 PBS group;<0.01 control group) and MMP9(<0.05),but their karyotype still remained 46XY without any abnormalities.
CONCLUSIONS
Piwil2-iCSC-derived exosomes can promote the proliferation,migration and invasion but does not cause cancer-like heterogeneity changes in hucMSCs.
Argonaute Proteins
;
Cell Movement
;
physiology
;
Cell Proliferation
;
physiology
;
Exosomes
;
physiology
;
Fetal Blood
;
cytology
;
Humans
;
Karyotyping
;
Mesenchymal Stem Cells
;
pathology
;
Neoplasm Invasiveness
;
Neoplastic Stem Cells
;
Umbilical Cord
;
Wound Healing

Result Analysis
Print
Save
E-mail