Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38.379
Filter
1.
Nat Commun ; 15(1): 7020, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147733

ABSTRACT

Mechanosensitive PIEZO2 ion channels play roles in touch, proprioception, and inflammatory pain. Currently, there are no small molecule inhibitors that selectively inhibit PIEZO2 over PIEZO1. The TMEM120A protein was shown to inhibit PIEZO2 while leaving PIEZO1 unaffected. Here we find that TMEM120A expression elevates cellular levels of phosphatidic acid and lysophosphatidic acid (LPA), aligning with its structural resemblance to lipid-modifying enzymes. Intracellular application of phosphatidic acid or LPA inhibits PIEZO2 but not PIEZO1 activity. Extended extracellular exposure to the non-hydrolyzable phosphatidic acid and LPA analog carbocyclic phosphatidic acid (ccPA) also inhibits PIEZO2. Optogenetic activation of phospholipase D (PLD), a signaling enzyme that generates phosphatidic acid, inhibits PIEZO2 but not PIEZO1. Conversely, inhibiting PLD leads to increased PIEZO2 activity and increased mechanical sensitivity in mice in behavioral experiments. These findings unveil lipid regulators that selectively target PIEZO2 over PIEZO1, and identify the PLD pathway as a regulator of PIEZO2 activity.


Subject(s)
Ion Channels , Lysophospholipids , Phosphatidic Acids , Ion Channels/metabolism , Ion Channels/genetics , Animals , Phosphatidic Acids/metabolism , Humans , Mice , Lysophospholipids/metabolism , HEK293 Cells , Phospholipase D/metabolism , Phospholipase D/genetics , Mechanotransduction, Cellular , Mice, Inbred C57BL , Male , Optogenetics
2.
Sci Adv ; 10(32): eadn0367, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121219

ABSTRACT

The development of noninvasive approaches to precisely control neural activity in mammals is highly desirable. Here, we used the ion channel transient receptor potential ankyrin-repeat 1 (TRPA1) as a proof of principle, demonstrating remote near-infrared (NIR) activation of endogenous neuronal channels in mice through an engineered nanoagonist. This achievement enables specific neurostimulation in nongenetically modified mice. Initially, target-based screening identified flavins as photopharmacological agonists, allowing for the photoactivation of TRPA1 in sensory neurons upon ultraviolet A/blue light illumination. Subsequently, upconversion nanoparticles (UCNPs) were customized with an emission spectrum aligned to flavin absorption and conjugated with flavin adenine dinucleotide, creating a nanoagonist capable of NIR activation of TRPA1. Following the intrathecal injection of the nanoagonist, noninvasive NIR stimulation allows precise bidirectional control of nociception in mice through remote activation of spinal TRPA1. This study demonstrates a noninvasive NIR neurostimulation method with the potential for adaptation to various endogenous ion channels and neural processes by combining photochemical toolboxes with customized UCNPs.


Subject(s)
Infrared Rays , Nanoparticles , TRPA1 Cation Channel , Animals , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/agonists , Mice , Nanoparticles/chemistry , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Sensory Receptor Cells/drug effects , Ion Channels/metabolism , Nociception/drug effects
3.
Handb Clin Neurol ; 203: 1-23, 2024.
Article in English | MEDLINE | ID: mdl-39174242

ABSTRACT

Ion channels are membrane proteins that allow the passage of ions across the membrane. They characteristically contain a pore where the selectivity of certain ion species is determined and gates that open and close the pore are found. The pore is often connected to additional domains or subunits that regulate its function. Channels are grouped into families based on their selectivity for specific ions and the stimuli that control channel opening and closing, such as voltage or ligands. Ion channels are fundamental to the electrical properties of excitable tissues. Dysfunction of channels can lead to abnormal electrical signaling of neurons and muscle cells, accompanied by clinical manifestations, known as channelopathies. Many naturally occurring toxins target ion channels and affect excitable cells where the channels are expressed. Furthermore, ion channels, as membrane proteins and key regulators of a number of physiologic functions, are an important target for drugs in clinical use. In this chapter, we give a general overview of the classification, genetics and structure-function features of the main ion channel families, and address some pharmacologic aspects relevant to neurologic channelopathies.


Subject(s)
Channelopathies , Ion Channels , Humans , Channelopathies/metabolism , Channelopathies/genetics , Ion Channels/metabolism , Animals , Structure-Activity Relationship
4.
Handb Clin Neurol ; 203: 39-58, 2024.
Article in English | MEDLINE | ID: mdl-39174253

ABSTRACT

Periodic paralysis is a rare, dominantly inherited disorder of skeletal muscle in which episodic attacks of weakness are caused by a transient impairment of fiber excitability. Attacks of weakness are often elicited by characteristic environmental triggers, which were the basis for clinically delineating subtypes of periodic paralysis and are an important distinction for optimal disease management. All forms of familial periodic paralysis are caused by mutations of ion channels, often selectively expressed in skeletal muscle, that destabilize the resting potential. The missense mutations usually alter channel function through gain-of-function changes rather than producing a complete loss-of-function null. The knowledge of which channel gene harbors a variant, whether that variant is expected to (or known to) alter function, and how altered function impairs fiber excitability aides in the interpretation of patient signs and symptoms, the interpretation of gene test results, and how to optimize therapeutic intervention for symptom management and improve quality of life.


Subject(s)
Paralyses, Familial Periodic , Humans , Paralyses, Familial Periodic/genetics , Paralyses, Familial Periodic/diagnosis , Paralyses, Familial Periodic/therapy , Mutation/genetics , Ion Channels/genetics , Muscle, Skeletal/physiopathology
5.
Nat Commun ; 15(1): 7023, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174529

ABSTRACT

Neutrophil infiltration and subsequent extracellular trap formation (NETosis) is a contributing factor in sterile inflammation. Furthermore, neutrophil extracellular traps (NETs) are prothrombotic, as they provide a scaffold for platelets and red blood cells to attach to. In circulation, neutrophils are constantly exposed to hemodynamic forces such as shear stress, which in turn regulates many of their biological functions such as crawling and NETosis. However, the mechanisms that mediate mechanotransduction in neutrophils are not fully understood. In this study, we demonstrate that shear stress induces NETosis, dependent on the shear stress level, and increases the sensitivity of neutrophils to NETosis-inducing agents such as adenosine triphosphate and lipopolysaccharides. Furthermore, shear stress increases intracellular calcium levels in neutrophils and this process is mediated by the mechanosensitive ion channel Piezo1. Activation of Piezo1 in response to shear stress mediates calpain activity and cytoskeleton remodeling, which consequently induces NETosis. Thus, activation of Piezo1 in response to shear stress leads to a stepwise sequence of cellular events that mediates NETosis and thereby places neutrophils at the centre of localized inflammation and prothrombotic effects.


Subject(s)
Calcium , Extracellular Traps , Ion Channels , Mechanotransduction, Cellular , Neutrophils , Stress, Mechanical , Neutrophils/metabolism , Ion Channels/metabolism , Ion Channels/genetics , Humans , Extracellular Traps/metabolism , Calcium/metabolism , Adenosine Triphosphate/metabolism , Calpain/metabolism , Lipopolysaccharides/pharmacology , Cytoskeleton/metabolism , Neutrophil Infiltration , Inflammation/metabolism
6.
BMC Neurosci ; 25(1): 37, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174899

ABSTRACT

BACKGROUND: Adipose and muscle tissue wasting outlines the cachectic process during tumor progression. The sympathetic nervous system (SNS) is known to promote tumor progression and research suggests that it might also contribute to cancer-associated cachexia (CAC) energetic expenditure through fat wasting. METHODS: We sympathectomized L5178Y-R tumor-bearing male BALB/c mice by intraperitoneally administering 6-hydroxydopamine to evaluate morphometric, inflammatory, and molecular indicators of CAC and tumor progression. RESULTS: Tumor burden was associated with cachexia indicators, including a 10.5% body mass index (BMI) decrease, 40.19% interscapular, 54% inguinal, and 37.17% visceral adipose tissue loss, a 12% food intake decrease, and significant (p = 0.038 and p = 0.0037) increases in the plasmatic inflammatory cytokines IL-6 and IFN-γ respectively. Sympathectomy of tumor-bearing mice was associated with attenuated BMI and visceral adipose tissue loss, decreased interscapular Ucp-1 gene expression to basal levels, and 2.6-fold reduction in Mmp-9 relative gene expression, as compared with the unsympathectomized mice control group. CONCLUSION: The SNS contributes to CAC-associated morphometric and adipose tissue alterations and promotes tumor progression in a murine model.


Subject(s)
Cachexia , Disease Progression , Mice, Inbred BALB C , Sympathetic Nervous System , Animals , Cachexia/metabolism , Cachexia/pathology , Cachexia/etiology , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Male , Mice , Uncoupling Protein 1/metabolism , Cell Line, Tumor , Ion Channels/metabolism , Matrix Metalloproteinase 9/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Oxidopamine , Sympathectomy, Chemical , Interleukin-6/metabolism , Body Mass Index , Neoplasms/complications , Neoplasms/pathology , Neoplasms/metabolism
7.
J Am Chem Soc ; 146(33): 23230-23239, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39116214

ABSTRACT

TMEM175 is a lysosomal potassium and proton channel that is associated with the development of Parkinson's disease. Advances in understanding the physiological roles of TMEM175 have been hampered by the absence of selective inhibitors, and studies involving genetic perturbations have yielded conflicting results. Here, we report the discovery and characterization of the first reported TMEM175-selective inhibitors, 2-phenylpyridin-4-ylamine (2-PPA), and AP-6. Cryo-EM structures of human TMEM175 bound by 2-PPA and AP-6 reveal that they act as pore blockers, binding at distinct sites in the pore and occluding the ion permeation pathway. Acute inhibition of TMEM175 by 2-PPA or AP-6 increases the level of lysosomal macromolecule catabolism, thereby accelerating macropinocytosis and other digestive processes. These inhibitors may serve as valuable tools to study the roles of TMEM175 in regulating lysosomal function and provide useful templates for future therapeutic development in Parkinson's disease.


Subject(s)
Lysosomes , Parkinson Disease , Humans , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Lysosomes/metabolism , Drug Discovery , Ion Channels/antagonists & inhibitors , Ion Channels/metabolism , Ion Channels/chemistry , Pyridines/chemistry , Pyridines/pharmacology , Models, Molecular , Cryoelectron Microscopy , Potassium Channels
8.
J Exp Med ; 221(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39167075

ABSTRACT

Changes in mechanosensitive ion channels following radiation have seldom been linked to therapeutic sensitivity or specific factors involved in antitumor immunity. Here, in this study, we found that the mechanical force sensor, Piezo2, was significantly upregulated in tumor cells after radiation, and Piezo2 knockout in tumor cells enhanced tumor growth suppression by radiotherapy. Specifically, loss of Piezo2 in tumor cells induced their IL-15 expression via unleashing JAK2/STAT1/IRF-1 axis after radiation. This increase in IL-15 activates IL-15Rα on tumor-infiltrating CD8+ T cells, thereby leading to their augmented effector and stem cell-like properties, along with reduced terminal exhausted feature. Importantly, Piezo2 expression was negatively correlated with CD8 infiltration, as well as with radiosensitivity of patients with rectum adenocarcinoma receiving radiotherapy treatment. Together, our findings reveal that tumor cell-intrinsic Piezo2 induces radioresistance by dampening the IRF-1/IL-15 axis, thus leading to impaired CD8+ T cell-dependent antitumor responses, providing insights into the further development of combination strategies to treat radioresistant cancers.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-15 , Ion Channels , Radiation Tolerance , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Animals , Humans , Ion Channels/metabolism , Ion Channels/genetics , Radiation Tolerance/genetics , Mice , Interleukin-15/metabolism , Interleukin-15/genetics , Cell Line, Tumor , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Mice, Inbred C57BL , Female , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Male , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Signal Transduction
9.
Cell Mol Life Sci ; 81(1): 331, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107572

ABSTRACT

The rising incidences of atherosclerosis have necessitated efforts to identify novel targets for therapeutic interventions. In the present study, we observed increased expression of the mechanosensitive calcium channel Piezo1 transcript in mouse and human atherosclerotic plaques, correlating with infiltration of PIEZO1-expressing macrophages. In vitro administration of Yoda1, a specific agonist for PIEZO1, led to increased foam cell apoptosis and enhanced phagocytosis by macrophages. Mechanistically, PIEZO1 activation resulted in intracellular F-actin rearrangement, elevated mitochondrial ROS levels and induction of mitochondrial fragmentation upon PIEZO1 activation, as well as increased expression of anti-inflammatory genes. In vivo, ApoE-/- mice treated with Yoda1 exhibited regression of atherosclerosis, enhanced stability of advanced lesions, reduced plaque size and necrotic core, increased collagen content, and reduced expression levels of inflammatory markers. Our findings propose PIEZO1 as a novel and potential therapeutic target in atherosclerosis.


Subject(s)
Apoptosis , Atherosclerosis , Foam Cells , Ion Channels , Macrophages , Phagocytosis , Animals , Ion Channels/metabolism , Ion Channels/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Mice , Foam Cells/metabolism , Foam Cells/pathology , Humans , Macrophages/metabolism , Mice, Inbred C57BL , Thiophenes/pharmacology , Male , Reactive Oxygen Species/metabolism , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/genetics , Mitochondria/metabolism , Pyrazines , Thiadiazoles
10.
Channels (Austin) ; 18(1): 2393088, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39169878

ABSTRACT

Mechanosensitive ion channel Piezo1 is known to mediate a variety of inflammatory pathways and is also involved in the occurrence and development of many orthopedic diseases. Although its role in the inflammatory mechanism of knee osteoarthritis (KOA) has been reported, a systematic explanation is yet to be seen. This article aims to summarize the role of inflammatory responses in the pathogenesis of KOA and elucidate the mechanism by which the Piezo1-mediated inflammatory response contributes to the pathogenesis of KOA, providing a theoretical basis for the prevention and treatment of knee osteoarthritis. The results indicate that in the mechanism leading to knee osteoarthritis, Piezo1 can mediate the inflammatory response through chondrocytes and synovial cells, participating in the pathological progression of KOA. Piezo1 has the potential to become a new target for the prevention and treatment of this disease. Additionally, as pain is one of the most severe manifestations in KOA patients, the inflammatory response mediated by Piezo1, which causes the release of inflammatory mediators and pro-inflammatory factors leading to pain, can be further explored.


Subject(s)
Inflammation , Ion Channels , Osteoarthritis, Knee , Ion Channels/metabolism , Humans , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Inflammation/metabolism , Animals , Chondrocytes/metabolism , Mechanotransduction, Cellular
11.
J Am Chem Soc ; 146(33): 22869-22873, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39115272

ABSTRACT

Tubular structures exist broadly in biological systems and exhibit important functions including mediating cellular communications. The construction of artificial analogues in living cells would provide a new strategy for chemotherapy. In this report, a kind of supramolecular channel has been constructed within intercellular gaps by mimicking the assembly process and structure of natural gap junctional channels, which consist of hydrophobic tubular modules located in the adjacent cell membranes and hydrophilic modules within the extracellular space. The assembly of the channels was driven by electrostatic interactions. The channels could inhibit tumor cell invasion by preventing cell migration.


Subject(s)
Cell Movement , Humans , Cell Movement/drug effects , Gap Junctions/metabolism , Hydrophobic and Hydrophilic Interactions , Ion Channels/metabolism , Ion Channels/chemistry , Cell Line, Tumor
12.
Proc Natl Acad Sci U S A ; 121(33): e2403903121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39116127

ABSTRACT

Connexin hemichannels were identified as the first members of the eukaryotic large-pore channel family that mediate permeation of both atomic ions and small molecules between the intracellular and extracellular environments. The conventional view is that their pore is a large passive conduit through which both ions and molecules diffuse in a similar manner. In stark contrast to this notion, we demonstrate that the permeation of ions and of molecules in connexin hemichannels can be uncoupled and differentially regulated. We find that human connexin mutations that produce pathologies and were previously thought to be loss-of-function mutations due to the lack of ionic currents are still capable of mediating the passive transport of molecules with kinetics close to those of wild-type channels. This molecular transport displays saturability in the micromolar range, selectivity, and competitive inhibition, properties that are tuned by specific interactions between the permeating molecules and the N-terminal domain that lies within the pore-a general feature of large-pore channels. We propose that connexin hemichannels and, likely, other large-pore channels, are hybrid channel/transporter-like proteins that might switch between these two modes to promote selective ion conduction or autocrine/paracrine molecular signaling in health and disease processes.


Subject(s)
Connexins , Humans , Connexins/metabolism , Connexins/genetics , Ion Transport , Animals , Mutation , Ions/metabolism , Gap Junctions/metabolism , Ion Channels/metabolism , Ion Channels/genetics
13.
J Gen Physiol ; 156(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39051992

ABSTRACT

Thermosensation requires the activation of a unique collection of ion channels and receptors that work in concert to transmit thermal information. It is widely accepted that transient receptor potential melastatin 8 (TRPM8) activation is required for normal cold sensing; however, recent studies have illuminated major roles for other ion channels in this important somatic sensation. In addition to TRPM8, other TRP channels have been reported to contribute to cold transduction mechanisms in diverse sensory neuron populations, with both leak- and voltage-gated channels being identified for their role in the transmission of cold signals. Whether the same channels that contribute to physiological cold sensing also mediate noxious cold signaling remains unclear; however, recent work has found a conserved role for the kainite receptor, GluK2, in noxious cold sensing across species. Additionally, cold-sensing neurons likely engage in functional crosstalk with nociceptors to give rise to cold pain. This Review will provide an update on our understanding of the relationship between various ion channels in the transduction and transmission of cold and highlight areas where further investigation is required.


Subject(s)
Cold Temperature , Thermosensing , Animals , Humans , Thermosensing/physiology , Ion Channels/metabolism , Signal Transduction/physiology , TRPM Cation Channels/metabolism , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism
14.
Prog Biophys Mol Biol ; 191: 25-39, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971325

ABSTRACT

Cancer is a pernicious and pressing medical problem; moreover, it is a failure of multicellular morphogenesis that sheds much light on evolutionary developmental biology. Numerous classes of pharmacological agents have been considered as cancer therapeutics and evaluated as potential carcinogenic agents; however, these are spread throughout the primary literature. Here, we briefly review recent work on ion channel drugs as promising anti-cancer treatments and present a systematic review of the known cancer-relevant effects of 109 drugs targeting ion channels. The roles of ion channels in cancer are consistent with the importance of bioelectrical parameters in cell regulation and with the functions of bioelectric signaling in morphogenetic signals that act as cancer suppressors. We find that compounds that are well-known for having targets in the nervous system, such as voltage-gated ion channels, ligand-gated ion channels, proton pumps, and gap junctions are especially relevant to cancer. Our review suggests further opportunities for the repurposing of numerous promising candidates in the field of cancer electroceuticals.


Subject(s)
Ion Channels , Neoplasms , Phenotype , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Ion Channels/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Animals , Electrophysiological Phenomena/drug effects
15.
PLoS Genet ; 20(7): e1011339, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38980841

ABSTRACT

BACKGROUND: Varicose veins (VV) are one of the common human diseases, but the role of genetics in its development is not fully understood. METHODS: We conducted an exome-wide association study of VV using whole-exome sequencing data from the UK Biobank, and focused on common and rare variants using single-variant association analysis and gene-level collapsing analysis. FINDINGS: A total of 13,823,269 autosomal genetic variants were obtained after quality control. We identified 36 VV-related independent common variants mapping to 34 genes by single-variant analysis and three rare variant genes (PIEZO1, ECE1, FBLN7) by collapsing analysis, and most associations between genes and VV were replicated in FinnGen. PIEZO1 was the closest gene associated with VV (P = 5.05 × 10-31), and it was found to reach exome-wide significance in both single-variant and collapsing analyses. Two novel rare variant genes (ECE1 and METTL21A) associated with VV were identified, of which METTL21A was associated only with females. The pleiotropic effects of VV-related genes suggested that body size, inflammation, and pulmonary function are strongly associated with the development of VV. CONCLUSIONS: Our findings highlight the importance of causal genes for VV and provide new directions for treatment.


Subject(s)
Exome Sequencing , Exome , Genetic Predisposition to Disease , Genome-Wide Association Study , Varicose Veins , Humans , Varicose Veins/genetics , Female , Male , Exome/genetics , Polymorphism, Single Nucleotide , Endothelin-Converting Enzymes/genetics , Middle Aged , Genetic Variation , Adult , Ion Channels
16.
Methods Mol Biol ; 2836: 219-233, 2024.
Article in English | MEDLINE | ID: mdl-38995543

ABSTRACT

Channels, tunnels, and pores serve as pathways for the transport of molecules and ions through protein structures, thus participating to their functions. MOLEonline ( https://mole.upol.cz ) is an interactive web-based tool with enhanced capabilities for detecting and characterizing channels, tunnels, and pores within protein structures. MOLEonline has two distinct calculation modes for analysis of channel and tunnels or transmembrane pores. This application gives researchers rich analytical insights into channel detection, structural characterization, and physicochemical properties. ChannelsDB 2.0 ( https://channelsdb2.biodata.ceitec.cz/ ) is a comprehensive database that offers information on the location, geometry, and physicochemical characteristics of tunnels and pores within macromolecular structures deposited in Protein Data Bank and AlphaFill databases. These tunnels are sourced from manual deposition from literature and automatic detection using software tools MOLE and CAVER. MOLEonline and ChannelsDB visualization is powered by the LiteMol Viewer and Mol* viewer, ensuring a user-friendly workspace. This chapter provides an overview of user applications and usage.


Subject(s)
Databases, Protein , Software , Protein Conformation , User-Computer Interface , Models, Molecular , Ion Channels/metabolism , Ion Channels/chemistry , Computational Biology/methods , Proteins/chemistry , Proteins/metabolism , Web Browser
17.
Nat Commun ; 15(1): 5521, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951553

ABSTRACT

The microgeometry of the cellular microenvironment profoundly impacts cellular behaviors, yet the link between it and the ubiquitously expressed mechanosensitive ion channel PIEZO1 remains unclear. Herein, we describe a fluorescent micropipette aspiration assay that allows for simultaneous visualization of intracellular calcium dynamics and cytoskeletal architecture in real-time, under varied micropipette geometries. By integrating elastic shell finite element analysis with fluorescent lifetime imaging microscopy and employing PIEZO1-specific transgenic red blood cells and HEK cell lines, we demonstrate a direct correlation between the microscale geometry of aspiration and PIEZO1-mediated calcium signaling. We reveal that increased micropipette tip angles and physical constrictions lead to a significant reorganization of F-actin, accumulation at the aspirated cell neck, and subsequently amplify the tension stress at the dome of the cell to induce more PIEZO1's activity. Disruption of the F-actin network or inhibition of its mobility leads to a notable decline in PIEZO1 mediated calcium influx, underscoring its critical role in cellular mechanosensing amidst geometrical constraints.


Subject(s)
Actins , Calcium , Cytoskeleton , Ion Channels , Mechanotransduction, Cellular , Humans , Ion Channels/metabolism , Actins/metabolism , HEK293 Cells , Cytoskeleton/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Finite Element Analysis , Animals , Microscopy, Fluorescence/methods
18.
Biomolecules ; 14(7)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39062518

ABSTRACT

The gastrointestinal (GI) tract is an organ actively involved in mechanical processes, where it detects forces via a mechanosensation mechanism. Mechanosensation relies on specialized cells termed mechanoreceptors, which convert mechanical forces into electrochemical signals via mechanosensors. The mechanosensitive Piezo1 and Piezo2 are widely expressed in various mechanosensitive cells that respond to GI mechanical forces by altering transmembrane ionic currents, such as epithelial cells, enterochromaffin cells, and intrinsic and extrinsic enteric neurons. This review highlights recent research advances on mechanosensitive Piezo channels in GI physiology and pathology. Specifically, the latest insights on the role of Piezo channels in the intestinal barrier, GI motility, and intestinal mechanosensation are summarized. Additionally, an overview of Piezo channels in the pathogenesis of GI disorders, including irritable bowel syndrome, inflammatory bowel disease, and GI cancers, is provided. Overall, the presence of mechanosensitive Piezo channels offers a promising new perspective for the treatment of various GI disorders.


Subject(s)
Gastrointestinal Tract , Ion Channels , Mechanotransduction, Cellular , Humans , Ion Channels/metabolism , Animals , Gastrointestinal Tract/metabolism , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/pathology , Gastrointestinal Motility/physiology
19.
Bioorg Med Chem ; 110: 117839, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39029438

ABSTRACT

Peptaibols are a class of short peptides, typically 7 to 20 amino acids long, characterized by noncanonical amino acid residues such as aminoisobutyric acid (Aib). Although the helix length is shorter than the membrane thickness, the 11-residue peptaibol trichorovin-XII (TV-XII) can form ion channels in membranes. Assuming that a higher proportion of isoleucine (Ile) relative to leucine (Leu) residues is crucial for maintaining the ion channel activity of TV-XII, peptide analogs of TV-XII with varying Ile content were designed, synthesized, and evaluated. The secondary structure of all derivatives under hydrophobic conditions was confirmed by CD measurement as an α-helix-like ß-bend ribbon spiral structure. The most stable ion channel activity was found in compound 4a with maximum Ile. Furthermore, the C-terminal Ile analog showed greater ion channel activity compared to the Leu analog. This suggests that the choice between Leu and Ile can influence the expression of ion channel activity, which will be crucial for the de novo designed functional peptides.


Subject(s)
Isoleucine , Peptaibols , Isoleucine/chemistry , Isoleucine/analogs & derivatives , Peptaibols/chemistry , Peptaibols/pharmacology , Peptaibols/chemical synthesis , Ion Channels/metabolism , Ion Channels/chemistry , Protein Structure, Secondary , Structure-Activity Relationship , Amino Acid Sequence , Circular Dichroism
SELECTION OF CITATIONS
SEARCH DETAIL