Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
BMC Musculoskelet Disord ; 25(1): 388, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762738

ABSTRACT

BACKGROUND: A variety of measurement methods and imaging modalities are in use to quantify the morphology of lateral femoral condyle (LFC), but the most reliable method remains elusive in patients with lateral patellar dislocation (LPD). The purpose of this study was to determine the intra- and inter-observer reliability of different measurement methods for evaluating the morphology of LFC on different imaging modalities in patients with LPD. METHODS: Seventy-three patients with LPD were included. Four parameters for quantifying the morphology of LFC were retrospectively measured by three observers on MRI, sagittal CT image, conventional radiograph (CR), and three-dimensional CT (3D-CT). The intra-class correlation coefficient was calculated to determine the intra- and inter-observer reliability. Bland-Altman analysis was conducted to identify the bias between observers. RESULTS: The lateral femoral condyle index (LFCI) showed better intra- and inter-observer reliability on MRI and 3D-CT than on CR and sagittal CT images. The mean difference in the LFCI between observers was lowest on 3D-CT (0.047), higher on MRI (0.053), and highest on sagittal CT images (0.062). The LFCI was associated with the lateral femoral condyle ratio (ρ = 0.422, P = 0.022), lateral condyle index (r = 0.413, P = 0.037), and lateral femoral condyle distance (r = 0.459, P = 0.014). The LFCI could be reliably measured by MRI and 3D-CT. CONCLUSION: The LFCI could be reliably measured by MRI and 3D-CT. The LFCI was associated with both the height and length of LFC and could serve as a comprehensive parameter for quantifying the morphology of LFC in patients with LPD.


Subject(s)
Femur , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Observer Variation , Patellar Dislocation , Tomography, X-Ray Computed , Humans , Female , Male , Reproducibility of Results , Patellar Dislocation/diagnostic imaging , Magnetic Resonance Imaging/methods , Femur/diagnostic imaging , Retrospective Studies , Young Adult , Adult , Imaging, Three-Dimensional/methods , Adolescent
2.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38673895

ABSTRACT

Voltage-gated potassium (Kv) channels and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels share similar structures but have opposite gating polarity. Kv channels have a strong coupling (>109) between the voltage sensor (S4) and the activation gate: when S4s are activated, the gate is open to >80% but, when S4s are deactivated, the gate is open <10-9 of the time. Using noise analysis, we show that the coupling between S4 and the gate is <200 in HCN channels. In addition, using voltage clamp fluorometry, locking the gate open in a Kv channel drastically altered the energetics of S4 movement. In contrast, locking the gate open or decreasing the coupling between S4 and the gate in HCN channels had only minor effects on the energetics of S4 movement, consistent with a weak coupling between S4 and the gate. We propose that this loose coupling is a prerequisite for the reversed voltage gating in HCN channels.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channel Gating , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Animals , Patch-Clamp Techniques , Humans
3.
Exp Biol Med (Maywood) ; 248(15): 1279-1287, 2023 08.
Article in English | MEDLINE | ID: mdl-37897221

ABSTRACT

Osteoarthritis (OA) is one of the most prevalent joint disorders associated with the degradation of articular cartilage and an abnormal mechanical microenvironment. Mechanical stimuli, including compression, shear stress, stretching strain, osmotic challenge, and the physical properties of the matrix microenvironment, play pivotal roles in the tissue homeostasis of articular cartilage. The primary cilium, as a mechanosensory and chemosensory organelle, is important for detecting and transmitting both mechanical and biochemical signals in chondrocytes within the matrix microenvironment. Growing evidence indicates that primary cilia are critical for chondrocytes signaling transduction and the matrix homeostasis of articular cartilage. Furthermore, the ability of primary cilium to regulate cellular signaling is dynamic and dependent on the cellular matrix microenvironment. In the current review, we aim to elucidate the key mechanisms by which primary cilia mediate chondrocytes sensing and responding to the matrix mechanical microenvironment. This might have potential therapeutic applications in injuries and OA-associated degeneration of articular cartilage.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Chondrocytes/metabolism , Mechanotransduction, Cellular/physiology , Cilia/physiology , Signal Transduction , Cartilage, Articular/metabolism , Osteoarthritis/metabolism
4.
Int J Mol Sci ; 24(15)2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37569465

ABSTRACT

Long QT syndrome (LQTS) can lead to ventricular arrhythmia and sudden cardiac death. The most common congenital cause of LQTS is mutations in the channel subunits generating the cardiac potassium current IKs. Zebrafish (Danio rerio) have been proposed as a powerful system to model human cardiac diseases due to the similar electrical properties of the zebrafish heart and the human heart. We used high-resolution all-optical electrophysiology on ex vivo zebrafish hearts to assess the effects of IKs analogues on the cardiac action potential. We found that chromanol 293B (an IKs inhibitor) prolonged the action potential duration (APD) in the presence of E4031 (an IKr inhibitor applied to drug-induced LQT2), and to a lesser extent, in the absence of E4031. Moreover, we showed that PUFA analogues slightly shortened the APD of the zebrafish heart. However, PUFA analogues failed to reverse the APD prolongation in drug-induced LQT2. However, a more potent IKs activator, ML-277, partially reversed the APD prolongation in drug-induced LQT2 zebrafish hearts. Our results suggest that IKs plays a limited role in ventricular repolarizations in the zebrafish heart under resting conditions, although it plays a more important role when the IKr is compromised, as if the IKs in zebrafish serves as a repolarization reserve as in human hearts. This study shows that potent IKs activators can restore the action potential duration in drug-induced LQT2 in the zebrafish heart.


Subject(s)
Long QT Syndrome , Potassium Channels, Voltage-Gated , Animals , Humans , Anti-Arrhythmia Agents/pharmacology , Zebrafish , Heart , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Long QT Syndrome/drug therapy , Long QT Syndrome/genetics , Action Potentials , Potassium Channels, Voltage-Gated/pharmacology
5.
J Gen Physiol ; 155(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-36752823

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels contribute to the rhythmic firing of pacemaker neurons and cardiomyocytes. Mutations in HCN channels are associated with cardiac arrhythmia and epilepsy. HCN channels belong to the superfamily of voltage-gated K+ channels, most of which are activated by depolarization. HCN channels, however, are activated by hyperpolarization. The mechanism behind this reversed gating polarity of HCN channels is not clear. We here show that sea urchin HCN (spHCN) channels with mutations in the C-terminal part of the voltage sensor use the same voltage-sensor movement to either close or open in response to hyperpolarizations depending on the absence or presence of cAMP. Our results support that non-covalent interactions at the C-terminal end of the voltage sensor are critical for HCN gating polarity. These interactions are also critical for the proper closing of the channels because these mutations exhibit large constitutive currents. Since a similar voltage-sensor movement can cause both depolarization- and hyperpolarization-activation in the same channel, this suggests that the coupling between the voltage sensor and the pore is changed to create channels opened by different polarities. We also show an identical voltage-sensor movement in activated and inactivated spHCN channels and suggest a model for spHCN activation and inactivation. Our results suggest the possibility that channels open by opposite voltage dependence, such as HCN and the related EAG channels, use the same voltage-sensor movement but different coupling mechanisms between the voltage sensor and the gate.


Subject(s)
Ion Channel Gating , Potassium Channels , Potassium Channels/metabolism , Ion Channel Gating/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mutation , Cyclic Nucleotide-Gated Cation Channels/genetics
6.
Cancer Med ; 12(2): 1643-1654, 2023 01.
Article in English | MEDLINE | ID: mdl-35789211

ABSTRACT

BACKGROUND: Kushenol A is natural flavonoid extract discovered in recent years, with potential anti-tumor activity. Its role in breast cancer is poorly understood. METHODS: To investigate biological function of Kushenol A in breast cancer (BC), Cell Counting Kit-8 assay, colony formation assay, flow cytometry, western blotting, qPCR analysis, and xenograft mouse model were performed. RESULTS: We found that Kushenol A treatment reduced proliferative capability and induced G0/G1 phase cell cycle arrest and apoptosis of BC cells in a concentration-dependent manner. Besides, Kushenol A treatment contributed to the upregulation of apoptosis-related and cell cycle-associated genes. In nude mice, Kushenol A administration repressed BC xenograft tumor growth. Mechanistically, phosphorylation levels of AKT and mTOR were markedly attenuated in Kushenol A-treated BC cells; however, there were no significant differences in total AKT and mTOR expressions. Moreover, PI3K inhibitor combined with Kushenol A exhibited synergistic inhibitory activity on cell proliferation. CONCLUSIONS: Taken together, our findings suggested that Kushenol A suppressed BC cell proliferation by modulating PI3K/AKT/mTOR signaling pathway. Kushenol A may be a promising therapeutic drug for treating BC.


Subject(s)
Breast Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Animals , Mice , Female , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Mice, Nude , TOR Serine-Threonine Kinases/metabolism , Flavonoids/pharmacology , Breast Neoplasms/pathology , Apoptosis , Cell Proliferation , Cell Line, Tumor
7.
Front Physiol ; 13: 902224, 2022.
Article in English | MEDLINE | ID: mdl-36505078

ABSTRACT

The congenital Long QT Syndrome (LQTS) is an inherited disorder in which cardiac ventricular repolarization is delayed and predisposes patients to cardiac arrhythmias and sudden cardiac death. LQT1 and LQT5 are LQTS variants caused by mutations in KCNQ1 or KCNE1 genes respectively. KCNQ1 and KCNE1 co-assemble to form critical IKS potassium channels. Beta-blockers are the standard of care for the treatment of LQT1, however, doing so based on mechanisms other than correcting the loss-of-function of K+ channels. ML277 and R-L3 are compounds that enhance IKS channels and slow channel deactivation in a manner that is dependent on the stoichiometry of KCNE1 subunits in the assembled channels. In this paper, we used expression of IKS channels in Chinese hamster ovary (CHO) cells and Xenopus oocytes to study the potential of these two drugs (ML277 and R-L3) for the rescue of LQT1 and LQT5 mutant channels. We focused on the LQT1 mutation KCNQ1-S546L, and two LQT5 mutations, KCNE1-L51H and KCNE1-G52R. We found ML277 and R-L3 potentiated homozygote LQTS mutations in the IKS complexes-KCNE1-G52R and KCNE1-L51H and in heterogeneous IKS channel complexes which mimic heterogeneous expression of mutations in patients. ML277 and R-L3 increased the mutant IKS current amplitude and slowed current deactivation, but not in wild type (WT) IKS. We obtained similar results in the LQT1 mutant (KCNQ1 S546L/KCNE1) with ML277 and R-L3. ML277 and R-L3 had a similar effect on the LQT1 and LQT5 mutants, however, ML277 was more effective than R-L3 in this modulation. Importantly we found that not all LQT5 mutants expressed with KCNQ1 resulted in channels that are potentiated by these drugs as the KCNE1 mutant D76N inhibited drug action when expressed with KCNQ1. Thus, our work shows that by directly studying the treatment of LQT1 and LQT5 mutations with ML277 and R-L3, we will understand the potential utility of these activators as options in specific LQTS therapeutics.

8.
Elife ; 112022 06 01.
Article in English | MEDLINE | ID: mdl-35642783

ABSTRACT

Neuronal KCNQ channels mediate the M-current, a key regulator of membrane excitability in the central and peripheral nervous systems. Mutations in KCNQ2 channels cause severe neurodevelopmental disorders, including epileptic encephalopathies. However, the impact that different mutations have on channel function remains poorly defined, largely because of our limited understanding of the voltage-sensing mechanisms that trigger channel gating. Here, we define the parameters of voltage sensor movements in wt-KCNQ2 and channels bearing epilepsy-associated mutations using cysteine accessibility and voltage clamp fluorometry (VCF). Cysteine modification reveals that a stretch of eight to nine amino acids in the S4 becomes exposed upon voltage sensing domain activation of KCNQ2 channels. VCF shows that the voltage dependence and the time course of S4 movement and channel opening/closing closely correlate. VCF reveals different mechanisms by which different epilepsy-associated mutations affect KCNQ2 channel voltage-dependent gating. This study provides insight into KCNQ2 channel function, which will aid in uncovering the mechanisms underlying channelopathies.


Subject(s)
Epilepsy , KCNQ2 Potassium Channel , Neurodevelopmental Disorders , Cysteine/genetics , Epilepsy/genetics , Humans , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , Mutation , Neurodevelopmental Disorders/genetics
9.
Acta Biochim Biophys Sin (Shanghai) ; 54(1): 113-125, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35130619

ABSTRACT

The pericellular matrix stiffness is strongly associated with its biochemical and structural changes during the aging and osteoarthritis progress of articular cartilage. However, how substrate stiffness modulates the chondrocyte regulatory volume decrease (RVD) and calcium signaling in chondrocytes remains unknown. This study aims to investigate the effects of substrate stiffness on the chondrocyte RVD and calcium signaling by recapitulating the physiologically relevant substrate stiffness. Our results showed that substrate stiffness induces completely different dynamical deformations between the cell swelling and recovering progresses. Chondrocytes swell faster on the soft substrate but recovers slower than the stiff substrate during the RVD response induced by the hypo-osmotic challenge. We found that stiff substrate enhances the cytosolic Ca oscillation of chondrocytes in the iso-osmotic medium. Furthermore, chondrocytes exhibit a distinctive cytosolic Ca oscillation during the RVD response. Soft substrate significantly improves the Ca oscillation in the cell swelling process whereas stiff substrate enhances the cytosolic Ca oscillation in the cell recovering process. Our work also suggests that the TRPV4 channel is involved in the chondrocyte sensing substrate stiffness by mediating Ca signaling in a stiffness-dependent manner. This helps to understand a previously unidentified relationship between substrate stiffness and RVD response under the hypo-osmotic challenge. A better understanding of substrate stiffness regulating chondrocyte volume and calcium signaling will aid the development of novel cell-instructive biomaterial to restore cellular functions.


Subject(s)
Cartilage, Articular , Osteoarthritis , Calcium/metabolism , Calcium Signaling , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Humans , Osmosis/physiology , Osteoarthritis/metabolism
10.
Integr Psychol Behav Sci ; 56(2): 426-458, 2022 06.
Article in English | MEDLINE | ID: mdl-34988873

ABSTRACT

Over more than three decades New Zealand (NZ) has abolished its racially-biased immigration policy and changed to select immigrants based on personal merits, since 1997 new Chinese immigrants from China have become the second-largest immigrant group in NZ. Reflecting the rapid changes in Mainland Chinese society, different waves of Chinese immigrants have arrived in NZ during the past three decades, each carrying distinctive characteristics. Despite the magnitude of this immigrant population both globally and in NZ, the subethnicities of these immigrants have never been conceptualised theoretically. Based on the digitally- enhanced research techniques together with ethnographically-based study, the paper aims to remedy this research gap in Chinese diaspora studies. Two theoretical concepts are used in this research. The first is the concept of sub-ethnicity which refers to finer boundaries drawn within an ethnic group by nationality, language, region of origin, class, or other distinctions. The second is the concept of ethnoburb - a model of ethnic settlement where the suburban ethnic communities and clusters of associated residential areas and business districts in large metropolitan cities are highly concentrated. This research considers these two concepts are correlated to each other since certain ethnoburb has particular attraction to certain sub-ethnic group; and vice versa, certain subethnic group intends to gather in same ethnoburb unintentionally or unconsciously. Applying these concepts, the research considers Albany as an example of a distinctive Chinese ethnoburb for the China-born new immigrants, especially for the most recent arrivals. The paper makes theoretical contribution to understand the complementarity between these two concepts and their methodological implementation towards studying new Chinese migration.


Subject(s)
Emigrants and Immigrants , Ethnicity , China , Demography , Humans , New Zealand , Population Dynamics
11.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504015

ABSTRACT

Rhythmic activity in pacemaker cells, as in the sino-atrial node in the heart, depends on the activation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. As in depolarization-activated K+ channels, the fourth transmembrane segment S4 functions as the voltage sensor in hyperpolarization-activated HCN channels. But how the inward movement of S4 in HCN channels at hyperpolarized voltages couples to channel opening is not understood. Using voltage clamp fluorometry, we found here that S4 in HCN channels moves in two steps in response to hyperpolarizations and that the second S4 step correlates with gate opening. We found a mutation in sea urchin HCN channels that separate the two S4 steps in voltage dependence. The E356A mutation in S4 shifts the main S4 movement to positive voltages, but channel opening remains at negative voltages. In addition, E356A reveals a second S4 movement at negative voltages that correlates with gate opening. Cysteine accessibility and molecular models suggest that the second S4 movement opens up an intracellular crevice between S4 and S5 that would allow radial movement of the intracellular ends of S5 and S6 to open HCN channels.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Animals , Biological Clocks/physiology , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Ion Channel Gating/physiology , Membrane Potentials/physiology , Patch-Clamp Techniques/methods , Potassium Channels/metabolism , Sea Urchins/metabolism
12.
Commun Biol ; 4(1): 887, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34285340

ABSTRACT

Voltage-gated KCNQ1 channels contain four separate voltage-sensing domains (VSDs) and a pore domain (PD). KCNQ1 expressed alone opens when the VSDs are in an intermediate state. In cardiomyocytes, KCNQ1 co-expressed with KCNE1 opens mainly when the VSDs are in a fully activated state. KCNE1 also drastically slows the opening of KCNQ1 channels and shifts the voltage dependence of opening by >40 mV. We here show that mutations of conserved residues at the VSD-PD interface alter the VSD-PD coupling so that the mutant KCNQ1/KCNE1 channels open in the intermediate VSD state. Using recent structures of KCNQ1 and KCNE beta subunits in different states, we present a mechanism by which KCNE1 rotates the VSD relative to the PD and affects the VSD-PD coupling of KCNQ1 channels in a non-canonical way, forcing KCNQ1/KCNE1 channels to open in the fully-activated VSD state. This would explain many of the KCNE1-induced effects on KCNQ1 channels.


Subject(s)
Ion Channel Gating , KCNQ1 Potassium Channel/genetics , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/genetics , KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism
13.
Diabetes Metab Syndr Obes ; 14: 305-313, 2021.
Article in English | MEDLINE | ID: mdl-33519219

ABSTRACT

BACKGROUND AND OBJECTIVE: The 12-month follow-up effect of the self-efficacy-focused structured education program (SSEP) requires in-depth confirmation. This study aims to verify whether the benefits of SSEP can be maintained in 12 months. MATERIALS AND METHODS: A multicenter randomized controlled trial with 12-month follow-up conducted among 265 type 2 diabetes patients not on insulin from 4 hospitals in mainland China. The intervention group (n = 133) was administrated with SSEP, and the control group (n = 132) received the routine education. The indicators of metabolic and psychosocial aspects of the patients were assessed at baseline and 12 months. RESULTS: As opposed to the control group, the primary outcomes of HbA1c in the intervention group were improved obviously in the 12th month during the 12-month follow-up (-1.13%, P < 0.001). The secondary outcomes (ie, waist circumference, total cholesterol, low-density lipoprotein cholesterol, diabetes self-efficacy, diabetes self-management behaviors, diabetes knowledge and diabetes distress) were improved significantly in the intervention group as compared with the control group in the 12th month during the 12-month follow-up (-3.14 cm, P = 0.001; -0.30 mmol/L, P = 0.032; -0.25 mmol/L, P = 0.008; 0.87, P < 0.001; 10.67, P < 0.001; 3.42, P < 0.001; -4.97, P < 0.001). The non-significant difference in the secondary outcomes (ie, systolic pressure, diastolic pressure, triglycerides and high-density lipoprotein cholesterol) was identified between the two groups in the 12th month during the 12-month follow-up (P > 0.05). CONCLUSION: The SSEP provided sustainable benefits in outcomes of HbA1c, waist circumference, total cholesterol, low-density lipoprotein cholesterol, diabetes knowledge, diabetes distress, diabetes self-efficacy and diabetes self-management behaviors for type 2 diabetes patients not on insulin in the 12th month during the 12-month follow-up. Thus, it will be an effective education model capable of being generalized nationwide, and it can be referenced for the nations and regions under consistent conditions. CLINICAL TRIAL REGISTRY: Chinese Clinical Trial Registry (ChiCTR-IOR-17011007).

14.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 10-18, 2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33210711

ABSTRACT

Cell migration and invasion are two essential processes during cancer metastasis. Increasing evidence has shown that the Piezo1 channel is involved in mediating cell migration and invasion in some types of cancers. However, the role of Piezo1 in the breast cancer and its underlying mechanisms have not been clarified yet. Here, we show that Piezo1 is high-expressed in breast cancer cell (BCC) lines, despite its complex expression in clinical patient database. Piezo1 knockdown (Piezo1-KD) promotes unconfined BCC migration, but impedes confined cell migration. Piezo1 may mediate BCC migration through the balances of cell adhesion, cell stiffness, and contractility. Furthermore, Piezo1-KD inhibits BCC invasion by impairing the invadopodium formation and suppressing the expression of metalloproteinases (MMPs) as well. However, the proliferation and cell cycle of BCCs are not significantly affected by Piezo1. Our study highlights a crucial role of Piezo1 in regulating migration and invasion of BCCs, indicating Piezo1 channel might be a new prognostic and therapeutic target in BCCs.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Ion Channels/genetics , Ion Channels/metabolism , Actins/metabolism , Biomechanical Phenomena , Breast Neoplasms/immunology , Cell Line, Tumor , Cell Movement/genetics , Databases, Genetic , Female , Focal Adhesions/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness/genetics , Podosomes/metabolism
15.
Int J Mol Sci ; 21(24)2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33322401

ABSTRACT

The delayed rectifier potassium IKs channel is an important regulator of the duration of the ventricular action potential. Hundreds of mutations in the genes (KCNQ1 and KCNE1) encoding the IKs channel cause long QT syndrome (LQTS). LQTS is a heart disorder that can lead to severe cardiac arrhythmias and sudden cardiac death. A better understanding of the IKs channel (here called the KCNQ1/KCNE1 channel) properties and activities is of great importance to find the causes of LQTS and thus potentially treat LQTS. The KCNQ1/KCNE1 channel belongs to the superfamily of voltage-gated potassium channels. The KCNQ1/KCNE1 channel consists of both the pore-forming subunit KCNQ1 and the modulatory subunit KCNE1. KCNE1 regulates the function of the KCNQ1 channel in several ways. This review aims to describe the current structural and functional knowledge about the cardiac KCNQ1/KCNE1 channel. In addition, we focus on the modulation of the KCNQ1/KCNE1 channel and its potential as a target therapeutic of LQTS.


Subject(s)
KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Humans , KCNQ1 Potassium Channel/genetics , Long QT Syndrome/metabolism , Potassium Channels, Voltage-Gated/genetics
17.
Elife ; 92020 03 24.
Article in English | MEDLINE | ID: mdl-32207683

ABSTRACT

The cardiac ventricular action potential depends on several voltage-gated ion channels, including NaV, CaV, and KV channels. Mutations in these channels can cause Long QT Syndrome (LQTS) which increases the risk for ventricular fibrillation and sudden cardiac death. Polyunsaturated fatty acids (PUFAs) have emerged as potential therapeutics for LQTS because they are modulators of voltage-gated ion channels. Here we demonstrate that PUFA analogues vary in their selectivity for human voltage-gated ion channels involved in the ventricular action potential. The effects of specific PUFA analogues range from selective for a specific ion channel to broadly modulating cardiac ion channels from all three families (NaV, CaV, and KV). In addition, a PUFA analogue selective for the cardiac IKs channel (Kv7.1/KCNE1) is effective in shortening the cardiac action potential in human-induced pluripotent stem cell-derived cardiomyocytes. Our data suggest that PUFA analogues could potentially be developed as therapeutics for LQTS and cardiac arrhythmia.


Subject(s)
Calcium Channels, L-Type/drug effects , Fatty Acids, Unsaturated/pharmacology , KCNQ1 Potassium Channel/drug effects , NAV1.5 Voltage-Gated Sodium Channel/drug effects , Potassium Channels, Voltage-Gated/drug effects , Xenopus Proteins/drug effects , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Calcium Channels, L-Type/physiology , Induced Pluripotent Stem Cells/cytology , KCNQ1 Potassium Channel/physiology , Long QT Syndrome/drug therapy , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel/physiology , Potassium Channels, Voltage-Gated/physiology , Xenopus Proteins/physiology , Xenopus laevis
18.
J Biomech ; 104: 109729, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32147239

ABSTRACT

In articular cartilage, the function of chondrocytes is strongly related to their zone-specific microniche geometry defined by pericellular matrix. Microniche geometry is critical for regulating the phenotype and function of the chondrocyte in native cartilage and tissue engineering constructs. However the role of microniche geometry in the mechanical properties and calcium signaling of chondrocytes remains unknown. To recapitulate microniche geometry at single-cell level, we engineered three basic physiological-related polydimethylsiloxane (PDMS) microniches geometries fabricated using soft lithography. We cultured chondrocytes in these microniche geometries and quantified cell mechanical properties using atomic force microscopy (AFM). Fluorescent calcium indicator was used to record and quantify cytosolic Ca2+ oscillation of chondrocytes in different geometries. Our work showed that microniche geometry modulated the mechanical behavior and calcium signaling of chondrocytes. The ellipsoidal microniches significantly enhanced the mechanical properties of chondrocytes compared to spheroidal microniche. Additionally, ellipsoidal microniches can markedly improved the amplitude but weakened the frequency of cytosolic Ca2+ oscillation in chondrocytes than spheroidal microniche. Our work might reveal a novel understanding of chondrocyte mechanotransduction and therefore be useful for designing cell-instructive scaffolds for functional cartilage tissue engineering.


Subject(s)
Cartilage, Articular , Chondrocytes , Calcium Signaling , Cartilage, Articular/metabolism , Mechanotransduction, Cellular , Tissue Engineering
19.
ACS Biomater Sci Eng ; 6(8): 4623-4630, 2020 08 10.
Article in English | MEDLINE | ID: mdl-33455169

ABSTRACT

Cancer stem cells (CSCs) play a critical role in the cancer metastasis and account for tumor heterogeneity. Growing evidence indicates that the CSC phenotypes are related to the tumor microenvironment. In this study, we report that the gradient of mechanical stresses guides the spatial patterning of the expression of CD44 and Yes-associated protein (YAP) in the geometrically confined multicellular sheets. Our study shows that the cytoskeletal contraction regulates the expression of CD44 through the translocation of YAP into the nucleus. The results demonstrate that geometric confinement and mechanical stresses are the regulators in the spatial patterning of CSC. It may help to understand the relationship between the tumor microenvironment and oncogenesis.


Subject(s)
Neoplasms , Traction , Biomarkers, Tumor , Humans , Hyaluronan Receptors/genetics , Neoplasms/genetics , Neoplastic Stem Cells , Tumor Microenvironment
20.
J Gen Physiol ; 152(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-31865382

ABSTRACT

Repolarization and termination of the ventricular cardiac action potential is highly dependent on the activation of the slow delayed-rectifier potassium IKs channel. Disruption of the IKs current leads to the most common form of congenital long QT syndrome (LQTS), a disease that predisposes patients to ventricular arrhythmias and sudden cardiac death. We previously demonstrated that polyunsaturated fatty acid (PUFA) analogues increase outward K+ current in wild type and LQTS-causing mutant IKs channels. Our group has also demonstrated the necessity of a negatively charged PUFA head group for potent activation of the IKs channel through electrostatic interactions with the voltage-sensing and pore domains. Here, we test whether the efficacy of the PUFAs can be tuned by the presence of different functional groups in the PUFA head, thereby altering the electrostatic interactions of the PUFA head group with the voltage sensor or the pore. We show that PUFA analogues with taurine and cysteic head groups produced the most potent activation of IKs channels, largely by shifting the voltage dependence of activation. In comparison, the effect on voltage dependence of PUFA analogues with glycine and aspartate head groups was half that of the taurine and cysteic head groups, whereas the effect on maximal conductance was similar. Increasing the number of potentially negatively charged moieties did not enhance the effects of the PUFA on the IKs channel. Our results show that one can tune the efficacy of PUFAs on IKs channels by altering the pKa of the PUFA head group. Different PUFAs with different efficacy on IKs channels could be developed into more personalized treatments for LQTS patients with a varying degree of IKs channel dysfunction.


Subject(s)
Fatty Acids, Unsaturated/metabolism , Potassium Channels, Voltage-Gated/metabolism , Action Potentials/physiology , Animals , Arrhythmias, Cardiac/metabolism , Heart Ventricles/metabolism , KCNQ1 Potassium Channel , Long QT Syndrome/metabolism , Myocytes, Cardiac/metabolism , Oocytes/physiology , Potassium/pharmacokinetics , Xenopus laevis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...