1.Glutamatergic neurons in thalamic paraventricular nucleus may be involved in the regulation of abnormal sleep behavior of Shank3 gene knockout mice.
Chang-Feng CHEN ; Lie-Cheng WANG ; Yong LIU ; Lei CHEN
Acta Physiologica Sinica 2025;77(5):792-800
The purpose of this study was to investigate the anxiety-like behaviors, circadian rhythms and sleep, and to elucidate the possible underlying mechanisms of the abnormal sleep behavior in Shank3 gene knockout (Shank3-KO) mice. The anxiety-like behaviors were detected by elevated plus-maze (EPM) test, open field test (OFT) and tail suspension test (TST). The circadian rhythms were detected by running wheel test. The electroencephalogram (EEG)/electromyogram (EMG) recordings were performed synchronically by polysomnograph. The distribution of SHANK3 in anterior cingulate cortex (ACC), paraventricular thalamus (PVT), nucleus accumbens (NAc), basolateral amygdala (BLA) and hippocampal CA2 region in wild type (WT) mice was detected by immunofluorescence assay. The protein expression of c-Fos in PVT, ACC and NAc was also detected by immunofluorescence assay during light cycle. The colocalization of c-Fos and vesicular glutamate transporter 2 (Vglut2, a marker for glutamatergic neurons) in the PVT was detected by immunofluorescence double labeling experiment. The results of EPM test showed that, compared with the WT mice, the Shank3-KO mice showed less time in open arms and less number of open arm entries. The results of OFT showed that the Shank3-KO mice showed less time in central area and less number of central area entries. The immobility time of Shank3-KO mice was increased in the TST. The results of running wheel rhythm test showed that the phase shift time of Shank3-KO mice in the continuous dark period was increased. The results of EEG/EMG recording showed that, compared with the WT mice, the duration of wakefulness in Shank3-KO mice was increased and the duration of non-rapid eye movement (NREM) sleep was decreased during light phase; The bout number of wakefulness was increased, the bout number of NREM sleep was decreased, NREM-wake transitions were increased, and wake-NREM transitions were decreased during light phase. SHANK3 was expressed in ACC, PVT, NAc and BLA in the WT mice. The expression of c-Fos in the PVT of Shank3-KO mice was up-regulated 2 h after entering the light phase, and majority of c-Fos was co-localized with Vglut2. These results suggest that the anxiety level of Shank3-KO mice is increased, the regulation of the internal rhythms is decreased, and the bout number of wakefulness is increased during light phase. The glutamatergic neurons in PVT may be involved in the regulation of abnormal sleep behavior in Shank3-KO mice during the light phase.
Animals
;
Mice, Knockout
;
Mice
;
Neurons/metabolism*
;
Nerve Tissue Proteins/physiology*
;
Male
;
Midline Thalamic Nuclei/cytology*
;
Circadian Rhythm/physiology*
;
Sleep/physiology*
;
Anxiety/physiopathology*
;
Proto-Oncogene Proteins c-fos/metabolism*
;
Vesicular Glutamate Transport Protein 2/metabolism*
;
Mice, Inbred C57BL
;
Microfilament Proteins
2.Effect of removing microglia from spinal cord on nerve repair after spinal cord injury in mice.
Qi JIANG ; Chao QI ; Yuerong SUN ; Shiyuan XUE ; Xinyi WEI ; Haitao FU
Chinese Journal of Reparative and Reconstructive Surgery 2025;39(6):754-761
OBJECTIVE:
To investigate the effects of removing microglia from spinal cord on nerve repair and functional recovery after spinal cord injury (SCI) in mice.
METHODS:
Thirty-nine 6-week-old female C57BL/6 mice were randomly divided into control group ( n=12), SCI group ( n=12), and PLX3397+SCI group ( n=15). The PLX3397+SCI group received continuous feeding of PLX3397, a colony-stimulating factor 1 receptor inhibitor, while the other two groups were fed a standard diet. After 14 days, both the SCI group and the PLX3397+SCI group were tested for ionized calcium binding adapter molecule 1 (Iba1) to confirm that the PLX3397+SCI group had completely depleted the spinal cord microglia. The SCI model was then prepared by clamping the spinal cord in both the SCI group and the PLX3397+SCI group, while the control group underwent laminectomy. Preoperatively and at 1, 3, 7, 14, 21, and 28 days postoperatively, the Basso Mouse Scale (BMS) was used to assess the hind limb function of mice in each group. At 28 days, a footprint test was conducted to observe the gait of the mice. After SCI, spinal cord tissue from the injury site was taken, and Iba1 immunofluorescence staining was performed at 7 days to observe the aggregation and proliferation of microglia in the spinal cord. HE staining was used to observe the formation of glial scars at the injury site at 28 days; glial fibrillary acidic protein (GFAP) immunofluorescence staining was applied to astrocytes to assess the extent of the injured area; neuronal nuclei antigen (NeuN) immunofluorescence staining was used to evaluate neuronal survival. And 5-hydroxytryptamine (5-HT) immunofluorescence staining was performed to assess axonal survival at 60 days.
RESULTS:
All mice survived until the end of the experiment. Immunofluorescence staining revealed that the microglia in the spinal cord of the PLX3397+SCI group decreased by more than 95% compared to the control group after 14 days of continuous feeding with PLX3397 ( P<0.05). Compared to the control group, the BMS scores in the PLX3397+SCI group and the SCI group significantly decreased at different time points after SCI ( P<0.05). Moreover, the PLX3397+SCI group showed a further decrease in BMS scores compared to the SCI group, and exhibited a dragging gait. The differences between the two groups were significant at 14, 21, and 28 days ( P<0.05). HE staining at 28 days revealed that the SCI group had formed a well-defined and dense gliotic scar, while the PLX3397+SCI group also developed a gliotic scar, but with a more blurred and loose boundary. Immunofluorescence staining revealed that the number of microglia near the injury center at 7 days increased in the SCI group than in the control group, but the difference between groups was not significant ( P>0.05). In contrast, the PLX3397+SCI group showed a significant reduction in microglia compared to both the control and SCI groups ( P<0.05). At 28 days after SCI, the area of spinal cord injury in the PLX3397+SCI group was significantly larger than that in SCI group ( P<0.05); the surviving neurons significantly reduced compared with the control group and SCI group ( P<0.05). The axonal necrosis and retraction at 60 days after SCI were more obvious.
CONCLUSION
The removal of microglia in the spinal cord aggravate the tissue damage after SCI and affecte the recovery of motor function in mice, suggesting that microglia played a neuroprotective role in SCI.
Animals
;
Spinal Cord Injuries/surgery*
;
Microglia/pathology*
;
Female
;
Mice
;
Mice, Inbred C57BL
;
Nerve Regeneration/drug effects*
;
Spinal Cord/pathology*
;
Pyrroles/administration & dosage*
;
Aminopyridines/administration & dosage*
;
Recovery of Function
;
Disease Models, Animal
;
Calcium-Binding Proteins/metabolism*
;
Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors*
;
Microfilament Proteins/metabolism*
;
Glial Fibrillary Acidic Protein/metabolism*
3.Research on the mechanism of gentiopicroside preventing macrophage-mediated liver fibrosis by regulating the MIF-SPP1 signaling pathway in hepatic stellate cells.
Jixu WANG ; Yingbin ZHU ; Maoli CHEN ; Yongfeng HAN
Chinese Journal of Cellular and Molecular Immunology 2025;41(7):593-602
Objective To explore the mechanism by which gentiopicroside (GPS) prevents macrophage-mediated hepatic fibrosis by regulating the macrophage migration inhibitory factor (MIF)-secreted phosphoprotein 1 (SPP1) signaling pathway in hepatic stellate cells. Methods LX-2 cells were divided into control group, transforming growth factor β(TGF-β) group, and TGF-β combined with GPS (25, 50, 100, 150 μmol/mL) groups. Cell proliferation was detected by EDU assay, cell invasion was assessed by TranswellTM assay, and the protein expressions of α-smooth muscle actin (α-SMA) and type I collagen (COL1A1) were measured by Western blot. M1-type macrophage-conditioned medium (M1-CM) was used to treat LX-2 cells in the TGF-β group and TGF-β combined with GPS group. The concentrations of inducible nitric oxide synthase (iNOS) and arginase 1 (Arg1) in the cell supernatant, as well as cell proliferation, invasion ability, and the expressions of α-SMA and COL1A1 were detected. Bioinformatics analysis was performed to identify the target intersections of GPS, hepatic fibrosis, and macrophage-related genes. Drug affinity responsive target stability (DARTS) experiments and Western blot were used to verify the regulatory effect of GPS on MIF. Furthermore, LX-2 cells were divided into control group, TGF-β group, TGF-β combined with M2-CM group, TGF-β and oe-NC combined with M2-CM group, and TGF-β and oe-MIF combined with M2-CM group to analyze the concentrations of iNOS and Arg1 in the cell supernatant, as well as changes in cell proliferation, invasion, and the expressions of α-SMA and COL1A1. LX-2 cells were also divided into control group, TGF-β group, TGF-β combined with oe-NC group, TGF-β combined with oe-MIF group, and TGF-β and oe-MIF combined with GPS group to determine the protein expressions of MIF and SPP1 by Western blot. A rat model of hepatic fibrosis was constructed to explore the potential therapeutic effects of GPS on hepatic fibrosis in vivo. Results Compared with the control group, the proliferation and invasion abilities of LX-2 cells in the TGF-β group were increased, and the protein expressions of α-SMA and COL1A1 were enhanced. GPS intervention inhibited the proliferation and invasion of LX-2 cells under TGF-β conditions and reduced the expressions of α-SMA and COL1A1. Compared with the control group, the concentration of iNOS in the cell supernatant of the TGF-β group was upregulated, while the concentration of Arg1 was decreased. M1-CM treatment further increased the concentration of iNOS, decreased the concentration of Arg1, and promoted cell proliferation and invasion, as well as upregulated the expressions of α-SMA and COL1A1 on the basis of TGF-β intervention. However, GPS could reverse the effects of M1-CM intervention. Bioinformatics analysis revealed that MIF was one of the target intersections of GPS, hepatic fibrosis, and macrophage-related genes, and GPS could target and inhibit its expression. Compared with the TGF-β group, after M2-CM intervention, the concentration of iNOS in the cell supernatant decreased, the concentration of Arg1 increased, the proliferation and invasion abilities of LX-2 cells were reduced, and the expressions of α-SMA and COL1A1 were weakened. However, overexpression of MIF reversed the effects of M2-CM intervention. Western blot results showed that compared with the control group, the protein expressions of MIF and SPP1 were enhanced in the TGF-β group. Overexpression of MIF further enhanced the expressions of MIF and SPP1, while GPS intervention inhibited the expressions of MIF and SPP1. In the animal experiment, GPS intervention treatment alleviated liver injury in rats with hepatic fibrosis and inhibited the expressions of MIF and SPP1, as well as α-SMA and COL1A1 in liver tissue. Conclusion GPS may prevent macrophage-mediated hepatic fibrosis by inhibiting the MIF-SPP1 signaling pathway in hepatic stellate cells.
Hepatic Stellate Cells/metabolism*
;
Signal Transduction/drug effects*
;
Macrophage Migration-Inhibitory Factors/genetics*
;
Liver Cirrhosis/prevention & control*
;
Macrophages/drug effects*
;
Iridoid Glucosides/pharmacology*
;
Humans
;
Cell Proliferation/drug effects*
;
Animals
;
Cell Line
;
Collagen Type I/metabolism*
;
Collagen Type I, alpha 1 Chain
;
Intramolecular Oxidoreductases/genetics*
;
Rats
;
Transforming Growth Factor beta/pharmacology*
;
Actins/metabolism*
4.Leukocyte-specific protein 1 (LSP1): A key regulator of cytoskeletal dynamics and leukocyte function.
Puyuan ZHU ; Jinyi GU ; Yuejun LUO ; Yaming XI
Chinese Journal of Cellular and Molecular Immunology 2025;41(8):750-755
Leukocyte-specific protein 1 (LSP1) is an F-actin binding protein expressed in various leukocytes, including lymphocytes, mononuclear macrophages, and neutrophils. LSP1 is highly conserved across different species. Human LSP1 protein contains 339 amino acids, featuring a Ca2+ binding site in the acidic NH2-terminal region and multiple F-actin binding domains along with phosphorylatable sites in the basic COOH-terminal region. Under Ca2+ regulation, the COOH-terminal domain of LSP1 binds to F-actin to regulate cell movement and signal transduction. Additionally, LSP1 activates the mitogen-activated protein kinase (MAPK) signaling pathway through phosphorylation mediated by protein kinase C (PKC) and MAPK-activated protein kinase-2, thereby regulating leukocyte proliferation and chemotaxis. The main effects of LSP1 on leukocytes are as follows: LSP1 plays important roles in neutrophil and macrophage migration, affecting cell adhesion, polarization and movement. LSP1 also functions in endothelial cells to regulate leukocyte transendothelial migration. In addition, LSP1 regulates macrophage phagocytosis through interaction with myosin 1e. Moreover, LSP1 regulates leukocyte proliferation and differentiation and plays significant roles in the development of leukemia and other tumors. In summary, LSP1 regulates leukocyte morphology, movement and function through interactions with cytoskeletal and signaling proteins. This review provides a comprehensive summary of these aspects.
Humans
;
Leukocytes/cytology*
;
Animals
;
Cytoskeleton/metabolism*
;
Microfilament Proteins/physiology*
;
Cell Movement
;
Signal Transduction
5.Effects of human umbilical cord-derived mesenchymal stem cell therapy for cavernous nerve injury-induced erectile dysfunction in the rat model.
Wei WANG ; Ying LIU ; Zi-Hao ZHOU ; Kun PANG ; Jing-Kai WANG ; Peng-Fei HUAN ; Jing-Ru LU ; Tao ZHU ; Zuo-Bin ZHU ; Cong-Hui HAN
Asian Journal of Andrology 2025;27(4):508-515
Stem cell treatment may enhance erectile dysfunction (ED) in individuals with cavernous nerve injury (CNI). Nevertheless, no investigations have directly ascertained the implications of varying amounts of human umbilical cord-derived mesenchymal stem cells (HUC-MSCs) on ED. We compare the efficacy of three various doses of HUC-MSCs as a therapeutic strategy for ED. Sprague-Dawley rats (total = 175) were randomly allocated into five groups. A total of 35 rats underwent sham surgery and 140 rats endured bilateral CNI and were treated with vehicles or doses of HUC-MSCs (1 × 10 6 cells, 5 × 10 6 cells, and 1 × 10 7 cells in 0.1 ml, respectively). Penile tissues were harvested for histological analysis on 1 day, 3 days, 7 days, 14 days, 28 days, 60 days, and 90 days postsurgery. It was found that varying dosages of HUC-MSCs enhanced the erectile function of rats with bilateral CNI and ED. Moreover, there was no significant disparity in the effectiveness of various dosages of HUC-MSCs. However, the expression of endothelial markers (rat endothelial cell antigen-1 [RECA-1] and endothelial nitric oxide synthase [eNOS]), smooth muscle markers (alpha smooth muscle actin [α-SMA] and desmin), and neural markers (neurofilament [RECA-1] and neurogenic nitric oxide synthase [nNOS]) increased significantly with prolonged treatment time. Masson's staining demonstrated an increased in the smooth muscle cell (SMC)/collagen ratio. Significant changes were detected in the microstructures of various types of cells. In vivo imaging system (IVIS) analysis showed that at the 1 st day, the HUC-MSCs implanted moved to the site of damage. Additionally, the oxidative stress levels were dramatically reduced in the penises of rats administered with HUC-MSCs.
Male
;
Animals
;
Erectile Dysfunction/metabolism*
;
Rats, Sprague-Dawley
;
Mesenchymal Stem Cell Transplantation/methods*
;
Rats
;
Penis/pathology*
;
Humans
;
Disease Models, Animal
;
Umbilical Cord/cytology*
;
Peripheral Nerve Injuries/complications*
;
Mesenchymal Stem Cells
;
Nitric Oxide Synthase Type III/metabolism*
;
Actins/metabolism*
;
Nitric Oxide Synthase Type I/metabolism*
6.Genetic and clinical phenotypic analysis of Usher syndrome-associated gene variants.
Heng ZHAO ; Xiuli MA ; Yanli QU ; Guo LI ; Ken LIN ; Rui HUANG ; Lijuan ZHOU ; Jing MA
Journal of Clinical Otorhinolaryngology Head and Neck Surgery 2025;39(8):736-742
Objective:To investigate the molecular characteristics and clinical heterogeneity of Usher syndrome(USH) -related gene variants in patients with hereditary hearing loss in southwest China, providing a basis for early diagnosis and clinical management. Methods:Thirteen patients from twelve families with hearing loss who attended the Affiliated Children's Hospital of Kunming Medical University between January 2017 and March 2021 were enrolled. All patients were identified as carrying USH-related gene variants through next-generation sequencing. Sanger sequencing was performed for all patients and their parents to validate the pathogenic variants. Comprehensive clinical evaluations, including medical history collection, otologic and ophthalmologic examinations, and vestibular function assessments, were conducted. Results:Among the 13 patients, 4 were diagnosed with USH type 1 and 2 with USH type 2. A total of 19 pathogenic or likely pathogenic variants were detected in USH-related genes, including MYO7A,CDH23,USH1C, and USH2A. The causative gene was MYO7A in 3 probands, CDH23 in 5, USH1C in 3, and USH2Ain 2. All patients exhibited an autosomal recessive inheritance pattern. Vestibular dysfunction was observed in 4 patients, and retinitis pigmentosa(RP) in 3 patients. Based on the genotype-phenotype correlation, 6 patients were initially diagnosed with USH, while 7 were classified as having non-syndromic hearing loss(NSHL). Conclusion:This study revealed the clinical heterogeneity of USH-related gene variants in patients with hereditary deafness in southwest China. Although the clinical manifestations of USH are complex and there are overlapping characteristics between different subtypes, genetic testing provides an important basis for early diagnosis and precise clinical management. Especially for those with typical hearing loss, early genetic diagnosis can provide a window of time for early detection and intervention of retinitis pigmentosa.
Humans
;
Usher Syndromes/genetics*
;
Myosin VIIa
;
Phenotype
;
Male
;
Female
;
Myosins/genetics*
;
Mutation
;
Cadherins/genetics*
;
Child
;
Extracellular Matrix Proteins/genetics*
;
Adolescent
;
Pedigree
;
High-Throughput Nucleotide Sequencing
;
Cadherin Related Proteins
;
Cytoskeletal Proteins
;
Cell Cycle Proteins
7.Villin-like protein VILL suppresses proliferation of nasopharyngeal carcinoma cells by interacting with LMO7 protein.
Yumei ZENG ; Jike LI ; Zhongxi HUANG ; Yibo ZHOU
Journal of Southern Medical University 2025;45(5):954-961
OBJECTIVES:
To elucidate the molecular mechanism by which villin-like protein VILL (VILL) inhibits proliferation of nasopharyngeal carcinoma (NPC) cells.
METHODS:
Co-immunoprecipitation (CO-IP) assay, mass spectrometry, Western blotting, immunofluorescence staining, and GST pull-down assay were employed to identify and confirm the protein interacting with VILL that had the highest abundance in NPC cell lines. Transgenic experiments were conducted in both NPC cell lines and nude mice to validate the regulatory role of VILL and its target protein in NPC proliferation. Immunohistochemistry was utilized to assess the correlation of the expression levels of VILL and its target protein in clinical tissue specimens of NPC with the clinical features of the patients.
RESULTS:
In NPC cell lines (HONE1 EBV and S18), VILL was found to interact most abundantly with the E3 ubiquitin ligase LMO7, and both proteins co-localized in the cytoplasm with direct interactions. Overexpression of LMO7 partially counteracted the inhibitory effect of VILL on NPC cell proliferation. The expression of VILL was significantly downregulated in 136 NPC tissue samples compared to 67 non-cancerous nasopharyngeal tissues (P<0.00001) with close correlation with clinical T stage (P=0.04), N stage (P=0.01), and M stage (P=0.013), whereas LMO7 was highly expressed in all the NPC tissues.
CONCLUSIONS
VILL overexpression inhibits NPC proliferation probably by suppressing the oncogenic function of LMO7.
Nasopharyngeal Neoplasms/metabolism*
;
Humans
;
LIM Domain Proteins/metabolism*
;
Cell Proliferation
;
Cell Line, Tumor
;
Animals
;
Mice
;
Nasopharyngeal Carcinoma
;
Mice, Nude
;
Transcription Factors/metabolism*
;
Carcinoma
;
Female
;
Microfilament Proteins/genetics*
;
Male
;
Middle Aged
8.The Medial Prefrontal Cortex-Basolateral Amygdala Circuit Mediates Anxiety in Shank3 InsG3680 Knock-in Mice.
Jiabin FENG ; Xiaojun WANG ; Meidie PAN ; Chen-Xi LI ; Zhe ZHANG ; Meng SUN ; Tailin LIAO ; Ziyi WANG ; Jianhong LUO ; Lei SHI ; Yu-Jing CHEN ; Hai-Feng LI ; Junyu XU
Neuroscience Bulletin 2025;41(1):77-92
Anxiety disorder is a major symptom of autism spectrum disorder (ASD) with a comorbidity rate of ~40%. However, the neural mechanisms of the emergence of anxiety in ASD remain unclear. In our study, we found that hyperactivity of basolateral amygdala (BLA) pyramidal neurons (PNs) in Shank3 InsG3680 knock-in (InsG3680+/+) mice is involved in the development of anxiety. Electrophysiological results also showed increased excitatory input and decreased inhibitory input in BLA PNs. Chemogenetic inhibition of the excitability of PNs in the BLA rescued the anxiety phenotype of InsG3680+/+ mice. Further study found that the diminished control of the BLA by medial prefrontal cortex (mPFC) and optogenetic activation of the mPFC-BLA pathway also had a rescue effect, which increased the feedforward inhibition of the BLA. Taken together, our results suggest that hyperactivity of the BLA and alteration of the mPFC-BLA circuitry are involved in anxiety in InsG3680+/+ mice.
Animals
;
Prefrontal Cortex/metabolism*
;
Basolateral Nuclear Complex/metabolism*
;
Mice
;
Anxiety/metabolism*
;
Nerve Tissue Proteins/genetics*
;
Male
;
Gene Knock-In Techniques
;
Pyramidal Cells/physiology*
;
Mice, Transgenic
;
Neural Pathways/physiopathology*
;
Mice, Inbred C57BL
;
Microfilament Proteins
9.Deciphering the Role of Shank3 in Dendritic Morphology and Synaptic Function Across Postnatal Developmental Stages in the Shank3B KO Mouse.
Jing YANG ; Guaiguai MA ; Xiaohui DU ; Jinyi XIE ; Mengmeng WANG ; Wenting WANG ; Baolin GUO ; Shengxi WU
Neuroscience Bulletin 2025;41(4):583-599
Autism Spectrum Disorder (ASD) is marked by early-onset neurodevelopmental anomalies, yet the temporal dynamics of genetic contributions to these processes remain insufficiently understood. This study aimed to elucidate the role of the Shank3 gene, known to be associated with monogenic causes of autism, in early developmental processes to inform the timing and mechanisms for potential interventions for ASD. Utilizing the Shank3B knockout (KO) mouse model, we examined Shank3 expression and its impact on neuronal maturation through Golgi staining for dendritic morphology and electrophysiological recordings to measure synaptic function in the anterior cingulate cortex (ACC) across different postnatal stages. Our longitudinal analysis revealed that, while Shank3B KO mice displayed normal neuronal morphology at one week postnatal, significant impairments in dendritic growth and synaptic activity emerged by two to three weeks. These findings highlight the critical developmental window during which Shank3 is essential for neuronal and synaptic maturation in the ACC.
Animals
;
Nerve Tissue Proteins/metabolism*
;
Mice, Knockout
;
Dendrites/metabolism*
;
Mice
;
Synapses/metabolism*
;
Gyrus Cinguli/metabolism*
;
Male
;
Mice, Inbred C57BL
;
Autism Spectrum Disorder/genetics*
;
Microfilament Proteins
10.Recurrent eosinophilia with a novel homozygous ARPC1B mutation.
Gamze SONMEZ ; Baris ULUM ; Ates Kutay TENEKECI ; Canan CAKA ; Ali ŞAHIN ; Alp KAZANCIOĞLU ; Begum OZBEK ; İsmail YAZ ; Saliha ESENBOĞA ; Deniz ÇAĞDAŞ
Frontiers of Medicine 2025;19(1):174-180
Cytoskeletal network dysregulation is a pivotal determinant in various immunodeficiencies and autoinflammatory conditions. This report reviews the significance of actin remodeling in disease pathogenesis, focusing on the Arp2/3 complex and its regulatory subunit actin related protein 2/3 complex subunit 1B (ARPC1B). A spectrum of cellular dysfunctions associated with ARPC1B deficiency, impacting diverse immune cell types, is elucidated. The study presents a patient featuring recurrent and persistent eosinophilia attributed to homozygous ARPC1B mutation alongside concomitant compound heterozygous cystic fibrosis transmembrane conductance regulator (CFTR) gene mutations. We used ARPC1B antibody to stain the patient's peripheral blood lymphocytes and those of the control. The defect in the ARPC1B gene in the present patient caused absent/low expression by immunofluorescence microscopy. The intricate interplay between cytoskeletal defects and immunological manifestations underscores the complexity of disease phenotypes, warranting further exploration for targeted therapeutic strategies.
Humans
;
Actin-Related Protein 2-3 Complex/genetics*
;
Cystic Fibrosis Transmembrane Conductance Regulator/genetics*
;
Eosinophilia/genetics*
;
Homozygote
;
Mutation
;
Recurrence

Result Analysis
Print
Save
E-mail