Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.962
Filter
1.
Nat Commun ; 15(1): 6716, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112473

ABSTRACT

Myosin motors perform many fundamental functions in eukaryotic cells by providing force generation, transport or tethering capacity. Motor activity control within the cell involves on/off switches, however, few examples are known of how myosins regulate speed or processivity and fine-tune their activity to a specific cellular task. Here, we describe a phosphorylation event for myosins of class VI (MYO6) in the motor domain, which accelerates its ATPase activity leading to a 4-fold increase in motor speed determined by actin-gliding assays, single molecule mechanics and stopped flow kinetics. We demonstrate that the serine/threonine kinase DYRK2 phosphorylates MYO6 at S267 in vitro. Single-molecule optical-tweezers studies at low load reveal that S267-phosphorylation results in faster nucleotide-exchange kinetics without change in the working stroke of the motor. The selective increase in stiffness of the acto-MYO6 complex when proceeding load-dependently into the nucleotide-free rigor state demonstrates that S267-phosphorylation turns MYO6 into a stronger motor. Finally, molecular dynamic simulations of the nucleotide-free motor reveal an alternative interaction network within insert-1 upon phosphorylation, suggesting a molecular mechanism, which regulates insert-1 positioning, turning the S267-phosphorylated MYO6 into a faster motor.


Subject(s)
Molecular Dynamics Simulation , Myosin Heavy Chains , Phosphorylation , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Kinetics , Protein Serine-Threonine Kinases/metabolism , Nucleotides/metabolism , Humans , Animals , Protein Domains , Protein-Tyrosine Kinases/metabolism , Actins/metabolism
2.
Physiol Rep ; 12(16): e70012, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39169429

ABSTRACT

Aging is associated with cardiac contractile abnormalities, but the etiology of these contractile deficits is unclear. We hypothesized that cardiac contractile and regulatory protein expression is altered during aging. To investigate this possibility, left ventricular (LV) lysates were prepared from young (6 months) and old (24 months) Fischer344 rats. There are no age-related changes in SERCA2 expression or phospholamban phosphorylation. Additionally, neither titin isoform expression nor phosphorylation differed. However, there is a significant increase in ß-isoform of the myosin heavy chain (MyHC) expression and phosphorylation of TnI and MyBP-C during aging. In permeabilized strips of papillary muscle, force and Ca2+ sensitivity are reduced during aging, consistent with the increase in ß-MyHC expression and TnI phosphorylation. However, the increase in MyBP-C phosphorylation during aging may represent a mechanism to compensate for age-related contractile deficits. In isolated cardiomyocytes loaded with Fura-2, the peak of the Ca2+ transient is reduced, but the kinetics of the Ca2+ transient are not altered. Furthermore, the extent of shortening and the rates of both sarcomere shortening and re-lengthening are reduced. These results demonstrate that aging is associated with changes in contractile and regulatory protein expression and phosphorylation, which affect the mechanical properties of cardiac muscle.


Subject(s)
Aging , Myocardial Contraction , Myocytes, Cardiac , Rats, Inbred F344 , Animals , Male , Myocardial Contraction/physiology , Aging/metabolism , Aging/physiology , Rats , Phosphorylation , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Myosin Heavy Chains/metabolism , Calcium-Binding Proteins/metabolism , Connectin/metabolism , Troponin I/metabolism , Calcium/metabolism , Calmodulin-Binding Proteins/metabolism , Carrier Proteins
3.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125717

ABSTRACT

Acute myeloid leukemia (AML) is the most prevalent type of hematopoietic malignancy. Despite recent therapeutic advancements, the high relapse rate associated with extramedullary involvement remains a challenging issue. Moreover, therapeutic targets that regulate the extramedullary infiltration of AML cells are still not fully elucidated. The Aryl Hydrocarbon Receptor (AHR) is known to influence the progression and migration of solid tumors; however, its role in AML is largely unknown. This study explored the roles of AHR in the invasion and migration of AML cells. We found that suppressed expression of AHR target genes correlated with an elevated relapse rate in AML. Treatment with an AHR agonist on patient-derived AML cells significantly decreased genes associated with leukocyte trans-endothelial migration, cell adhesion, and regulation of the actin cytoskeleton. These results were further confirmed in THP-1 and U937 AML cell lines using AHR agonists (TCDD and FICZ) and inhibitors (SR1 and CH-223191). Treatment with AHR agonists significantly reduced Matrigel invasion, while inhibitors enhanced it, regardless of the Matrigel's stiffness. AHR agonists significantly reduced the migration rate and chemokinesis of both cell lines, but AHR inhibitors enhanced them. Finally, we found that the activity of AHR and the expression of NMIIA are negatively correlated. These findings suggest that AHR activity regulates the invasiveness and motility of AML cells, making AHR a potential therapeutic target for preventing extramedullary infiltration in AML.


Subject(s)
Cell Movement , Leukemia, Myeloid, Acute , Myosin Heavy Chains , Neoplasm Invasiveness , Receptors, Aryl Hydrocarbon , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/agonists , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIA/genetics , Cell Line, Tumor , Female , Male , Gene Expression Regulation, Leukemic , Middle Aged , Aged , THP-1 Cells , U937 Cells , Adult , Basic Helix-Loop-Helix Transcription Factors
4.
FASEB J ; 38(16): e70009, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39158138

ABSTRACT

Skeletal muscle comprises slow and fast myofibers, with slow myofibers excelling in aerobic metabolism and endurance. Quercetin, a polyphenol, is reported to induce slow myofibers in rodent skeletal muscle both in vitro and in vivo. However, its effect on human myofiber types remains unexplored. In this study, we evaluated quercetin's impact on slow myofiber induction using human skeletal muscle satellite cells. In a two-dimensional culture, quercetin enhanced gene expression, contributing to muscle differentiation, and significantly expanded the area of slow-type myosin heavy chain positive cells. It also elevated the gene expression of Pgc1α, an inducer of slow myofibers. Conversely, quercetin did not affect mitochondrial abundance, fission, or fusion, but it did increase the gene expression of Cox7A2L, which aids in promoting mitochondrial supercomplexity and endurance, and Mb, which contributes to oxidative phosphorylation. In a three-dimensional culture, quercetin significantly extended the time to peak tension and half relaxation time of the engineered human skeletal muscle tissues constructed on microdevices. Moreover, quercetin enhanced the muscle endurance of the tissues and curbed the rise in lactate secretion from the exercised tissues. These findings suggest that quercetin may induce slow myofibers in human skeletal muscle.


Subject(s)
Muscle, Skeletal , Quercetin , Quercetin/pharmacology , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/cytology , Tissue Engineering/methods , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Phenotype , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/cytology , Cells, Cultured , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Cell Differentiation/drug effects
5.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39201382

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a cardiovascular pathology that is caused by variants in genes encoding sarcomere-associated proteins. However, the clinical significance of numerous variants in HCM-associated genes is still unknown. CRISPR/Cas9 is a tool of nucleotide sequence editing that allows for the unraveling of different biological tasks. In this study, introducing a mutation with CRISPR/Cas9 into induced pluripotent stem cells (iPSCs) of a healthy donor and the directed differentiation of the isogenic iPSC lines into cardiomyocytes were used to assess the pathogenicity of a variant of unknown significance, p.M659I (c.1977G > A) in MYH7, which was found previously in an HCM patient. Using two single-stranded donor oligonucleotides with and without the p.M659I (c.1977G > A) mutation, together with CRISPR/Cas9, an iPSC line heterozygous at the p.M659I (c.1977G > A) variant in MYH7 was generated. No CRISPR/Cas9 off-target activity was observed. The iPSC line with the introduced p.M659I (c.1977G > A) mutation in MYH7 retained its pluripotent state and normal karyotype. Compared to the isogenic control, cardiomyocytes derived from the iPSCs with the introduced p.M659I (c.1977G > A) mutation in MYH7 recapitulated known HCM features: enlarged size, elevated diastolic calcium level, changes in the expression of HCM-related genes, and disrupted energy metabolism. These findings indicate the pathogenicity of the variant.


Subject(s)
CRISPR-Cas Systems , Cardiac Myosins , Cardiomyopathy, Hypertrophic , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Myosin Heavy Chains , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Humans , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/cytology , Cell Differentiation/genetics , Gene Editing/methods , Mutation , Cell Line
6.
Sci Rep ; 14(1): 19685, 2024 08 24.
Article in English | MEDLINE | ID: mdl-39181964

ABSTRACT

Prostate cancer (PRAD) is one of the leading malignancies in men all around the world. Here, we identified Myosin Heavy Chain 6 (MYH6) as a potential tumor suppressor gene in the development of prostate cancer. We found lower expression of MYH6 in prostate cancer tissues, and its lower gene expression was also associated with worse clinical outcomes. In vitro and in vivo assays indicated that overexpressed MYH6 could suppress the proliferation and migration progression of prostate cancer cells. RNA-seq was employed to investigate the mechanism, and KIT Proto-Oncogen (KIT) was determined as the downstream gene of MYH6, which was further confirmed using rescue assays. In all, we provide the evidence that MYH6 could serve as a tumor suppressor in prostate cancer. Our results highlight the potential role of MYH6 in the development of prostate cancer.


Subject(s)
Cell Movement , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Myosin Heavy Chains , Prostatic Neoplasms , Proto-Oncogene Proteins c-kit , Male , Humans , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Cell Movement/genetics , Animals , Down-Regulation , Mice , Cardiac Myosins
7.
Cell Commun Signal ; 22(1): 417, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39192336

ABSTRACT

Non-muscle myosin heavy chain IIA (MYH9), a member of the non-muscle myosin II (NM II) family, is widely expressed in cells. The interaction of MYH9 with actin in the cytoplasm can hydrolyze ATP, completing the conversion of chemical energy to mechanical motion. MYH9 participates in various cellular processes, such as cell adhesion, migration, movement, and even signal transduction. Mutations in MYH9 are often associated with autosomal dominant platelet disorders and kidney diseases. Over the past decade, tumor-related research has gradually revealed a close relationship between MYH9 and the occurrence and development of tumors. This article provides a review of the research progress on the role of MYH9 in cancer regulation. We also discussed the anti-cancer effects of MYH9 under special circumstances, as well as its regulation of T cell function. In addition, given the importance of MYH9 as a key hub in oncogenic signal transduction, we summarize the current therapeutic strategies targeting MYH9 as well as the ongoing challenges.


Subject(s)
Myosin Heavy Chains , Neoplasms , Humans , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Animals , Signal Transduction , Molecular Motor Proteins/metabolism , Molecular Motor Proteins/genetics
8.
Dis Model Mech ; 17(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39189070

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is an inherited heart muscle disease that is characterised by left ventricular wall thickening, cardiomyocyte disarray and fibrosis, and is associated with arrhythmias, heart failure and sudden death. However, it is unclear to what extent the electrophysiological disturbances that lead to sudden death occur secondary to structural changes in the myocardium or as a result of HCM cardiomyocyte electrophysiology. In this study, we used an induced pluripotent stem cell model of the R403Q variant in myosin heavy chain 7 (MYH7) to study the electrophysiology of HCM cardiomyocytes in electrically coupled syncytia, revealing significant conduction slowing and increased spatial dispersion of repolarisation - both well-established substrates for arrhythmia. Analysis of rhythmonome protein expression in MYH7 R403Q cardiomyocytes showed reduced expression of connexin-43 (also known as GJA1), sodium channels and inward rectifier potassium channels - a three-way hit that reduces electrotonic coupling and slows cardiac conduction. Our data represent a previously unreported, biophysical basis for arrhythmia in HCM that is intrinsic to cardiomyocyte electrophysiology. Later in the progression of the disease, these proarrhythmic phenotypes may be accentuated by myocyte disarray and fibrosis to contribute to sudden death.


Subject(s)
Cardiomyopathy, Hypertrophic , Connexin 43 , Heart Conduction System , Myocytes, Cardiac , Myosin Heavy Chains , Connexin 43/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Humans , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/physiopathology , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Induced Pluripotent Stem Cells/metabolism , Cardiac Myosins/metabolism , Cardiac Myosins/genetics , Giant Cells/metabolism , Giant Cells/pathology , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Action Potentials
9.
Nutrients ; 16(16)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39203737

ABSTRACT

Sialyllactose (SL) is a functional human milk oligosaccharide essential for immune support, brain development, intestinal maturation, and antiviral defense. However, despite its established health benefits, the effect of SL on exercise performance and muscle mass in mice remains unknown. Here, we aimed to investigate, for the first time, the effects of 6'-SL on muscle functions. Seven-week-old male C57BL/6J mice were administered 100 mg/kg 6'-SL for 12 weeks, after which exhaustive treadmill performance was conducted. Moreover, muscle strength was examined by grip strength, and muscle phenotype characteristics such as muscle mass, muscle fiber size, and muscle protein expression were also examined. The administration of 6'-SL significantly improved exhaustive treadmill performance metrics, including distance and exhaustion time. Grip strength was also increased by 6'-SL administration. Additionally, 6'-SL increased muscle mass in both the gastrocnemius (GAS) and soleus. 6'-SL administration led to an increase in the minimum Feret's diameter and the protein expression of total myosin heavy chain in the GAS muscle. In conclusion, 6'-SL administration in vivo led to increased running distance and time by increasing muscle mass and strength. These findings collectively indicate that 6'-SL is a potential agent for improving muscle health and exercise performance.


Subject(s)
Mice, Inbred C57BL , Muscle Strength , Muscle, Skeletal , Physical Conditioning, Animal , Animals , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle Strength/drug effects , Physical Conditioning, Animal/physiology , Mice , Lactose/analogs & derivatives , Lactose/pharmacology , Myosin Heavy Chains/metabolism , Muscle Proteins/metabolism
10.
Physiol Rep ; 12(13): e16052, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987200

ABSTRACT

We previously observed lifelong endurance exercise (LLE) influenced quadriceps whole-muscle and myofiber size in a fiber-type and sex-specific manner. The current follow-up exploratory investigation examined myofiber size regulators and myofiber size distribution in vastus lateralis biopsies from these same LLE men (n = 21, 74 ± 1 years) and women (n = 7, 72 ± 2 years) as well as old, healthy nonexercisers (OH; men: n = 10, 75 ± 1 years; women: n = 10, 75 ± 1 years) and young exercisers (YE; men: n = 10, 25 ± 1 years; women: n = 10, 25 ± 1 years). LLE exercised ~5 days/week, ~7 h/week for the previous 52 ± 1 years. Slow (myosin heavy chain (MHC) I) and fast (MHC IIa) myofiber nuclei/fiber, myonuclear domain, satellite cells/fiber, and satellite cell density were not influenced (p > 0.05) by LLE in men and women. The aging groups had ~50%-60% higher proportion of large (>7000 µm2) and small (<3000 µm2) myofibers (OH; men: 44%, women: 48%, LLE; men: 42%, women: 42%, YE; men: 27%, women: 29%). LLE men had triple the proportion of large slow fibers (LLE: 21%, YE: 7%, OH: 7%), while LLE women had more small slow fibers (LLE: 15%, YE: 8%, OH: 9%). LLE reduced by ~50% the proportion of small fast (MHC II containing) fibers in the aging men (OH: 14%, LLE: 7%) and women (OH: 35%, LLE: 18%). These data, coupled with previous findings, suggest that myonuclei and satellite cell content are uninfluenced by lifelong endurance exercise in men ~60-90 years, and this now also extends to septuagenarian lifelong endurance exercise women. Additionally, lifelong endurance exercise appears to influence the relative abundance of small and large myofibers (fast and slow) differently between men and women.


Subject(s)
Exercise , Muscle Fibers, Fast-Twitch , Muscle Fibers, Slow-Twitch , Physical Endurance , Satellite Cells, Skeletal Muscle , Humans , Female , Male , Satellite Cells, Skeletal Muscle/physiology , Satellite Cells, Skeletal Muscle/cytology , Adult , Physical Endurance/physiology , Exercise/physiology , Aged , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Slow-Twitch/physiology , Muscle Fibers, Slow-Twitch/cytology , Cell Nucleus/physiology , Myosin Heavy Chains/metabolism , Quadriceps Muscle/cytology , Quadriceps Muscle/physiology , Aging/physiology , Young Adult
11.
Biomed Res ; 45(4): 173-177, 2024.
Article in English | MEDLINE | ID: mdl-39010193

ABSTRACT

Sarcopenia is a common complication of chronic kidney disease (CKD) and has a detrimental effect on prognosis. Previous studies have explored the role of secondary calciprotein particles (CPP2) in determining the progression of complications and poor outcomes in patients with CKD. However, no study has demonstrated that CPP2 impairs skeletal myogenesis. Our study revealed that CPP2 exposure inhibits skeletal myogenesis by suppressing myotube formation and expression of skeletal muscle-specific myosin heavy chain and actin in human primary myoblasts. Moreover, CPP2 exposure altered the expression patterns of lineage-determinative transcription factors responsible for regulating myotube differentiation marker genes. This study first demonstrated that CPP2 interferes with myoblast differentiation and myotube formation in vitro.


Subject(s)
Cell Differentiation , Muscle Development , Myoblasts , Humans , Myoblasts/metabolism , Myoblasts/cytology , Cells, Cultured , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Gene Expression Regulation
12.
Pathol Res Pract ; 260: 155476, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39038387

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) are involved in the regulation of colorectal cancer (CRC) progression and chemoresistence. Here, we attempted to reveal the function and mechanism of circ_0000395 in CRC chemoresistence. METHODS: The expression levels of circ_0000395, microRNA (miR)-153-5p, and myosin VI (MYO6) were determined by quantitative real-time PCR. Cell growth, metastasis and oxaliplatin resistance were evaluated via EdU assay, colony formation assay, flow cytometry, transwell assay, and cell counting kit 8 assay. Xenograft tumor model was adopted to evaluate the role of circ_0000395 on CRC tumor growth and oxaliplatin sensitivity. Protein expression of drug-resistance markers and MYO6 was analyzed by western blot. The target relationship between miR-153-5p and circ_0000395 or MYO6 was validated via dual-luciferase reporter assay and RIP assay. RESULTS: Circ_0000395 expression was enhanced in CRC tissues and cells. Silencing of circ_0000395 repressed CRC cell proliferation, migration and invasion, while promoted apoptosis and oxaliplatin sensitivity. Besides, circ_0000395 knockdown also reduced CRC tumor growth and enhanced the sensitivity of tumor to oxaliplatin. Additionally, circ_0000395 acted as a sponge for miR-153-5p, and miR-153-5p targeted MYO6. Functional experiments suggested that miR-153-5p inhibitor or MYO6 overexpression could reverse the suppressive effect of circ_0000395 knockdown on CRC cell growth, metastasis and oxaliplatin resistance. CONCLUSION: Circ_0000395 promoted CRC cell growth, metastasis and oxaliplatin resistance via the miR-153-5p/MYO6 axis, which might provide new insights into the treatment of CRC.


Subject(s)
Cell Proliferation , Colorectal Neoplasms , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , MicroRNAs , Myosin Heavy Chains , Oxaliplatin , RNA, Circular , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , RNA, Circular/genetics , RNA, Circular/metabolism , Drug Resistance, Neoplasm/genetics , Cell Proliferation/drug effects , Animals , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Mice , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Mice, Nude , Cell Movement/drug effects , Apoptosis/drug effects , Male , Mice, Inbred BALB C
13.
J Exp Biol ; 227(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39023116

ABSTRACT

The cheetah is considered the fastest land animal, but studies on their skeletal muscle properties are scarce. Vastus lateralis biopsies, obtained from male and female cheetahs as well as humans, were analysed and compared for fibre type and size, and metabolism. Overall, cheetah muscle had predominantly type IIX fibres, which was confirmed by the myosin heavy chain isoform content (mean±s.d. type I: 17±8%, type IIA: 21±6%, type IIX: 62±12%), whereas human muscle contained predominantly type I and IIA fibres (type I: 49±14%, type IIA: 43±8%, type IIX: 7±7%). Cheetahs had smaller fibres than humans, with larger fibres in the males compared with their female counterparts. Citrate synthase (16±6 versus 28±7 µmol min-1 g-1 protein, P<0.05) and 3-hydroxyacyl co-enzyme A dehydrogenase (30±11 versus 47±15 µmol min-1 g-1 protein, P<0.05) activities were lower in cheetahs than in humans, whereas lactate dehydrogenase activity was 6 times higher in cheetahs (2159±827 versus 382±161 µmol min-1 g-1 protein, P<0.001). The activities of creatine kinase (4765±1828 versus 6485±1298, P<0.05 µmol min-1 g-1 protein) and phosphorylase (111±29 versus 216±92 µmol min-1 g-1 protein) were higher in humans, irrespective of the higher type IIX fibres in cheetahs. Superoxide dismutase and catalase, markers of antioxidant capacity, were higher in humans, but overall antioxidant capacity was higher in cheetahs. To conclude, fibre type, fibre size and metabolism differ between cheetahs and humans, with limited differences between the sexes.


Subject(s)
Acinonyx , Acinonyx/physiology , Acinonyx/metabolism , Male , Female , Animals , Humans , Muscle, Skeletal/metabolism , Adult , Myosin Heavy Chains/metabolism , Sex Characteristics , Sex Factors
14.
Eur J Pharmacol ; 979: 176806, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38986830

ABSTRACT

Chronic kidney disease (CKD) is a clinical syndrome characterized by persistent renal function decline. Renal fibrosis is the main pathological process in CKD, but an effective treatment does not exist. Stratifin (SFN) is a highly-conserved, multi-function soluble acidic protein. Therefore, this study explored the effects of SFN on renal fibrosis. First, we found that SFN was highly expressed in patients with CKD, as well as in renal fibrosis animal and cell models. Next, transforming growth factor-beta 1 (TGF-ß1) induced injury and fibrosis in human renal tubule epithelial cells, and SFN knockdown reversed these effects. Furthermore, SFN knockdown mitigated unilateral ureteral obstruction (UUO)-induced renal tubular dilatation and renal interstitial fibrosis in mice. Liquid chromatography-tandem mass spectrometry/mass spectrometry (LC-MS/MS), co-immunoprecipitation (Co-IP), and immunofluorescence co-localization assays demonstrated that SFN bound the non-muscle myosin-encoding gene, myosin heavy chain 9 (MYH9), in the cytoplasm of renal tubular epithelial cells. MYH9 knockdown also reduced Col-1 and α-SMA expression, which are fibrosis markers. Finally, silencing SFN decreased MYH9 expression, alleviating renal fibrosis. These results suggest that SFN promotes renal fibrosis in CKD by interacting with MYH9. This study may provide potential strategies for the treatment of CKD.


Subject(s)
Kidney , Myosin Heavy Chains , Renal Insufficiency, Chronic , Animals , Humans , Male , Mice , Cell Line , Disease Models, Animal , Fibrosis , Kidney/pathology , Kidney/metabolism , Mice, Inbred C57BL , Molecular Motor Proteins/metabolism , Molecular Motor Proteins/genetics , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Protein Binding , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/genetics , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/pathology , Ureteral Obstruction/metabolism , Ureteral Obstruction/complications
15.
Forensic Sci Int ; 361: 112121, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971138

ABSTRACT

Sudden unexplained death (SUD) is not uncommon in forensic pathology. Yet, diagnosis of SUD remains challenging due to lack of specific biomarkers. This study aimed to screen differentially expressed proteins (DEPs) and validate their usefulness as diagnostic biomarkers for SUD cases. We designed a three-phase investigation, where in the discovery phase, formalin-fixed paraffin-embedded (FFPE) heart specimens were screened through label-free proteomic analysis of cases dying from SUD, mechanical injury and carbon monoxide (CO) intoxication. A total of 26 proteins were identified to be DEPs for the SUD cases after rigorous criterion. Bioinformatics and Adaboost-recursive feature elimination (RFE) analysis further revealed that three of the 26 proteins (MYH6, COX5B and TNNT2) were potential discriminative biomarkers. In the training phase, MYH6 and COX5B were verified to be true DEPs in cardiac tissues from 29 independent SUD cases as compared with a serial of control cases (n = 42). Receiver operating characteristic (ROC) analysis illustrated that combination of MYH6 and COX5B achieved optimal diagnostic sensitivity (89.7 %) and specificity (84.4 %), with area under the curve (AUC) being 0.91. A diagnostic software based on the logistic regression formula derived from the training phase was then constructed. In the validation phase, the diagnostic software was applied to eight authentic SUD cases, seven (87.5 %) of which were accurately recognized. Our study provides a valid strategy towards practical diagnosis of SUD by integrating cardiac MYH6 and COX5B as dual diagnostic biomarkers.


Subject(s)
Biomarkers , Myocardium , Myosin Heavy Chains , Proteomics , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Biomarkers/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Case-Control Studies , Death, Sudden/etiology , Forensic Pathology/methods , Myocardium/metabolism , Myocardium/chemistry , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , ROC Curve , Sensitivity and Specificity
16.
Stem Cell Res ; 80: 103491, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39047410

ABSTRACT

Variants in MYH7 cause cardiomyopathies as well as myosin storage myopathy and Laing early-onset distal myopathy (MPD1). MPD1 is characterized by muscle weakness and atrophy usually beginning in the lower legs. Here, we generated iPSC lines from lymphoblastoid cells of three unrelated individuals heterozygous for the most common MPD1-causing variant; p.Lys1617del. iPSC lines showed typical morphology, expressed pluripotency markers, demonstrated trilineage differentiation potential, and had a normal karyotype. These lines represent the first iPSCs derived from MPD1 patients and complement existing MPD1 animal models. They can provide in vitro platforms to better understand and model MPD1 pathomechanisms and test therapies.


Subject(s)
Cardiac Myosins , Distal Myopathies , Induced Pluripotent Stem Cells , Myosin Heavy Chains , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Distal Myopathies/genetics , Distal Myopathies/pathology , Distal Myopathies/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Male , Female , Cell Line , Cell Differentiation , Adult
17.
Proc Natl Acad Sci U S A ; 121(30): e2319267121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39008679

ABSTRACT

Migrasomes, vesicular organelles generated on the retraction fibers of migrating cells, play a crucial role in migracytosis, mediating intercellular communication. The cargoes determine the functional specificity of migrasomes. Migrasomes harbor numerous intraluminal vesicles, a pivotal component of their cargoes. The mechanism underlying the transportation of these intraluminal vesicles to the migrasomes remains enigmatic. In this study, we identified that Rab10 and Caveolin-1 (CAV1) mark the intraluminal vesicles in migrasomes. Transport of Rab10-CAV1 vesicles to migrasomes required the motor protein Myosin Va and adaptor proteins RILPL2. Notably, the phosphorylation of Rab10 by the kinase LRRK2 regulated this process. Moreover, CSF-1 can be transported to migrasomes through this mechanism, subsequently fostering monocyte-macrophage differentiation in skin wound healing, which served as a proof of the physiological importance of this transporting mechanism.


Subject(s)
Caveolin 1 , Cell Movement , rab GTP-Binding Proteins , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Humans , Caveolin 1/metabolism , Caveolin 1/genetics , Macrophages/metabolism , Phosphorylation , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Myosin Type V/metabolism , Myosin Type V/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Biological Transport , Wound Healing/physiology , Organelles/metabolism
18.
J Ethnopharmacol ; 334: 118527, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38971342

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ulcerative colitis (UC), a recurrent inflammatory bowel disease, continues to challenge effective pharmacologic management. Disulfidptosis, a recently identified form of cell death, appears implicated in the progression of various diseases. Scientific studies have demonstrated that Modified Gegen Qinlian decoction (MGQD) alleviates UC symptoms. However, the underlying mechanisms remain inadequately elucidated. AIM OF THE STUDY: This study investigated the role of disulfidptosis in UC and explored the potential of MGQD to ameliorate UC by mediating disulfidptosis. METHODS: Microarray data were utilized to identify disulfidptosis-related genes stably expressed in UC, and integrated genomic analyses were conducted to elucidate the landscape of disulfidptosis in UC. Subsequently, C57BL/6J mice were administered 3% dextran sodium sulfate (DSS) to induce experimental colitis and treated with MGQD. Quantitative real-time polymerase chain reaction and immunohistochemical analysis of colonic tissues from colitis mice were performed to validate the microarray data findings. Finally, molecular docking was employed to explore the binding interactions between MGQD components and disulfidptosis biomarkers. RESULTS: Myosin heavy chain 10 (MYH10) and filamin A (FLNA) were identified as stably expressed in UC, demonstrating high diagnostic value for the disease. Correlation analysis indicated that disulfidptosis-related genes are associated with elevated levels of immune cells in UC. Single gene set enrichment analysis further clarified that these genes might be involved in the pathological processes of UC via immune-related pathways. Subsequent animal experiments revealed that MYH10 and FLNA were significantly upregulated in mice with colitis, a condition reversed by MGQD treatment. Molecular docking results showed that MYH10 and FLNA serve as stable binding targets for the primary components of MGQD. CONCLUSIONS: The study identified a connection between the disulfidptosis-related landscape and immune infiltration in UC, suggesting that MGQD may modulate disulfidptosis by inhibiting MYH10 and FLNA, thereby alleviating UC.


Subject(s)
Colitis, Ulcerative , Dextran Sulfate , Drugs, Chinese Herbal , Mice, Inbred C57BL , Animals , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Drugs, Chinese Herbal/pharmacology , Male , Mice , Molecular Docking Simulation , Disease Models, Animal , Colon/drug effects , Colon/pathology , Colon/metabolism , Colon/immunology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism
19.
Mol Biol Rep ; 51(1): 822, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39023774

ABSTRACT

BACKGROUND: Testicular descent is a physiological process regulated by many factors. Eventually, disturbances in the embryological/fetal development path facilitate the occurrence of scrotal hernia, a congenital malformation characterized by the presence of intestinal portions within the scrotal sac due to the abnormal expansion of the inguinal ring. In pigs, some genes have been related to this anomaly, but the genetic mechanisms involved remain unclear. This study aimed to investigate the expression profile of a set of genes potentially involved with the manifestation of scrotal hernia in the inguinal ring tissue. METHODS AND RESULTS: Tissue samples from the inguinal ring/canal of normal and scrotal hernia-affected male pigs with approximately 30 days of age were used. Relative expression analysis was performed using qPCR to confirm the expression profile of 17 candidate genes previously identified in an RNA-Seq study. Among them, the Myosin heavy chain 1 (MYH1), Desmin (DES), and Troponin 1 (TNNI1) genes were differentially expressed between groups and had reduced levels of expression in the affected animals. These genes encode proteins involved in the formation of muscle tissue, which seems to be important for increasing the resistance of the inguinal ring to the abdominal pressure, which is essential to avoid the occurrence of scrotal hernia. CONCLUSIONS: The downregulation of muscular candidate genes in the inguinal tissue clarifies the genetic mechanisms involved with this anomaly in its primary site, providing useful information for developing strategies to control this malformation in pigs and other mammals.


Subject(s)
Down-Regulation , Scrotum , Animals , Male , Swine/genetics , Scrotum/metabolism , Scrotum/abnormalities , Scrotum/pathology , Down-Regulation/genetics , Hernia, Inguinal/genetics , Hernia, Inguinal/metabolism , Hernia, Inguinal/veterinary , Gene Expression Profiling/methods , Swine Diseases/genetics , Swine Diseases/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism
20.
Cell Commun Signal ; 22(1): 312, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38902769

ABSTRACT

African American (AA) women are twice as likely to develop triple-negative breast cancer (TNBC) as women of European descent. Additionally, AA women with TNBC present a much more aggressive disease course than their European American (EA) counterparts. Thus, there is an unmet clinical need to identify race-specific biomarkers and improve survival outcomes in AA patients with TNBC. The minus-end directed microtubule motor protein kinesin family member C1 (KIFC1) promotes centrosome clustering and chromosomal instability and is often overexpressed in TNBC. Previous findings suggest that KIFC1 plays a role in cell proliferation and migration in TNBC cells from AAs and that the levels of nuclear KIFC1 (nKIFC1) are particularly high in AA patients with TNBC. The nuclear localization of KIFC1 in interphase may underlie its previously unrecognized race-specific association. In this study, we found that in TNBC cells derived from AAs, nKIFC1 interacted with the tumor suppressor myosin heavy chain 9 (MYH9) over EA cells. Treatment of AA TNBC cells with commercial inhibitors of KIFC1 and MYH9 disrupted the interaction between KIFC1 and MYH9. To characterize the racial differences in the KIFC1-MYH9-MYC axis in TNBC, we established homozygous KIFC1 knockout (KO) TNBC cell lines. KIFC1 KO significantly inhibited proliferation, migration, and invasion in AA TNBC cells but not in EA TNBC cells. RNA sequencing analysis showed significant downregulation of genes involved in cell migration, invasion, and metastasis upon KIFC1 KO in TNBC cell lines from AAs compared to those from EAs. These data indicate that mechanistically, the role of nKIFC1 in driving TNBC progression and metastasis is stronger in AA patients than in EA patients, and that KIFC1 may be a critical therapeutic target for AA patients with TNBC.


Subject(s)
Kinesins , Myosin Heavy Chains , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/ethnology , Triple Negative Breast Neoplasms/metabolism , Kinesins/genetics , Kinesins/metabolism , Female , Cell Line, Tumor , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Cell Proliferation/genetics , Cell Movement/genetics , Black or African American/genetics , White People/genetics , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL