1.Genetic and phenotypic analysis of <i>MYO15Ai> rare variants associated with autosomal recessive hearing loss.
Journal of Clinical Otorhinolaryngology Head and Neck Surgery 2024;38(1):38-43
Objective:To analyze the phenotype and genotype characteristics of autosomal recessive hearing loss caused by <i>MYO15Ai> gene variants, and to provide genetic diagnosis and genetic counseling for patients and their families. Methods:Identification of <i>MYO15Ai> gene variants by next generation sequencing in two sporadic cases of hearing loss at Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine. The sequence variants were verified by Sanger sequencing.The pathogenicity of these variants was determined according to the American College of Medical Genetics and Genomics(ACMG) variant classification guidelines, in conjuction with clinical data. Results:The probands of the two families have bilateral,severe or complete hearing loss.Four variants of <i>MYO15Ai> were identified, including one pathogenic variant that has been reported, two likely pathogenic variants,and one splicing variant of uncertain significance. Patient I carries c. 3524dupA(p. Ser1176Valfs*14), a reported pathogenic variant, and a splicing variant c. 10082+3G>A of uncertain significance according to the ACMG guidelines. Patient I was treated with bilateral hearing aids with satisfactory effect, demonstrated average hearing thresholds of 37.5 dB in the right ear and 33.75 dB in the left ear. Patient Ⅱ carries c. 7441_7442del(p. Leu2481Glufs*86) and c. 10250_10252del(p. Ser3417del),a pair of as likely pathogenic variants according to the ACMG guidelines. Patient Ⅱ, who underwent right cochlear implantation eight years ago, achieved scores of 9 on the Categorical Auditory Performance-Ⅱ(CAP-Ⅱ) and 5 on the Speech Intelligibility Rating(SIR). Conclusion:This study's discovery of the rare c. 7441_7442del variant and the splicing variant c. 10082+3G>A in the <i>MYO15Ai> gene is closely associated with autosomal recessive hearing loss, expanding the <i>MYO15Ai> variant spectrum. Additionally, the pathogenicity assessment of the splicing variant facilitates classification of splicing variations.
Humans
;
Pedigree
;
China
;
Deafness/genetics*
;
Hearing Loss/genetics*
;
Phenotype
;
Hearing Loss, Sensorineural/genetics*
;
Mutation
;
Myosins/genetics*
2.Non-muscle myosin heavy chain 9 gene-related disorders with thrombocytopenia: report of two pedigrees and literature review.
Shu Ting MAO ; Bai LI ; Dao WANG ; Shan Shan LIU ; Shu Fang SU ; Lin Lin WEI ; Fang Yuan CHAI ; Ying LIU ; Yu Feng LIU
Chinese Journal of Pediatrics 2023;61(9):833-838
Objective: To summarize the clinical characteristics and gene variants of 2 pedigrees of non-muscle myosin heavy chain 9 related diseases (MYH9-RD) in children. Methods: The basic information, clinical features, gene variants and laboratory tests of MYH9-RD patients from 2 pedigrees confirmed in the First Affiliated Hospital of Zhengzhou University in November 2021 and July 2022 were analyzed retrospectively. "Non-muscle myosin heavy chain 9 related disease" "MYH9" and "children" were used as key words to search at Pubmed database, CNKI and Wanfang database up to February 2023. The MYH9-RD gene variant spectrum and clinical data were analyzed and summarized. Results: Proband 1 (male, 11 years old) sought medical attention due to epistaxis, the eldest sister and second sister of proband 1 only showed excessive menstrual bleeding, the skin and mucous membrane of the their mother were prone to ecchymosis after bumping, the uncle of proband 1 had kidney damage, and the maternal grandmother and maternal great-grandmother of proband 1 had a history of cataracts. There were 7 cases of phenotypic abnormalities in this pedigree. High-throughput sequencing showed that the proband 1 MYH9 gene had c.279C>G (p.N93K) missense variant, and family verification analysis showed that the variant was inherited from the mother. A total of 4 patients including proband 1 and family members were diagnosed with MYH9-RD. The proband 2 (female, 1 year old) sought medical attention duo to fever and cough, and the father's physical examination revealed thrombocytopenia. There were 2 cases of phenotypic abnormalities in this pedigree. High-throughput sequencing showed that there was a c.4270G>A (p.D1424N) missense variant in the proband 2 MYH9 gene, and family verification analysis showed that the variant was inherited from the father. A total of 2 patients including proband 2 and his father were diagnosed with MYH9-RD. A total of 99 articles were retrieved, including 32 domestic literatures and 67 foreign literatures. The MYH9-RD cases totaled 149 pedigrees and 197 sporadic patients, including 2 pedigrees in our study. There were 101 cases with complete clinical data, including 62 sporadic cases and 39 pedigrees. There were 56 males and 45 females, with an average age of 6.9 years old. The main clinical manifestations were thrombocytopenia, skin ecchymosis, and epistaxis. Most patients didn't receive special treatment after diagnosis. Six English literatures related to MYH9-RD caused by c.279C>G mutation in MYH9 gene were retrieved. Italy reported the highest number of cases (3 cases). Twelve literatures related to MYH9-RD caused by c.4270G>A mutation in MYH9 gene were retrieved. China reported the highest number of cases (9 cases). Conclusions: The clinical manifestations of patients in the MYH9-RD pedigrees varied greatly. MYH9 gene c.279C>G and c.4270G>A mutations are the cause of MYH9-RD.
Infant
;
Humans
;
Female
;
Male
;
Child
;
Myosin Heavy Chains/genetics*
;
Ecchymosis
;
Epistaxis
;
Pedigree
;
Retrospective Studies
;
Muscular Diseases
;
Thrombocytopenia
;
Cytoskeletal Proteins
3.High expression of MYH9 inhibits apoptosis of non-small cell lung cancer cells through activating the AKT/c-Myc pathway.
Fang LIU ; Lanzhu PENG ; Jingle XI
Journal of Southern Medical University 2023;43(4):527-536
OBJECTIVE:
To investigate the role of myosin heavy chain 9 (MYH9) in regulation of cell proliferation, apoptosis, and cisplatin sensitivity of non-small cell lung cancer (NSCLC).
METHODS:
Six NSCLC cell lines (A549, H1299, H1975, SPCA1, H322, and H460) and a normal bronchial epithelial cell line (16HBE) were examined for MYH9 expression using Western blotting. Immunohistochemical staining was used to detect MYH9 expression in a tissue microarray containing 49 NSCLC and 43 adjacent tissue specimens. MYH9 knockout cell models were established in H1299 and H1975 cells using CRISPR/Cas9 technology, and the changes in cell proliferation cell were assessed using cell counting kit-8 (CCK8) and clone formation assays; Western blotting and flow cytometry were used to detect apoptosis of the cell models, and cisplatin sensitivity of the cells was evaluated using IC50 assay. The growth of tumor xenografts derived from NSCLC with or without MYH9 knockout was observed in nude mice.
RESULTS:
MYH9 expression was significantly upregulated in NSCLC (<i>Pi> < 0.001), and the patients with high MYH9 expression had a significantly shorter survival time (<i>Pi>=0.023). In cultured NSCLC cells, MYH9 knockout obviously inhibited cell proliferation (<i>Pi> < 0.001), promoted cell apoptosis (<i>Pi> < 0.05), and increased their chemosensitivity of cisplatin. In the tumor-bearing mouse models, the NSCLC cells with MYH9 knockout showed a significantly lower growth rate (<i>Pi> < 0.05). Western blotting showed that MYH9 knockout inactivated the AKT/c- Myc axis (<i>Pi> < 0.05) to inhibit the expression of BCL2- like protein 1 (<i>Pi> < 0.05), promoted the expression of BH3- interacting domain death agonist and the apoptosis regulator BAX (<i>Pi> < 0.05), and activated apoptosis-related proteins caspase-3 and caspase-9 (<i>Pi> < 0.05).
CONCLUSION
High expression of MYH9 contributes to NSCLC progression by inhibiting cell apoptosis <i>viai> activating the AKT/c-Myc axis.
Animals
;
Humans
;
Mice
;
Apoptosis
;
Carcinoma, Non-Small-Cell Lung/metabolism*
;
Cell Line, Tumor
;
Cell Proliferation
;
Cisplatin/pharmacology*
;
Cytoskeletal Proteins/metabolism*
;
Lung Neoplasms/metabolism*
;
Mice, Nude
;
Myosin Heavy Chains/metabolism*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Signal Transduction
4.Recent research on childhood hypertrophic cardiomyopathy caused by <i>MYH7i> gene mutations.
Kui ZHENG ; Lu LIU ; Ying-Qian ZHANG
Chinese Journal of Contemporary Pediatrics 2023;25(4):425-430
Hypertrophic cardiomyopathy (HCM) is the most common monogenic inherited myocardial disease in children, and mutations in sarcomere genes (such as <i>MYH7i> and <i>MYBPC3i>) are the most common genetic etiology of HCM, among which mutations in the <i>MYH7i> gene are the most common and account for 30%-50%. <i>MYH7i> gene mutations have the characteristics of being affected by environmental factors, coexisting with multiple genetic variations, and age-dependent penetrance, which leads to different or overlapping clinical phenotypes in children, including various cardiomyopathies and skeletal myopathies. At present, the pathogenesis, course, and prognosis of HCM caused by <i>MYH7i> gene mutations in children remain unclear. This article summarizes the possible pathogenesis, clinical phenotype, and treatment of HCM caused by <i>MYH7i> gene mutations, in order to facilitate the accurate prognostic evaluation and individualized management and treatment of the children with this disorder.
Child
;
Humans
;
Cardiomyopathy, Hypertrophic/therapy*
;
Phenotype
;
Troponin T/genetics*
;
Mutation
;
Carrier Proteins/genetics*
;
Myosin Heavy Chains/genetics*
;
Cardiac Myosins/genetics*
5.Phenotype and genotype characteristics of children with cardiomyopathy associated with <i>MYH7i> gene mutation: a retrospective analysis.
Lu LIU ; Kui ZHENG ; Ying-Qian ZHANG
Chinese Journal of Contemporary Pediatrics 2023;25(11):1156-1160
OBJECTIVES:
To investigate the clinical phenotype and genotype characteristics of children withcardiomyopathy (CM) associated with <i>MYH7i> gene mutation.
METHODS:
A retrospective analysis was conducted on the medical data of five children with CM caused by <i>MYH7i> gene mutation who were diagnosed and treated in the Department of Cardiology, Hebei Children's Hospital.
RESULTS:
Among the five children with CM, there were three girls and two boys, all of whom carried <i>MYH7i> gene mutation. Seven mutation sites were identified, among which five were not reported before. Among the five children, there were three children with hypertrophic cardiomyopathy, one child with dilated cardiomyopathy, and one child with noncompaction cardiomyopathy. The age ranged from 6 to 156 months at the initial diagnosis. At the initial diagnosis, two children had the manifestations of heart failure such as cough, shortness of breath, poor feeding, and cyanosis of lips, as well as delayed development; one child had palpitation, blackness, and syncope; one child had fever, runny nose, and abnormal liver function; all five children had a reduction in activity endurance. All five children received pharmacotherapy for improving cardiac function and survived after follow-up for 7-24 months.
CONCLUSIONS
The age of onset varies in children with CM caused by <i>MYH7i> gene mutation, and most children lack specific clinical manifestations at the initial diagnosis and may have the phenotype of hypertrophic cardiomyopathy, dilated cardiomyopathy or noncompaction cardiomyopathy. The children receiving early genetic diagnosis and pharmacological intervention result in a favorable short-term prognosis.
Male
;
Female
;
Child
;
Humans
;
Retrospective Studies
;
Cardiomyopathy, Dilated/genetics*
;
Pedigree
;
Phenotype
;
Genotype
;
Mutation
;
Cardiomyopathy, Hypertrophic/diagnosis*
;
Myosin Heavy Chains/genetics*
;
Cardiac Myosins/genetics*
6.Clinical phenotype and genetic analysis of patients with left ventricular noncompaction caused by the biallelic mutation of MYBPC3 and MYH7.
Ya Hui ZHANG ; Xiao Yan LI ; Bang Rong SONG ; Yue Li WANG ; Jun Rui ZHANG ; Yan Long REN
Chinese Journal of Cardiology 2023;51(11):1160-1165
Objective: To explore the relationship between pathogenic gene, mutation and phenotype of left ventricular noncompaction (LVNC) patients and their family members. Methods: The subjects were the proband with LVNC and her family members. The medical history including electrocardiogram, echocardiography and cardiac magnetic resonance examination of the proband and family members were collected. Whole exome sequencing of the proband was performed, bioinformatics analysis focused on the genes related to hereditary cardiomyopathy. Candidate pathogenic sites were validated by Sanger sequencing. The clinical interpretation of sequence variants were classified according to American College of Medical Genetics and Genomics (ACMG) guidelines. Results: The proband carried a heterozygous variation of the MYBPC3 gene c.C2827T and the MYH7 gene c.G2221C. The proband's sister carried heterozygous variation of MYBPC3 gene c.C2827T. According to the ACMG guidelines, the variant was determined to be pathogenic. Conclusion: The missense variant of MYBPC3 gene c.C2827T and MYH7 gene c.G2221C are identified from the proband with LVNC and her family member, which provides a genetic basis for clinical diagnosis and genetic counseling of the patients and the family members with LVNC.
Female
;
Humans
;
Cardiac Myosins/genetics*
;
Heart Defects, Congenital
;
Mutation
;
Mutation, Missense
;
Myosin Heavy Chains/genetics*
;
Pedigree
;
Phenotype
7.Clinical phenotype and genetic analysis of patients with left ventricular noncompaction caused by the biallelic mutation of MYBPC3 and MYH7.
Ya Hui ZHANG ; Xiao Yan LI ; Bang Rong SONG ; Yue Li WANG ; Jun Rui ZHANG ; Yan Long REN
Chinese Journal of Cardiology 2023;51(11):1160-1165
Objective: To explore the relationship between pathogenic gene, mutation and phenotype of left ventricular noncompaction (LVNC) patients and their family members. Methods: The subjects were the proband with LVNC and her family members. The medical history including electrocardiogram, echocardiography and cardiac magnetic resonance examination of the proband and family members were collected. Whole exome sequencing of the proband was performed, bioinformatics analysis focused on the genes related to hereditary cardiomyopathy. Candidate pathogenic sites were validated by Sanger sequencing. The clinical interpretation of sequence variants were classified according to American College of Medical Genetics and Genomics (ACMG) guidelines. Results: The proband carried a heterozygous variation of the MYBPC3 gene c.C2827T and the MYH7 gene c.G2221C. The proband's sister carried heterozygous variation of MYBPC3 gene c.C2827T. According to the ACMG guidelines, the variant was determined to be pathogenic. Conclusion: The missense variant of MYBPC3 gene c.C2827T and MYH7 gene c.G2221C are identified from the proband with LVNC and her family member, which provides a genetic basis for clinical diagnosis and genetic counseling of the patients and the family members with LVNC.
Female
;
Humans
;
Cardiac Myosins/genetics*
;
Heart Defects, Congenital
;
Mutation
;
Mutation, Missense
;
Myosin Heavy Chains/genetics*
;
Pedigree
;
Phenotype
8.Clinical Characteristics and Prognosis of Acute Myeloid Leukemia Patients with inv(16)/t(16;16)(p13.1;q22) and/or CBFβ-MYH11.
Ye-Min WANG ; Ping CAI ; Mei-Jia ZHOU ; Ying-Ying GONG ; Jin-Lan PAN ; Jian-Nong CEN ; Xiao-Fei YANG ; Su-Ning CHEN
Journal of Experimental Hematology 2022;30(2):367-372
OBJECTIVE:
To summarize the clinical and laboratory characteristics of patients with acute myeloid leukemia (AML) with inv(16)/t(16;16) (p13.1;q22), and to analyze the risk factors affecting the prognosis of the patients.
METHODS:
AML patients with inv(16)/t(16;16) (p13.1;q22) and/or CBFβ-MYH11+ admitted to the Department of Hematology, The First Affiliated Hospital of Soochow University from January 1, 2008 to October 30, 2019 were retrospective analyzed, the clinical and laboratory indicators, as well as treatment plans and efficacy evaluations of the patients were all recorded. Furthermore, related factors affecting the overall survival (OS) and event-free survival (EFS) of the patients were analyzed.
RESULTS:
Among 151 AML patients with inv(16)/t(16;16) (p13.1;q22) and/or CBFβ-MYH11+, the percentage of additional chromosomal abnormalities was about 27.8%, and the most common additional chromosomal abnormality was +22 (33/151, 21.8%), followed by +8 (11/151, 7.3%). There were 112 patients with perfect NGS examination, and the result showed the most common accompanying gene mutations were KIT mutation (34/112, 30.4%) and FLT3 mutation (23/112, 20.5%). Univariate analysis showed that factors affecting EFS included: NE≤0.5×109/L (P=0.006) and combined K-RAS mutation (P=0.002); Factors affecting OS included: Age≥50 years old (P<0.001) and NE≤0.5×109/L (P=0.016). Multivariate analysis showed that NE≤0.5×109/L (P=0.019) was the risk factors affecting OS. The proportion of bone marrow eosinophilia (BME)≥10.00% (P=0.029) was the risk factors affecting EFS.
CONCLUSION
The prognosis for those newly diagnosed AML patients who were of advanced age, the high proportion of bone marrow eosinophils, K-RAS mutations, and agranulocytosis is poor. The treatment plans can be adjusted in the early stage to improve the prognosis of such patients.
Chromosome Inversion
;
Humans
;
Leukemia, Myeloid, Acute/genetics*
;
Middle Aged
;
Myosin Heavy Chains/genetics*
;
Oncogene Proteins, Fusion
;
Prognosis
;
Retrospective Studies
9.Effects of non-muscle myosin Ⅱ silenced bone marrow-derived mesenchymal stem cells transplantation on lung extracellular matrix in rats after endotoxin/lipopolysaccharide-induced acute lung injury.
Xi YIN ; Wan Fang ZHOU ; Wen Jia HOU ; Ming Zhi FAN ; Guo Sheng WU ; Xiao Bin LIU ; Qi Min MA ; Yu Song WANG ; Feng ZHU
Chinese Journal of Burns 2022;38(5):422-433
Objective: To investigate the effects of non-muscle myosin Ⅱ (<i>NMⅡi>) gene silenced bone marrow-derived mesenchymal stem cells (BMMSCs) on pulmonary extracellular matrix (ECM) and fibrosis in rats with acute lung injury (ALI) induced by endotoxin/lipopolysaccharide (LPS). Methods: The experimental research methods were adopted. Cells from femur and tibial bone marrow cavity of four one-week-old male Sprague-Dawley rats were identified as BMMSCs by flow cytometry, and the third passage of BMMSCs were used in the following experiments. The cells were divided into NMⅡ silenced group transfected with pHBLV-U6-ZsGreen-Puro plasmid containing small interference RNA sequence of <i>NMⅡi> gene, vector group transfected with empty plasmid, and blank control group without any treatment, and the protein expression of NMⅡ at 72 h after intervention was detected by Western blotting (<i>ni>=3). The morphology of cells was observed by an inverted phase contrast microscope and cells labeled with chloromethylbenzoine (CM-DiⅠ) in vitro were observed by an inverted fluorescence microscope. Twenty 4-week-old male Sprague-Dawley rats were divided into blank control group, ALI alone group, ALI+BMMSC group, and ALI+NMⅡ silenced BMMSC group according to the random number table, with 5 rats in each group. Rats in blank control group were not treated, and rats in the other 3 groups were given LPS to induce ALI. Immediately after modeling, rats in ALI alone group were injected with 1 mL normal saline via tail vein, rats in ALI+BMMSC group and ALI+NMⅡ silenced BMMSC group were injected with 1×107/mL BMMSCs and <i>NMⅡi> gene silenced BMMSCs of 1 mL labelled with CM-DiⅠ via tail vein, and rats in blank control group were injected with 1 mL normal saline via tail vein at the same time point, respectively. At 24 h after intervention, the lung tissue was collected to observe intrapulmonary homing of the BMMSCs by an inverted fluorescence microscope. Lung tissue was collected at 24 h, in 1 week, and in 2 weeks after intervention to observe pulmonary inflammation by hematoxylin eosin staining and to observe pulmonary fibrosis by Masson staining, and the pulmonary fibrosis in 2 weeks after intervention was scored by modified Ashcroft score (<i>ni>=5). The content of α-smooth muscle actin (α-SMA), matrix metalloproteinase 2 (MMP-2), and MMP-9 was detected by immunohistochemistry in 2 weeks after intervention (<i>ni>=3), the activity of superoxide dismutase (SOD), malondialdehyde, myeloperoxidase (MPO) was detected by enzyme-linked immunosorbent assay at 24 h after intervention (<i>ni>=3), and the protein expressions of CD11b and epidermal growth factor like module containing mucin like hormone receptor 1 (EMR1) in 1 week after intervention were detected by immunofluorescence staining (<i>ni>=3). Data were statistically analyzed with one-way analysis of variance, Bonferroni method, and Kruskal-Wallis <i>Hi> test. Results: At 72 h after intervention, the NMⅡprotein expression of cells in NMⅡ silenced group was significantly lower than those in blank control group and vector group (with <i>Pi> values <0.01). BMMSCs were in long spindle shape and grew in cluster shaped like vortexes, which were labelled with CM-DiⅠ successfully in vitro. At 24 h after intervention, cell homing in lung of rats in ALI+NMⅡ silenced BMMSC group was more pronounced than that in ALI+BMMSC group, while no CM-DiⅠ-labelled BMMSCs were observed in lung of rats in blank control group and ALI alone group. There was no obvious inflammatory cell infiltration in lung tissue of rats in blank control group at all time points, while inflammatory cell infiltration in lung tissue of rats in ALI+BMMSC group and ALI+NMⅡ silenced BMMSC group was significantly less than that in ALI alone group at 24 h after intervention, and alveolar wall turned to be thinner and a small amount of congestion in local lung tissue appeared in rats of the two groups in 1 week and 2 weeks after intervention. In 1 week and 2 weeks after intervention, collagen fiber deposition in lung tissue of rats in ALI alone group, ALI+BMMSC group, and ALI+NMⅡ silenced BMMSC group was significantly aggravated compared with that in blank control group, while collagen fiber deposition in lung tissue of rats in ALI+BMMSC group and ALI+NMⅡ silenced BMMSC group was significantly improved compared with that in ALI alone group. In 2 weeks after intervention, modified Ashcroft scores for pulmonary fibrosis of rats in ALI alone group, ALI+BMMSC group, and ALI+NMⅡ silenced BMMSC group were 2.36±0.22, 1.62±0.16, 1.06±0.26, respectively, significantly higher than 0.30±0.21 in blank control group (<i>Pi><0.01). Modified Ashcroft scores for pulmonary fibrosis of rats in ALI+BMMSC group and ALI+NMⅡ silenced BMMSC group were significantly lower than that in ALI alone group (<i>Pi><0.01), and modified Ashcroft score for pulmonary fibrosis of rats in ALI+NMⅡ silenced BMMSC group was significantly lower than that in ALI+BMMSC group (<i>Pi><0.01). In 2 weeks after intervention, the content of α-SMA in lung tissue of rats in ALI+BMMSC group and ALI+NMⅡ silenced BMMSC group were significantly decreased compared with that in ALI alone group (<i>Pi><0.05 or <i>Pi><0.01). The content of MMP-2 in lung tissue of rats in the 4 groups was similar (<i>Pi>>0.05). The content of MMP-9 in lung tissue of rats in ALI alone group was significantly increased compared with that in blank control group (<i>Pi><0.01), and the content of MMP-9 in lung tissue of rats in ALI+BMMSC group and ALI+NMⅡ silenced BMMSC group was significantly decreased compared with that in ALI alone group (<i>Pi><0.01). At 24 h after intervention, the activity of malondialdehyde, SOD, and MPO in lung tissue of rats in ALI alone group, ALI+BMMSC group, and ALI+NMⅡ silenced BMMSC group were significantly increased compared with that in blank control group (<i>Pi><0.01), the activity of malondialdehyde in lung tissue of rats in ALI+NMⅡ silenced BMMSC group and the activity of SOD in lung tissue of rats in ALI+BMMSC group and ALI+NMⅡ silenced BMMSC group were significantly increased compared with that in ALI alone group (<i>Pi><0.05 or <i>Pi><0.01), and the activity of SOD in lung tissue of rats in ALI+NMⅡ silenced BMMSC group was significantly decreased compared with that in ALI+BMMSC group (<i>Pi><0.01). The activity of MPO in lung tissue of rats in ALI+BMMSC group and ALI+NMⅡ silenced BMMSC group was significantly decreased compared with that in ALI alone group (<i>Pi><0.01), and the activity of MPO in lung tissue of rats in ALI+NMⅡ silenced BMMSC group was significantly decreased compared with that in ALI+BMMSC group (<i>Pi><0.01). In 1 week after intervention, the protein expression of CD11b in lung tissue of rats in ALI+NMⅡ silenced BMMSC group was significantly increased compared with those in the other three groups (<i>Pi><0.05 or <i>Pi><0.01), while the protein expressions of EMR1 in lung tissue of rats in the four groups were similar (<i>Pi>>0.05). Conclusions: Transplantation of <i>NMⅡi> gene silenced BMMSCs can significantly improve the activity of ECM components in the lung tissue in LPS-induced ALI rats, remodel its integrity, and enhance its antioxidant capacity, and alleviate lung injury and pulmonary fibrosis.
Acute Lung Injury/therapy*
;
Animals
;
Bone Marrow
;
Collagen/metabolism*
;
Endotoxins
;
Extracellular Matrix
;
Lipopolysaccharides/adverse effects*
;
Lung
;
Male
;
Malondialdehyde/metabolism*
;
Matrix Metalloproteinase 2/metabolism*
;
Matrix Metalloproteinase 9/metabolism*
;
Mesenchymal Stem Cells/metabolism*
;
Myosin Type II/metabolism*
;
Pulmonary Fibrosis
;
Rats
;
Rats, Sprague-Dawley
;
Saline Solution/metabolism*
;
Superoxide Dismutase/metabolism*
10.Analysis of phenotype and MYH7 gene variant in a family of patients with hypertrophic cardiomyopathy.
Xueli ZHAO ; Bo WANG ; Xiaoli ZHU ; Qianli YANG ; Ying LIU ; Hong SHAO ; Lei ZUO ; Yun LUO ; Yue WANG ; Liwen LIU
Chinese Journal of Medical Genetics 2022;39(8):873-876
OBJECTIVE:
To analyze the clinical phenotype and MYH7 gene variant in a Chinese pedigree affected with hypertrophic cardiomyopathy (HCM).
METHODS:
The proband was screened for variant of 96 cardiomyopathy-associated genes by exonic amplification and high-throughput sequencing. Candidate variant was verified by Sanger sequencing among 300 healthy controls as well as family members of the proband. Co-segregation analysis of genotypes and clinical phenotypes was carried out for the pedigree. Clustal X software was used to analyze the sequence conservation of the variant among various species, and its pathogenicity was predicted by using bioinformatics software.
RESULTS:
6 out of 12 members from this pedigree were found to harbor heterozygous c.4124A>G (p.Tyr1375Cys) variant of the MYH7 gene, among whom five were diagnosed with HCM. The remaining one had failed to meet the diagnostic criteria for HCM, but had abnormal ECG. The same variant was not found in the 300 healthy controls. Amino acid sequence analysis showed that the variant is located in a highly conserved region, and bioinformatics analysis predicted that this variant may affect protein function and has a deleterious effect. Based on the American College of Medical Genetics and Genomics (ACMG) guidelines, the variant was predicted to be likely pathogenic (PM2+ PP1_Moderate+PP3+PP5).
CONCLUSION
The c.4124A>G (p.Tyr1375Cys) variant of the MYH7 gene probably underlay the pathogenesis in this pedigree. Above finding has important value for the early diagnosis of patients with HCM.
Cardiac Myosins/genetics*
;
Cardiomyopathy, Hypertrophic/genetics*
;
Genotype
;
Humans
;
Mutation
;
Myosin Heavy Chains/genetics*
;
Pedigree
;
Phenotype

Result Analysis
Print
Save
E-mail