A variety of neuromuscular conditions resulting from MUTATIONS in ION CHANNELS manifesting as episodes of EPILEPSY; HEADACHE DISORDERS; and DYSKINESIAS.
A heterogenous group of inherited disorders characterized by recurring attacks of rapidly progressive flaccid paralysis or myotonia. These conditions have in common a mutation of the gene encoding the alpha subunit of the sodium channel in skeletal muscle. They are frequently associated with fluctuations in serum potassium levels. Periodic paralysis may also occur as a non-familial process secondary to THYROTOXICOSIS and other conditions. (From Adams et al., Principles of Neurology, 6th ed, p1481)
An autosomal dominant familial disorder characterized by recurrent episodes of skeletal muscle weakness associated with falls in serum potassium levels. The condition usually presents in the first or second decade of life with attacks of trunk and leg paresis during sleep or shortly after awakening. Symptoms may persist for hours to days and generally are precipitated by exercise or a meal high in carbohydrates. (Adams et al., Principles of Neurology, 6th ed, p1483)
Diseases characterized by MYOTONIA, which may be inherited or acquired. Myotonia may be restricted to certain muscles (e.g., intrinsic hand muscles) or occur as a generalized condition.
Prolonged failure of muscle relaxation after contraction. This may occur after voluntary contractions, muscle percussion, or electrical stimulation of the muscle. Myotonia is a characteristic feature of MYOTONIC DISORDERS.
An autosomal dominant familial disorder which presents in infancy or childhood and is characterized by episodes of weakness associated with hyperkalemia. During attacks, muscles of the lower extremities are initially affected, followed by the lower trunk and arms. Episodes last from 15-60 minutes and typically occur after a period of rest following exercise. A defect in skeletal muscle sodium channels has been identified as the cause of this condition. Normokalemic periodic paralysis is a closely related disorder marked by a lack of alterations in potassium levels during attacks of weakness. (Adams et al., Principles of Neurology, 6th ed, p1481)
A rare neuromuscular disorder with onset usually in late childhood or early adulthood, characterized by intermittent or continuous widespread involuntary muscle contractions; FASCICULATION; hyporeflexia; MUSCLE CRAMP; MUSCLE WEAKNESS; HYPERHIDROSIS; TACHYCARDIA; and MYOKYMIA. Involvement of pharyngeal or laryngeal muscles may interfere with speech and breathing. The continuous motor activity persists during sleep and general anesthesia (distinguishing this condition from STIFF-PERSON SYNDROME). Familial and acquired (primarily autoimmune) forms have been reported. (From Ann NY Acad Sci 1998 May 13;841:482-496; Adams et al., Principles of Neurology, 6th ed, p1491)
Acquired, familial, and congenital disorders of SKELETAL MUSCLE and SMOOTH MUSCLE.
A condition that is characterized by episodes of fainting (SYNCOPE) and varying degree of ventricular arrhythmia as indicated by the prolonged QT interval. The inherited forms are caused by mutation of genes encoding cardiac ion channel proteins. The two major forms are ROMANO-WARD SYNDROME and JERVELL-LANGE NIELSEN SYNDROME.
A voltage-gated sodium channel subtype that mediates the sodium ion PERMEABILITY of CARDIOMYOCYTES. Defects in the SCN5A gene, which codes for the alpha subunit of this sodium channel, are associated with a variety of CARDIAC DISEASES that result from loss of sodium channel function.
A voltage-gated sodium channel subtype that mediates the sodium ion PERMEABILITY of SKELETAL MYOCYTES. Defects in the SCN4A gene, which codes for the alpha subunit of this sodium channel, are associated with several MYOTONIC DISORDERS.
Ion channels that specifically allow the passage of SODIUM ions. A variety of specific sodium channel subtypes are involved in serving specialized functions such as neuronal signaling, CARDIAC MUSCLE contraction, and KIDNEY function.
An autosomal dominant defect of cardiac conduction that is characterized by an abnormal ST-segment in leads V1-V3 on the ELECTROCARDIOGRAM resembling a right BUNDLE-BRANCH BLOCK; high risk of VENTRICULAR TACHYCARDIA; or VENTRICULAR FIBRILLATION; SYNCOPAL EPISODE; and possible sudden death. This syndrome is linked to mutations of gene encoding the cardiac SODIUM CHANNEL alpha subunit.
An inherited congenital myopathic condition characterized by weakness and hypotonia in infancy and delayed motor development. Muscle biopsy reveals a condensation of myofibrils and myofibrillar material in the central portion of each muscle fiber. (Adams et al., Principles of Neurology, 6th ed, p1452)
Diseases of the central and peripheral nervous system. This includes disorders of the brain, spinal cord, cranial nerves, peripheral nerves, nerve roots, autonomic nervous system, neuromuscular junction, and muscle.
Gated, ion-selective glycoproteins that traverse membranes. The stimulus for ION CHANNEL GATING can be due to a variety of stimuli such as LIGANDS, a TRANSMEMBRANE POTENTIAL DIFFERENCE, mechanical deformation or through INTRACELLULAR SIGNALING PEPTIDES AND PROTEINS.
The abrupt and unexplained death of an apparently healthy infant under one year of age, remaining unexplained after a thorough case investigation, including performance of a complete autopsy, examination of the death scene, and review of the clinical history. (Pediatr Pathol 1991 Sep-Oct;11(5):677-84)
Diseases that are caused by genetic mutations present during embryo or fetal development, although they may be observed later in life. The mutations may be inherited from a parent's genome or they may be acquired in utero.
Any disturbances of the normal rhythmic beating of the heart or MYOCARDIAL CONTRACTION. Cardiac arrhythmias can be classified by the abnormalities in HEART RATE, disorders of electrical impulse generation, or impulse conduction.
Rapid and excessive rise of temperature accompanied by muscular rigidity following general anesthesia.
Any detectable and heritable change in the genetic material that causes a change in the GENOTYPE and which is transmitted to daughter cells and to succeeding generations.
Potassium channels where the flow of K+ ions into the cell is greater than the outward flow.
The opening and closing of ion channels due to a stimulus. The stimulus can be a change in membrane potential (voltage-gated), drugs or chemical transmitters (ligand-gated), or a mechanical deformation. Gating is thought to involve conformational changes of the ion channel which alters selective permeability.
Potassium channel whose permeability to ions is extremely sensitive to the transmembrane potential difference. The opening of these channels is induced by the membrane depolarization of the ACTION POTENTIAL.
Unexpected rapid natural death due to cardiovascular collapse within one hour of initial symptoms. It is usually caused by the worsening of existing heart diseases. The sudden onset of symptoms, such as CHEST PAIN and CARDIAC ARRHYTHMIAS, particularly VENTRICULAR TACHYCARDIA, can lead to the loss of consciousness and cardiac arrest followed by biological death. (from Braunwald's Heart Disease: A Textbook of Cardiovascular Medicine, 7th ed., 2005)
Voltage-dependent cell membrane glycoproteins selectively permeable to calcium ions. They are categorized as L-, T-, N-, P-, Q-, and R-types based on the activation and inactivation kinetics, ion specificity, and sensitivity to drugs and toxins. The L- and T-types are present throughout the cardiovascular and central nervous systems and the N-, P-, Q-, & R-types are located in neuronal tissue.
An alkaloid found in the root of RAUWOLFIA SERPENTINA, among other plant sources. It is a class Ia antiarrhythmic agent that apparently acts by changing the shape and threshold of cardiac action potentials.
Recording of the moment-to-moment electromotive forces of the HEART as projected onto various sites on the body's surface, delineated as a scalar function of time. The recording is monitored by a tracing on slow moving chart paper or by observing it on a cardioscope, which is a CATHODE RAY TUBE DISPLAY.
Persistent and disabling ANXIETY.
Formed by the articulation of the talus with the calcaneus.
A form of heart block in which the electrical stimulation of HEART VENTRICLES is interrupted at either one of the branches of BUNDLE OF HIS thus preventing the simultaneous depolarization of the two ventricles.

Molecular basis of inherited calcium channelopathies: role of mutations in pore-forming subunits. (1/102)

The pore-forming alpha subunits of voltage-gated calcium channels contain the essential biophysical machinery that underlies calcium influx in response to cell depolarization. In combination with requisite auxiliary subunits, these pore subunits form calcium channel complexes that are pivotal to the physiology and pharmacology of diverse cells ranging from sperm to neurons. Not surprisingly, mutations in the pore subunits generate diverse pathologies, termed channelopathies, that range from failures in excitation-contraction coupling to night blindness. Over the last decade, major insights into the mechanisms of pathogenesis have been derived from animals showing spontaneous or induced mutations. In parallel, there has been considerable growth in our understanding of the workings of voltage-gated ion channels from a structure-function, regulation and cell biology perspective. Here we document our current understanding of the mutations underlying channelopathies involving the voltage-gated calcium channel alpha subunits in humans and other species.  (+info)

Voltage-gated calcium channels in genetic diseases. (2/102)

Voltage-gated calcium channels (VGCCs) mediate calcium entry into excitable cells in response to membrane depolarization. During the past decade, our understanding of the gating and functions of VGCCs has been illuminated by the analysis of mutations linked to a heterogeneous group of genetic diseases called "calcium channelopathies". Calcium channelopathies include muscular, neurological, cardiac and vision syndromes. Recent data suggest that calcium channelopathies result not only from electrophysiological defects but also from altered alpha(1)/Ca(V) subunit protein processing, including folding, posttranslational modifications, quality control and trafficking abnormalities. Overall, functional analyses of VGCC mutations provide a more comprehensive view of the corresponding human disorders and offer important new insights into VGCC function. Ultimately, the understanding of these pathogenic channel mutations should lead to improved treatments of such hereditary diseases in humans.  (+info)

Emerging perspectives in store-operated Ca2+ entry: roles of Orai, Stim and TRP. (3/102)

Depletion of intracellular Ca2+ stores induces Ca2+ influx across the plasma membrane through store-operated channels (SOCs). This store-operated Ca2+ influx is important for the replenishment of the Ca2+ stores, and is also involved in many signaling processes by virtue of the ability of intracellular Ca2+ to act as a second messenger. For many years, the molecular identities of particular SOCs, as well as the signaling mechanisms by which these channels are activated, have been elusive. Recently, however, the mammalian proteins STIM1 and Orai1 were shown to be necessary for the activation of store-operated Ca2+ entry in a variety of mammalian cells. Here we present molecular, pharmacological, and electrophysiological properties of SOCs, with particular focus on the roles that STIM1 and Orai1 may play in the signaling processes that regulate various pathways of store-operated entry.  (+info)

Chloride channelopathy in myotonic dystrophy resulting from loss of posttranscriptional regulation for CLCN1. (4/102)

Transmembrane chloride ion conductance in skeletal muscle increases during early postnatal development. A transgenic mouse model of myotonic dystrophy type 1 (DM1) displays decreased sarcolemmal chloride conductance. Both effects result from modulation of chloride channel 1 (CLCN1) expression, but the respective contributions of transcriptional vs. posttranscriptional regulation are unknown. Here we show that alternative splicing of CLCN1 undergoes a physiological splicing transition during the first 3 wk of postnatal life in mice. During this interval, there is a switch to production of CLCN1 splice products having an intact reading frame, an upregulation of CLCN1 mRNA encoding full-length channel protein, and an increase of CLCN1 function, as determined by patch-clamp analysis of single muscle fibers. In a transgenic mouse model of DM1, however, the splicing transition does not occur, CLCN1 channel function remains low throughout the postnatal interval, and muscle fibers display myotonic discharges. Thus alternative splicing is a posttranscriptional mechanism regulating chloride conductance during muscle development, and the chloride channelopathy in a transgenic mouse model of DM1 results from a failure to execute a splicing transition for CLCN1.  (+info)

TRPpathies. (5/102)

Many human diseases are caused by mutations in ion channels. Dissecting the pathogenesis of these 'channelopathies' has yielded important insights into the regulation of vital biological processes by ions and has become a productive tool of modern ion channel biology. One of the best examples of a synergism between the clinical and basic science aspects of a modern biological topic is cystic fibrosis. Not only did the identification of the ion channel mutated in cystic fibrosis pinpoint the root cause of this disease, but it also has significantly advanced our understanding of basic biological processes as diverse as protein folding and epithelial fluid and electrolyte secretion. The list of confirmed 'channelopathies' is growing and several members of the TRP family of ion channels have been implicated in human diseases such as mucolipidosis type IV (MLIV), autosomal dominant polycystic kidney disease (ADPKD), familial focal segmental glomerulosclerosis (FSG), hypomagnesemia with secondary hypocalcaemia (HSH), and several forms of cancer. Analysing pathogenesis of the diseases linked to TRP dysregulation provides an exciting means of identifying novel functions of TRP channels.  (+info)

Transient receptor potential cation channels in disease. (6/102)

The transient receptor potential (TRP) superfamily consists of a large number of cation channels that are mostly permeable to both monovalent and divalent cations. The 28 mammalian TRP channels can be subdivided into six main subfamilies: the TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin), TRPML (mucolipin), and the TRPA (ankyrin) groups. TRP channels are expressed in almost every tissue and cell type and play an important role in the regulation of various cell functions. Currently, significant scientific effort is being devoted to understanding the physiology of TRP channels and their relationship to human diseases. At this point, only a few channelopathies in which defects in TRP genes are the direct cause of cellular dysfunction have been identified. In addition, mapping of TRP genes to susceptible chromosome regions (e.g., translocations, breakpoint intervals, increased frequency of polymorphisms) has been considered suggestive of the involvement of these channels in hereditary diseases. Moreover, strong indications of the involvement of TRP channels in several diseases come from correlations between levels of channel expression and disease symptoms. Finally, TRP channels are involved in some systemic diseases due to their role as targets for irritants, inflammation products, and xenobiotic toxins. The analysis of transgenic models allows further extrapolations of TRP channel deficiency to human physiology and disease. In this review, we provide an overview of the impact of TRP channels on the pathogenesis of several diseases and identify several TRPs for which a causal pathogenic role might be anticipated.  (+info)

KATP channel mutation confers risk for vein of Marshall adrenergic atrial fibrillation. (7/102)

BACKGROUND: A 53-year-old female presented with a 10-year history of paroxysmal atrial fibrillation (AF), precipitated by activity and refractory to medical therapy. In the absence of traditional risk factors for disease, a genetic defect in electrical homeostasis underlying stress-induced AF was explored. INVESTIGATIONS: Echocardiography, cardiac perfusion stress imaging, invasive electrophysiology with isoproterenol provocation, genomic DNA sequencing of K(ATP) channel genes, exclusion of mutation in 2,000 individuals free of AF, reconstitution of channel defect with molecular phenotyping, and verification of pathogenic link in targeted knockout. DIAGNOSIS: K(ATP) channelopathy caused by missense mutation (Thr1547Ile) of the ABCC9 gene conferring predisposition to adrenergic AF originating from the vein of Marshall. MANAGEMENT: Disruption of arrhythmogenic gene-environment substrate at the vein of Marshall by radiofrequency ablation.  (+info)

TRP channels in disease. (8/102)

"Transient receptor potential" cation channels (TRP channels) play a unique role as cell sensors, are involved in a plethora of Ca(2+)-mediated cell functions, and play a role as "gate-keepers" in many homeostatic processes such as Ca(2+) and Mg(2+) reabsorption. The variety of functions to which TRP channels contribute and the polymodal character of their activation predict that failures in correct channel gating or permeation will likely contribute to complex pathophysiological mechanisms. Dysfunctions of TRPs cause human diseases but are also involved in a complex manner to contribute and determine the progress of several diseases. Contributions to this special issue discuss channelopathias for which mutations in TRP channels that induce "loss-" or "gain-of-function" of the channel and can be considered "disease-causing" have been identified. The role of TRPs will be further elucidated in complex diseases of the intestinal, renal, urogenital, respiratory, and cardiovascular systems. Finally, the role of TRPs will be discussed in neuronal diseases and neurodegenerative disorders.  (+info)

Channelopathies are genetic disorders that are caused by mutations in the genes that encode for ion channels. Ion channels are specialized proteins that regulate the flow of ions, such as sodium, potassium, and calcium, across cell membranes. These ion channels play a crucial role in various physiological processes, including the generation and transmission of electrical signals in the body.

Channelopathies can affect various organs and systems in the body, depending on the type of ion channel that is affected. For example, mutations in sodium channel genes can cause neuromuscular disorders such as epilepsy, migraine, and periodic paralysis. Mutations in potassium channel genes can cause cardiac arrhythmias, while mutations in calcium channel genes can cause neurological disorders such as episodic ataxia and hemiplegic migraine.

The symptoms of channelopathies can vary widely depending on the specific disorder and the severity of the mutation. Treatment typically involves managing the symptoms and may include medications, lifestyle modifications, or in some cases, surgery.

Familial periodic paralysis is a group of rare genetic disorders characterized by episodes of muscle weakness or paralysis that recur over time. There are several types of familial periodic paralysis, including hypokalemic periodic paralysis, hyperkalemic periodic paralysis, and normokalemic periodic paralysis, each with its own specific genetic cause and pattern of symptoms.

In general, these disorders are caused by mutations in genes that regulate ion channels in muscle cells, leading to abnormalities in the flow of ions such as potassium in and out of the cells. This can result in changes in muscle excitability and contractility, causing episodes of weakness or paralysis.

The episodes of paralysis in familial periodic paralysis can vary in frequency, duration, and severity. They may be triggered by factors such as rest after exercise, cold or hot temperatures, emotional stress, alcohol consumption, or certain medications. During an episode, the affected muscles may become weak or completely paralyzed, often affecting the limbs but sometimes also involving the muscles of the face, throat, and trunk.

Familial periodic paralysis is typically inherited in an autosomal dominant pattern, meaning that a child has a 50% chance of inheriting the disorder if one parent is affected. However, some cases may arise from new mutations in the affected gene and occur in people with no family history of the disorder.

Treatment for familial periodic paralysis typically involves avoiding triggers and managing symptoms during episodes. In some cases, medications such as potassium-binding agents or diuretics may be used to help prevent or reduce the severity of episodes. Lifestyle modifications, such as a low-carbohydrate or high-sodium diet, may also be recommended in some cases.

Hypokalemic Periodic Paralysis (HPP) is a group of rare inherited disorders characterized by episodes of muscle weakness or paralysis, often associated with low potassium levels in the blood (hypokalemia). During an attack, muscles may become weak or fully paralyzed, typically affecting the legs and arms. The episodes can last from several hours to days. HPP is caused by genetic mutations that affect ion channels in muscle cells, leading to an imbalance of electrolytes and impaired muscle function. There are two main types: primary (or classic) HPP and secondary HPP. Primary HPP is further divided into thyrotoxic HPP and normokalemic HPP. Secondary HPP can be caused by various factors, such as medications or underlying medical conditions that cause hypokalemia.

Myotonic disorders are a group of genetic muscle diseases characterized by the inability to relax muscles (myotonia) after contraction. Myotonia can cause symptoms such as stiffness, muscle spasms, and prolonged muscle contractions or cramps. These disorders may also be associated with other symptoms, including muscle weakness, wasting, and various systemic features.

The most common myotonic disorder is myotonic dystrophy type 1 (DM1), which is caused by a mutation in the DMPK gene. Myotonic dystrophy type 2 (DM2) is another form of myotonic dystrophy, resulting from a mutation in the CNBP gene. These two forms of myotonic dystrophy have distinct genetic causes but share similar clinical features, such as myotonia and muscle weakness.

Other less common myotonic disorders include:

1. Myotonia congenita - A group of inherited conditions characterized by muscle stiffness from birth or early childhood. There are two main types: Thomsen's disease (autosomal dominant) and Becker's disease (autosomal recessive).
2. Paramyotonia congenita - An autosomal dominant disorder characterized by muscle stiffness triggered by cold temperatures or physical exertion.
3. Potassium-aggravated myotonia (PAM) - A rare, autosomal dominant condition with symptoms similar to paramyotonia congenita but without the cold sensitivity.
4. Myotonia fluctuans - A rare, autosomal dominant disorder characterized by fluctuating muscle stiffness and cramps.
5. Acquired myotonia - Rare cases of myotonia caused by factors other than genetic mutations, such as medication side effects or underlying medical conditions.

Myotonic disorders can significantly impact a person's quality of life, making daily activities challenging. Proper diagnosis and management are essential to help alleviate symptoms and improve overall well-being.

Myotonia is a condition characterized by the delayed relaxation of a muscle after voluntary contraction or electrical stimulation, resulting in stiffness or difficulty with relaxing the muscles. It's often associated with certain neuromuscular disorders such as myotonic dystrophy and myotonia congenita. The prolonged muscle contraction can cause stiffness, especially after periods of rest, and may improve with repeated contractions (warm-up phenomenon).

Hyperkalemic periodic paralysis (HypPK) is a rare genetic disorder characterized by episodes of muscle weakness or paralysis due to an abnormality in the ion channels that regulate the movement of potassium into and out of muscle fibers. This results in an excessive accumulation of potassium in the blood (hyperkalemia) during attacks, which can interfere with the ability of nerve cells to communicate with muscles and cause temporary muscle weakness or paralysis.

HypPK is caused by mutations in the SCN4A gene, which encodes a sodium channel protein found in skeletal muscle. These genetic changes disrupt the normal functioning of the ion channels, leading to an imbalance in the flow of ions across the muscle cell membrane and affecting muscle excitability.

Episodes of paralysis in HypPK typically begin in childhood or adolescence and can last from several hours to days. Triggers for attacks may include rest following exercise, cold or hot weather, stress, alcohol consumption, infection, or certain medications. Between episodes, individuals with HypPK usually have normal muscle strength and function.

Management of HypPK includes avoiding triggers, maintaining a low-potassium diet, and using medications to prevent or treat attacks. Medications such as thiazide diuretics, acetazolamide, or dichlorphenamide can help lower potassium levels in the blood and reduce the frequency and severity of episodes. In some cases, intravenous glucose and insulin may be used to drive potassium into cells during an attack, thereby reducing its concentration in the blood and alleviating symptoms.

Isaac's syndrome, also known as neuromyotonia, is a rare neurological disorder characterized by continuous muscle fiber activity leading to stiffness, cramps, and delayed relaxation after contraction. This condition results from hyperexcitability of the peripheral nerves due to dysfunction of voltage-gated potassium channels.

The symptoms may include:

1. Muscle stiffness (rigidity)
2. Muscle twitching or cramping (myokymia)
3. Delayed relaxation after contraction (percussion myotonia)
4. Involuntary muscle activity (neuromyotonia)
5. Hyperhidrosis (excessive sweating)
6. Paresthesias (abnormal sensations)

Isaac's syndrome can be associated with other conditions, such as autoimmune disorders, paraneoplastic syndromes, or genetic factors. The diagnosis typically involves clinical examination, electromyography (EMG), and nerve conduction studies. Treatment options may include medications that reduce neuronal excitability, such as anticonvulsants, plasma exchange, or intravenous immunoglobulin therapy.

Muscular diseases, also known as myopathies, refer to a group of conditions that affect the functionality and health of muscle tissue. These diseases can be inherited or acquired and may result from inflammation, infection, injury, or degenerative processes. They can cause symptoms such as weakness, stiffness, cramping, spasms, wasting, and loss of muscle function.

Examples of muscular diseases include:

1. Duchenne Muscular Dystrophy (DMD): A genetic disorder that results in progressive muscle weakness and degeneration due to a lack of dystrophin protein.
2. Myasthenia Gravis: An autoimmune disease that causes muscle weakness and fatigue, typically affecting the eyes and face, throat, and limbs.
3. Inclusion Body Myositis (IBM): A progressive muscle disorder characterized by muscle inflammation and wasting, typically affecting older adults.
4. Polymyositis: An inflammatory myopathy that causes muscle weakness and inflammation throughout the body.
5. Metabolic Myopathies: A group of inherited disorders that affect muscle metabolism, leading to exercise intolerance, muscle weakness, and other symptoms.
6. Muscular Dystonias: Involuntary muscle contractions and spasms that can cause abnormal postures or movements.

It is important to note that muscular diseases can have a significant impact on an individual's quality of life, mobility, and overall health. Proper diagnosis and treatment are crucial for managing symptoms and improving outcomes.

Long QT syndrome (LQTS) is a cardiac electrical disorder characterized by a prolonged QT interval on the electrocardiogram (ECG), which can potentially trigger rapid, chaotic heartbeats known as ventricular tachyarrhythmias, such as torsades de pointes. These arrhythmias can be life-threatening and lead to syncope (fainting) or sudden cardiac death. LQTS is often congenital but may also be acquired due to certain medications, medical conditions, or electrolyte imbalances. It's essential to identify and manage LQTS promptly to reduce the risk of severe complications.

NAV1.5, also known as SCN5A, is a specific type of voltage-gated sodium channel found in the heart muscle cells (cardiomyocytes). These channels play a crucial role in the generation and transmission of electrical signals that coordinate the contraction of the heart.

More specifically, NAV1.5 channels are responsible for the rapid influx of sodium ions into cardiomyocytes during the initial phase of the action potential, which is the electrical excitation of the cell. This rapid influx of sodium ions helps to initiate and propagate the action potential throughout the heart muscle, allowing for coordinated contraction and proper heart function.

Mutations in the SCN5A gene, which encodes the NAV1.5 channel, have been associated with various cardiac arrhythmias, including long QT syndrome, Brugada syndrome, and familial atrial fibrillation, among others. These genetic disorders can lead to abnormal heart rhythms, syncope, and in some cases, sudden cardiac death.

NAV1.4, also known as SCN4A, is a gene that encodes for the α subunit of the voltage-gated sodium channel in humans. This channel, specifically located in the skeletal muscle, is responsible for the rapid influx of sodium ions during the initiation and propagation of action potentials, which are critical for muscle contraction.

The NAV1.4 Voltage-Gated Sodium Channel plays a crucial role in the functioning of skeletal muscles. Mutations in this gene can lead to various neuromuscular disorders such as hyperkalemic periodic paralysis, paramyotonia congenita, and potassium-aggravated myotonia, which are characterized by muscle stiffness, cramps, and episodes of weakness or paralysis.

Sodium channels are specialized protein structures that are embedded in the membranes of excitable cells, such as nerve and muscle cells. They play a crucial role in the generation and transmission of electrical signals in these cells. Sodium channels are responsible for the rapid influx of sodium ions into the cell during the initial phase of an action potential, which is the electrical signal that travels along the membrane of a neuron or muscle fiber. This sudden influx of sodium ions causes the membrane potential to rapidly reverse, leading to the depolarization of the cell. After the action potential, the sodium channels close and become inactivated, preventing further entry of sodium ions and helping to restore the resting membrane potential.

Sodium channels are composed of a large alpha subunit and one or two smaller beta subunits. The alpha subunit forms the ion-conducting pore, while the beta subunits play a role in modulating the function and stability of the channel. Mutations in sodium channel genes have been associated with various inherited diseases, including certain forms of epilepsy, cardiac arrhythmias, and muscle disorders.

Brugada Syndrome is a genetic disorder characterized by abnormal electrocardiogram (ECG) findings and an increased risk of sudden cardiac death. It is typically caused by a mutation in the SCN5A gene, which encodes for a sodium channel protein in the heart. This mutation can lead to abnormal ion transport in the heart cells, causing changes in the electrical activity of the heart that can trigger dangerous arrhythmias.

The ECG findings associated with Brugada Syndrome include a distinct pattern of ST-segment elevation in the right precordial leads (V1-V3), which can appear spontaneously or be induced by certain medications. The syndrome is often classified into two types based on the presence or absence of symptoms:

* Type 1 Brugada Syndrome: This type is characterized by a coved-type ST-segment elevation of at least 2 mm in height in at least one right precordial lead, with a negative T wave. This pattern must be present to make the diagnosis, and it should not be transient or induced by any medication or condition. Type 1 Brugada Syndrome is associated with a higher risk of sudden cardiac death.
* Type 2 Brugada Syndrome: This type is characterized by a saddleback-type ST-segment elevation of at least 2 mm in height in at least one right precordial lead, with a positive or biphasic T wave. The ST segment should return to the baseline level or below within 0.08 seconds after the J point (the junction between the QRS complex and the ST segment). Type 2 Brugada Syndrome is associated with a lower risk of sudden cardiac death compared to Type 1, but it can still pose a significant risk in some individuals.

Brugada Syndrome can affect people of any age, gender, or ethnicity, although it is more commonly diagnosed in middle-aged men of Asian descent. The syndrome can be inherited in an autosomal dominant manner, meaning that a child has a 50% chance of inheriting the mutation from a parent who carries the gene. However, not all individuals with the genetic mutation will develop symptoms or have abnormal ECG findings.

Treatment for Brugada Syndrome typically involves implanting a cardioverter-defibrillator (ICD) to prevent sudden cardiac death. Medications such as quinidine or isoproterenol may also be used to reduce the risk of arrhythmias. Lifestyle modifications, such as avoiding alcohol and certain medications that can trigger arrhythmias, may also be recommended.

Central core myopathy is a rare genetic muscle disorder that is typically present at birth or appears in early childhood. It is characterized by the presence of distinctive rod-like structures, called cores, in the center of the muscle fibers. These cores are devoid of normal mitochondria and other organelles, which can lead to muscle weakness and wasting.

Central core myopathy is often associated with mutations in the ryanodine receptor 1 (RYR1) gene, which provides instructions for making a protein that plays a critical role in calcium signaling within muscles. Abnormalities in calcium signaling can lead to muscle weakness and wasting.

The symptoms of central core myopathy can vary widely, even among members of the same family with the same genetic mutation. Some people with this condition may have only mild muscle weakness, while others may be severely affected and have difficulty walking or performing other physical activities. The condition typically does not worsen over time, and life expectancy is usually normal. However, some people with central core myopathy may be at increased risk of malignant hyperthermia, a potentially life-threatening reaction to certain anesthetics.

Nervous system diseases, also known as neurological disorders, refer to a group of conditions that affect the nervous system, which includes the brain, spinal cord, nerves, and muscles. These diseases can affect various functions of the body, such as movement, sensation, cognition, and behavior. They can be caused by genetics, infections, injuries, degeneration, or tumors. Examples of nervous system diseases include Alzheimer's disease, Parkinson's disease, multiple sclerosis, epilepsy, migraine, stroke, and neuroinfections like meningitis and encephalitis. The symptoms and severity of these disorders can vary widely, ranging from mild to severe and debilitating.

Ion channels are specialized transmembrane proteins that form hydrophilic pores or gaps in the lipid bilayer of cell membranes. They regulate the movement of ions (such as sodium, potassium, calcium, and chloride) across the cell membrane by allowing these charged particles to pass through selectively in response to various stimuli, including voltage changes, ligand binding, mechanical stress, or temperature changes. This ion movement is essential for many physiological processes, including electrical signaling, neurotransmission, muscle contraction, and maintenance of resting membrane potential. Ion channels can be categorized based on their activation mechanisms, ion selectivity, and structural features. Dysfunction of ion channels can lead to various diseases, making them important targets for drug development.

Sudden Infant Death Syndrome (SIDS) is defined by the American Academy of Pediatrics as "the sudden unexpected death of an infant

Inborn genetic diseases, also known as inherited genetic disorders, are conditions caused by abnormalities in an individual's DNA that are present at conception. These abnormalities can include mutations, deletions, or rearrangements of genes or chromosomes. In many cases, these genetic changes are inherited from one or both parents and may be passed down through families.

Inborn genetic diseases can affect any part of the body and can cause a wide range of symptoms, which can vary in severity depending on the specific disorder. Some genetic disorders are caused by mutations in a single gene, while others are caused by changes in multiple genes or chromosomes. In some cases, environmental factors may also contribute to the development of these conditions.

Examples of inborn genetic diseases include cystic fibrosis, sickle cell anemia, Huntington's disease, Duchenne muscular dystrophy, and Down syndrome. These conditions can have significant impacts on an individual's health and quality of life, and many require ongoing medical management and treatment. In some cases, genetic counseling and testing may be recommended for individuals with a family history of a particular genetic disorder to help them make informed decisions about their reproductive options.

Cardiac arrhythmias are abnormal heart rhythms that result from disturbances in the electrical conduction system of the heart. The heart's normal rhythm is controlled by an electrical signal that originates in the sinoatrial (SA) node, located in the right atrium. This signal travels through the atrioventricular (AV) node and into the ventricles, causing them to contract and pump blood throughout the body.

An arrhythmia occurs when there is a disruption in this electrical pathway or when the heart's natural pacemaker produces an abnormal rhythm. This can cause the heart to beat too fast (tachycardia), too slow (bradycardia), or irregularly.

There are several types of cardiac arrhythmias, including:

1. Atrial fibrillation: A rapid and irregular heartbeat that starts in the atria (the upper chambers of the heart).
2. Atrial flutter: A rapid but regular heartbeat that starts in the atria.
3. Supraventricular tachycardia (SVT): A rapid heartbeat that starts above the ventricles, usually in the atria or AV node.
4. Ventricular tachycardia: A rapid and potentially life-threatening heart rhythm that originates in the ventricles.
5. Ventricular fibrillation: A chaotic and disorganized electrical activity in the ventricles, which can be fatal if not treated immediately.
6. Heart block: A delay or interruption in the conduction of electrical signals from the atria to the ventricles.

Cardiac arrhythmias can cause various symptoms, such as palpitations, dizziness, shortness of breath, chest pain, and fatigue. In some cases, they may not cause any symptoms and go unnoticed. However, if left untreated, certain types of arrhythmias can lead to serious complications, including stroke, heart failure, or even sudden cardiac death.

Treatment for cardiac arrhythmias depends on the type, severity, and underlying causes. Options may include lifestyle changes, medications, cardioversion (electrical shock therapy), catheter ablation, implantable devices such as pacemakers or defibrillators, and surgery. It is essential to consult a healthcare professional for proper evaluation and management of cardiac arrhythmias.

Malignant hyperthermia (MH) is a rare, but potentially life-threatening genetic disorder that can occur in susceptible individuals as a reaction to certain anesthetic drugs or other triggers. The condition is characterized by a rapid and uncontrolled increase in body temperature (hyperthermia), muscle rigidity, and metabolic rate due to abnormal skeletal muscle calcium regulation.

MH can develop quickly during or after surgery, usually within the first hour of exposure to triggering anesthetics such as succinylcholine or volatile inhalational agents (e.g., halothane, sevoflurane, desflurane). The increased metabolic rate and muscle activity lead to excessive production of heat, carbon dioxide, lactic acid, and potassium, which can cause severe complications such as heart rhythm abnormalities, kidney failure, or multi-organ dysfunction if not promptly recognized and treated.

The primary treatment for MH involves discontinuing triggering anesthetics, providing supportive care (e.g., oxygen, fluid replacement), and administering medications to reduce body temperature, muscle rigidity, and metabolic rate. Dantrolene sodium is the specific antidote for MH, which works by inhibiting calcium release from the sarcoplasmic reticulum in skeletal muscle cells, thereby reducing muscle contractility and metabolism.

Individuals with a family history of MH or who have experienced an episode should undergo genetic testing and counseling to determine their susceptibility and take appropriate precautions when receiving anesthesia.

A mutation is a permanent change in the DNA sequence of an organism's genome. Mutations can occur spontaneously or be caused by environmental factors such as exposure to radiation, chemicals, or viruses. They may have various effects on the organism, ranging from benign to harmful, depending on where they occur and whether they alter the function of essential proteins. In some cases, mutations can increase an individual's susceptibility to certain diseases or disorders, while in others, they may confer a survival advantage. Mutations are the driving force behind evolution, as they introduce new genetic variability into populations, which can then be acted upon by natural selection.

Inwardly rectifying potassium channels (Kir) are a type of potassium channel that allow for the selective passage of potassium ions (K+) across cell membranes. The term "inwardly rectifying" refers to their unique property of allowing potassium ions to flow more easily into the cell (inward current) than out of the cell (outward current). This characteristic is due to the voltage-dependent blockage of these channels by intracellular magnesium and polyamines at depolarized potentials.

These channels play crucial roles in various physiological processes, including:

1. Resting membrane potential maintenance: Kir channels help establish and maintain the negative resting membrane potential in cells by facilitating potassium efflux when the membrane potential is near the potassium equilibrium potential (Ek).
2. Action potential repolarization: In excitable cells like neurons and muscle fibers, Kir channels contribute to the rapid repolarization phase of action potentials, allowing for proper electrical signaling.
3. Cell volume regulation: Kir channels are involved in regulating cell volume by mediating potassium influx during osmotic stress or changes in intracellular ion concentrations.
4. Insulin secretion: In pancreatic β-cells, Kir channels control the membrane potential and calcium signaling necessary for insulin release.
5. Renal function: Kir channels are essential for maintaining electrolyte balance and controlling renal tubular transport in the kidneys.

There are several subfamilies of inwardly rectifying potassium channels (Kir1-7), each with distinct biophysical properties, tissue distributions, and functions. Mutations in genes encoding these channels can lead to various human diseases, including cardiac arrhythmias, epilepsy, and Bartter syndrome.

Ion channel gating refers to the process by which ion channels in cell membranes open and close in response to various stimuli, allowing ions such as sodium, potassium, and calcium to flow into or out of the cell. This movement of ions is crucial for many physiological processes, including the generation and transmission of electrical signals in nerve cells, muscle contraction, and the regulation of hormone secretion.

Ion channel gating can be regulated by various factors, including voltage changes across the membrane (voltage-gated channels), ligand binding (ligand-gated channels), mechanical stress (mechanosensitive channels), or other intracellular signals (second messenger-gated channels). The opening and closing of ion channels are highly regulated and coordinated processes that play a critical role in maintaining the proper functioning of cells and organ systems.

Voltage-gated potassium channels are a type of ion channel found in the membrane of excitable cells such as nerve and muscle cells. They are called "voltage-gated" because their opening and closing is regulated by the voltage, or electrical potential, across the cell membrane. Specifically, these channels are activated when the membrane potential becomes more positive, a condition that occurs during the action potential of a neuron or muscle fiber.

When voltage-gated potassium channels open, they allow potassium ions (K+) to flow out of the cell down their electrochemical gradient. This outward flow of K+ ions helps to repolarize the membrane, bringing it back to its resting potential after an action potential has occurred. The precise timing and duration of the opening and closing of voltage-gated potassium channels is critical for the normal functioning of excitable cells, and abnormalities in these channels have been linked to a variety of diseases, including cardiac arrhythmias, epilepsy, and neurological disorders.

Sudden cardiac death (SCD) is a sudden, unexpected natural death caused by the cessation of cardiac activity. It is often caused by cardiac arrhythmias, particularly ventricular fibrillation, and is often associated with underlying heart disease, although it can occur in people with no known heart condition. SCD is typically defined as a natural death due to cardiac causes that occurs within one hour of the onset of symptoms, or if the individual was last seen alive in a normal state of health, it can be defined as occurring within 24 hours.

It's important to note that sudden cardiac arrest (SCA) is different from SCD, although they are related. SCA refers to the sudden cessation of cardiac activity, which if not treated immediately can lead to SCD.

Calcium channels are specialized proteins that span the membrane of cells and allow calcium ions (Ca²+) to flow in and out of the cell. They are crucial for many physiological processes, including muscle contraction, neurotransmitter release, hormone secretion, and gene expression.

There are several types of calcium channels, classified based on their biophysical and pharmacological properties. The most well-known are:

1. Voltage-gated calcium channels (VGCCs): These channels are activated by changes in the membrane potential. They are further divided into several subtypes, including L-type, P/Q-type, N-type, R-type, and T-type. VGCCs play a critical role in excitation-contraction coupling in muscle cells and neurotransmitter release in neurons.
2. Receptor-operated calcium channels (ROCCs): These channels are activated by the binding of an extracellular ligand, such as a hormone or neurotransmitter, to a specific receptor on the cell surface. ROCCs are involved in various physiological processes, including smooth muscle contraction and platelet activation.
3. Store-operated calcium channels (SOCCs): These channels are activated by the depletion of intracellular calcium stores, such as those found in the endoplasmic reticulum. SOCCs play a critical role in maintaining calcium homeostasis and signaling within cells.

Dysregulation of calcium channel function has been implicated in various diseases, including hypertension, arrhythmias, migraine, epilepsy, and neurodegenerative disorders. Therefore, calcium channels are an important target for drug development and therapy.

Ajmaline is a type of medication known as a Class I antiarrhythmic agent, which is used to treat certain types of abnormal heart rhythms. It works by blocking the sodium channels in the heart muscle, which helps to slow down the conduction of electrical signals within the heart and can help to restore a normal heart rhythm.

Ajmaline is typically administered intravenously (through a vein) in a hospital setting, as it acts quickly and its effects can be closely monitored by healthcare professionals. It may be used to diagnose certain types of heart rhythm disturbances or to treat acute episodes of arrhythmias that are not responding to other treatments.

Like all medications, ajmaline can have side effects, including dizziness, headache, nausea, and chest pain. It is important for patients to be closely monitored while taking this medication and to report any unusual symptoms to their healthcare provider. Ajmaline should only be used under the close supervision of a qualified healthcare professional.

Electrocardiography (ECG or EKG) is a medical procedure that records the electrical activity of the heart. It provides a graphic representation of the electrical changes that occur during each heartbeat. The resulting tracing, called an electrocardiogram, can reveal information about the heart's rate and rhythm, as well as any damage to its cells or abnormalities in its conduction system.

During an ECG, small electrodes are placed on the skin of the chest, arms, and legs. These electrodes detect the electrical signals produced by the heart and transmit them to a machine that amplifies and records them. The procedure is non-invasive, painless, and quick, usually taking only a few minutes.

ECGs are commonly used to diagnose and monitor various heart conditions, including arrhythmias, coronary artery disease, heart attacks, and electrolyte imbalances. They can also be used to evaluate the effectiveness of certain medications or treatments.

Anxiety disorders are a category of mental health disorders characterized by feelings of excessive and persistent worry, fear, or anxiety that interfere with daily activities. They include several different types of disorders, such as:

1. Generalized Anxiety Disorder (GAD): This is characterized by chronic and exaggerated worry and tension, even when there is little or nothing to provoke it.
2. Panic Disorder: This is characterized by recurring unexpected panic attacks and fear of experiencing more panic attacks.
3. Social Anxiety Disorder (SAD): Also known as social phobia, this is characterized by excessive fear, anxiety, or avoidance of social situations due to feelings of embarrassment, self-consciousness, and concern about being judged or viewed negatively by others.
4. Phobias: These are intense, irrational fears of certain objects, places, or situations. When a person with a phobia encounters the object or situation they fear, they may experience panic attacks or other severe anxiety responses.
5. Agoraphobia: This is a fear of being in places where it may be difficult to escape or get help if one has a panic attack or other embarrassing or incapacitating symptoms.
6. Separation Anxiety Disorder (SAD): This is characterized by excessive anxiety about separation from home or from people to whom the individual has a strong emotional attachment (such as a parent, sibling, or partner).
7. Selective Mutism: This is a disorder where a child becomes mute in certain situations, such as at school, but can speak normally at home or with close family members.

These disorders are treatable with a combination of medication and psychotherapy (cognitive-behavioral therapy, exposure therapy). It's important to seek professional help if you suspect that you or someone you know may have an anxiety disorder.

The subtalar joint is a joint in the foot that is located between the talus and calcaneus (heel) bones. It is called a "joint" because it allows for movement, specifically inversion and eversion, which are the movements that allow the foot to roll inward or outward. The subtalar joint plays an essential role in the biomechanics of the foot and ankle, helping to absorb shock during walking and running, and contributing to the stability of the foot during standing and walking. Issues with the subtalar joint can lead to various foot and ankle problems, such as flatfoot or chronic ankle instability.

Bundle-branch block (BBB) is a type of conduction delay or block in the heart's electrical system that affects the way electrical impulses travel through the ventricles (the lower chambers of the heart). In BBB, one of the two main bundle branches that conduct electrical impulses to the ventricles is partially or completely blocked, causing a delay in the contraction of one of the ventricles.

There are two types of bundle-branch block: right bundle-branch block (RBBB) and left bundle-branch block (LBBB). In RBBB, the right bundle branch is affected, while in LBBB, the left bundle branch is affected. The symptoms and severity of BBB can vary depending on the underlying cause and the presence of other heart conditions.

In some cases, BBB may not cause any noticeable symptoms and may only be detected during a routine electrocardiogram (ECG). However, if BBB occurs along with other heart conditions such as coronary artery disease, heart failure, or cardiomyopathy, it can increase the risk of serious complications such as arrhythmias, syncope, and even sudden cardiac death.

Treatment for bundle-branch block depends on the underlying cause and the severity of the condition. In some cases, no treatment may be necessary, while in others, medications, pacemakers, or other treatments may be recommended to manage symptoms and prevent complications.

Rüdel, R (2000). "Muscle chloride channelopathies: myotonia congenita". In Lehmann-Horn, Frank; Jurkat-Rott, Karin (eds.). ... Rüdel, R (2000). "Muscle chloride channelopathies: myotonia congenita". In Lehmann-Horn, Frank; Jurkat-Rott, Karin (eds.). ... Channelopathies. Burlington: Elsevier. pp. 44-46. ISBN 9780080528854. Breed data sheet: Myotonic / United States of America ( ... Channelopathies. Burlington: Elsevier. pp. 44-46. ISBN 9780080528854. "On the Sacred Disease", trans. Francis Adams, in ...
... are the most common cause of channelopathies. Acquired channelopathies are caused by acquired disorders, drug use, toxins, etc ... Channelopathies are a group of diseases caused by the dysfunction of ion channel subunits or their interacting proteins. These ... Channelopathies can be categorized based on the organ system which they are associated with. In the cardiovascular system, the ... Channelopathies affecting synaptic function are a type of synaptopathy. Mutations in genes encoding ion channels, which cause ...
Other channelopathies may cause it as well. It is also associated with Schwartz-Jampel syndrome. Hyperekplexia Neuromuscular ... Kim, June-Bum (January 2014). "Channelopathies". Korean Journal of Pediatrics. 57 (1): 1-18. doi:10.3345/kjp.2014.57.1.1. ISSN ... Myotonia is the defining symptom of many channelopathies (diseases of ion channel transport) such as myotonia congenita, ... Vicart, S.; Sternberg, D.; Fontaine, B.; Meola, G. (1 October 2005). "Human skeletal muscle sodium channelopathies". ...
Kullmann, Dimitri M. (2010). "Neurological channelopathies". Annual Review of Neuroscience. 33: 151-172. doi:10.1146/annurev- ... neurological channelopathies and Synaptopathies, gene therapy for epilepsy, and mechanisms of neural oscillations. Kullmann ...
Feske S (July 2010). "CRAC channelopathies". Pflügers Archiv. 460 (2): 417-35. doi:10.1007/s00424-009-0777-5. PMC 2885504. PMID ...
... cardiac ion channelopathies form a major group of heart diseases. Cardiac ion channelopathies may explain some of the cases of ... Skinner JR, Winbo A, Abrams D, Vohra J, Wilde AA (January 2019). "Channelopathies That Lead to Sudden Cardiac Death: Clinical ... Marbán, Eduardo (10 January 2002). "Cardiac channelopathies". Nature. 415 (6868): 213-218. Bibcode:2002Natur.415..213M. doi: ... Marban, Eduardo (1 July 2003). "Cardiac Channelopathies". Heart Views. 4 (3): 4. ISSN 1995-705X. ...
Planells-Cases R, Jentsch TJ (March 2009). "Chloride channelopathies" (PDF). Biochimica et Biophysica Acta (BBA) - Molecular ...
The concept of channelopathies and the link with specific ion channel mutations emerged at the end of the 20th century. Kung AW ... TPP is regarded as a model for related conditions, known as "channelopathies", which have been linked with mutations in ion ... Ryan DP, Ptácek LJ (October 2010). "Episodic neurological channelopathies". Neuron. 68 (2): 282-92. doi:10.1016/j.neuron. ...
Ryanodine receptor channelopathies. Biochem. Biophys. Res. Commun. 322:1280-1285. PMID 15336975 DOI: 10.1016/j.bbrc.2004.08.033 ... particularly for the treatment of RyR channelopathies. Xiao, Liang; Gurrola, Georgina B.; Zhang, Jing; Martin, Mario San; ...
Vicart S, Sternberg D, Fontaine B, Meola G (2005). "Human skeletal muscle sodium channelopathies". Neurol Sci. 26 (4): 194-202 ... Cannon S (2006). "Pathomechanisms in channelopathies of skeletal muscle and brain". Annu Rev Neurosci. 29: 387-415. doi:10.1146 ... Channelopathies, Myoneural junction and neuromuscular diseases, Rare diseases). ...
Vaeth, Martin; Feske, Stefan (2018-06-01). "Ion channelopathies of the immune system". Current Opinion in Immunology. 52: 39-50 ... Meisler, Miriam H.; Hill, Sophie F.; Yu, Wenxi (March 2021). "Sodium channelopathies in neurodevelopmental disorders". Nature ... "Cardiac Channelopathies and Sudden Death: Recent Clinical and Genetic Advances". Biology. 6 (1): 7. doi:10.3390/biology6010007 ...
Behere SP, Weindling SN (2014). "Inherited arrhythmias: The cardiac channelopathies". Annals of Pediatric Cardiology. 8 (3): ... of SIDS cases are believed to be due to channelopathies, which are inherited defects in the ion channels which play an ...
Jurkat-Rott K, Lehmann-Horn F (August 2005). "Muscle channelopathies and critical points in functional and genetic studies". J ... Cannon, Stephen C. (2018). "Sodium Channelopathies of Skeletal Muscle". Voltage-gated Sodium Channels: Structure, Function and ... Channelopathies. Handbook of Experimental Pharmacology. Springer International Publishing. 246: 309-330. doi:10.1007/164_2017_ ...
Demirbilek H, Galcheva S, Vuralli D, Al-Khawaga S, Hussain K (May 2019). "Ion Transporters, Channelopathies, and Glucose ... Channelopathies, Autosomal dominant disorders, Syndromes affecting the heart). ...
... to channelopathies (sodium channels, "SCN3A"). Patients with autism have overall higher levels of cortical gyrification, but ...
Beyder A, Farrugia G (October 2016). "Ion channelopathies in functional GI disorders". Am J Physiol Gastrointest Liver Physiol ... Channelopathy and muscular dystrophy: IBS and functional GI diseases are comorbidities of genetic channelopathies that cause ... Beyder A, Farrugia G (2016). "Ion channelopathies in functional GI disorders". American Journal of Physiology. Gastrointestinal ...
K, Jurkat-Rott; H, Lerche; N, Mitrovic; F, Lehmann-Horn (September 1999). "Teaching Course: Ion Channelopathies in Neurology". ...
Waxman SG (July 2013). "Painful Na-channelopathies: an expanding universe". Trends in Molecular Medicine. 19 (7): 406-9. doi: ...
Abraham MR, Jahangir A, Alekseev AE, Terzic A (November 1999). "Channelopathies of inwardly rectifying potassium channels". ...
Genetic and autoimmune disorders of ion channels and their modifiers are known as channelopathies. See Category:Channelopathies ...
Venetucci L, Denegri M, Napolitano C, Priori SG (October 2012). "Inherited calcium channelopathies in the pathophysiology of ... Obeyesekere MN, Antzelevitch C, Krahn AD (February 2015). "Management of ventricular arrhythmias in suspected channelopathies ...
Vincent A (2008). "Autoimmune channelopathies: John Newsom-Davis's work and legacy. A summary of the Newsom-Davis Memorial ...
Schorge, Stephanie (2018-02-14). "Channelopathies go above and beyond the channels" (PDF). Neuropharmacology. 132: 1-2. doi: ...
Function and Channelopathies. Handbook of Experimental Pharmacology. Vol. 246. pp. 147-160. doi:10.1007/164_2018_99. ISBN 978-3 ... channelopathies) in patients with irritable bowel syndrome". Gastroenterology. 146 (7): 1659-1668. doi:10.1053/j.gastro.2014.02 ...
"Inherited calcium channelopathies in the pathophysiology of arrhythmias". Nature Reviews. Cardiology. 9 (10): 561-575. doi: ...
In addition, channelomic studies screen potential drugs for their effectiveness at channelopathies, by examining the binding ... Membrane biology, Ion channels, Channelopathies, Electrophysiology, Integral membrane proteins). ...
"Towards Precision Medicine with Human iPSCs for Cardiac Channelopathies". Circulation Research. 125 (6): 653-658. doi:10.1161/ ...
Mutation studies allow experimenters to study genetically inherited channelopathies. A channelopathy is any disease that ...
Channelopathies, Blindness, Disorders of choroid and retina, Autosomal recessive disorders). ...
Jurkat-Rott K, Lehmann-Horn F (August 2005). "Muscle channelopathies and critical points in functional and genetic studies". ...
... Pflugers Arch. 2010 Jul;460(2):405-15. doi: 10.1007/s00424-010-0810-8. Epub 2010 Mar 8. ... While full evidence for neuronal HCN channelopathies is not yet available, several indications point to a link between temporal ... Here we briefly review the current knowledge of HCN-related channelopathies in the heart and the brain. ... which delayed the investigation of HCN-related channelopathies. In cardiac pacemaker cells, the main function of f-channels is ...
Muscle channelopathies are caused by mutations in ion channel genes, by antibodies directed against ion channel proteins, or by ...
Overview of Channelopathies - Etiology, pathophysiology, symptoms, signs, diagnosis & prognosis from the Merck Manuals - ... The most common genetic channelopathies are the Brugada syndrome (BrS) Brugada Syndrome Brugada syndrome is an inherited ... Channelopathies are a group of genetic, autoimmune, or inflammatory conditions that alter cardiomyocyte ion channel function in ... Together, these genetic channelopathies account for approximately 10% of sudden cardiac deaths. ...
CaV1.2 channelopathies: from arrhythmias to autism, bipolar disorder, and immunodeficiency.. CaV1.2 channelopathies: from ...
Overview of Channelopathies - Etiology, pathophysiology, symptoms, signs, diagnosis & prognosis from the MSD Manuals - Medical ... The most common genetic channelopathies are the Brugada syndrome (BrS) Brugada Syndrome Brugada syndrome is an inherited ... Channelopathies are a group of genetic, autoimmune, or inflammatory conditions that alter cardiomyocyte ion channel function in ... Together, these genetic channelopathies account for approximately 10% of sudden cardiac deaths. ...
Ion channelopathies are unlikely to be routinely considered in the differential diagnosis of children presenting with ... Electrifying dysmorphology: Potassium channelopathies causing dysmorphic syndromes.. Mark James Hamilton, Mohnish Suri. ... In this chapter, we review a distinct subgroup of rare potassium channelopathies whose presentations are instead suggestive of ...
Medichron Publications LLC is a publishing company for the online health and medical community. The company specializes in research in the field of chronobiology and news pertaining to a chronobiological approach to health. ...
Calcium Channels, Channelopathies, Humans, Mutation, NAV1.7 Voltage-Gated Sodium Channel, NAV1.8 Voltage-Gated Sodium Channel, ...
In all autoimmune channelopathies, the relationship between autoantibody specificity and clinical phenotype is complex. But ... A second class of channelopathies, characterized by autoantibodies against ligand- and voltage-gated ion channels, cause a ... with this new information, autoimmune channelopathies are detected and treated with increasing success, and future research ... In all autoimmune channelopathies, the relationship between autoantibody specificity and clinical phenotype is complex. But ...
Ion Channelopathies Congenital ion channel diseases (long QT or short QT syndromes, Brugada syndrome, catecholaminergic ...
Rüdel, R (2000). "Muscle chloride channelopathies: myotonia congenita". In Lehmann-Horn, Frank; Jurkat-Rott, Karin (eds.). ... Rüdel, R (2000). "Muscle chloride channelopathies: myotonia congenita". In Lehmann-Horn, Frank; Jurkat-Rott, Karin (eds.). ... Channelopathies. Burlington: Elsevier. pp. 44-46. ISBN 9780080528854. Breed data sheet: Myotonic / United States of America ( ... Channelopathies. Burlington: Elsevier. pp. 44-46. ISBN 9780080528854. "On the Sacred Disease", trans. Francis Adams, in ...
II - Arrhythmias and channelopathies. 1) Atrial fibrillation. AF increases with age and whilst for many there are no clinical ... 3) HRS/EHRA Expert Consensus Statement on the State of Genetic Testing for the Channelopathies and Cardiomyopathies. Ackerman ... ECG and prolonged ECG monitoring can detect conduction disorders, arrhythmias and channelopathies although we need to be ... approach to screening from detecting conduction and structural disorders through to arrhythmias and channelopathies. ...
Cardiac Channelopathies. Multigene panel. Clinical suspicion or family history of cardiac channelopathies. Diagnosis and ...
... to generate therapeutic antagonist and agonist monoclonal antibodies to ion channels to efficiently treat channelopathies ... Inherited channelopathies are individually rare. For instance, the prevalence of the most common form of congenital long-QT ... It is important to remember that channelopathies are governed by two fundamental concepts. On the one hand, different mutations ... Therapeutic antibodies & channelopathies : about the difficulty in generating antibodies against ion channels. ...
Lessons from the Heart: New Insights into Physiology from Ion Channelopathies Geoffrey Pitt, MD, PhD. Director of the ...
Cardiac Channelopathies: Recognition, Treatment, Management Kathleen T. Hickey, FNP-BC, ANP-BC, EdD; Kathleen T. Hickey, FNP-BC ... This article provides an overview of cardiac channelopathies that nurses might encounter in an array of clinical and research ... Kathleen T. Hickey, Amir Elzomor; Cardiac Channelopathies: Recognition, Treatment, Management. AACN Adv Crit Care 15 March 2018 ... Cardiac arrhythmias caused by disorders of ion channels are known as cardiac channelopathies. Nurses are important members of ...
HERG1 channelopathies. Pflugers Arch. 2010 Jul;460(2):265-76. doi: 10.1007/s00424-009-0758-8. Epub 2009 Nov 22. Citation on ...
... which have previously been identified in genetic forms of cardiac channelopathies, including pacemaker channels (HCN4-Brugada ...
From Proteins to Body Surface ECG: Computational Investigations of Ion Channelopathies. * Ansprechperson: Axel Loewe, Gunnar ... From Proteins to Body Surface ECG: Computational Investigations of Ion Channelopathies. Download als PDF ...
Skeletal Muscle Channelopathies. Vivekanandam V, Munot P, Hanna MG, Matthews E. Neurol Clin. 2020 Aug;38(3):481-491. doi: ... Concurrent sodium channelopathies and amyotrophic lateral sclerosis supports shared pathogenesis. Franklin JP, Cooper-Knock J, ... Concurrent sodium channelopathies and amyotrophic lateral sclerosis supports shared pathogenesis. Franklin JP, Cooper-Knock J, ... COVID-19 infection and vaccination in patients with skeletal muscle channelopathies. Vivekanandam V, Jayaseelan D, Hanna MG. ...
Hereditary Channelopathies Caused by TRPV4 Mutations. 2014, 413-440. https://doi.org/10.1007/978-3-642-40282-1_21. ... The puzzle of TRPV4 channelopathies. EMBO reports 2013, 14 (2) , 152-163. https://doi.org/10.1038/embor.2012.219. ...
Ion channelopathies in endocrinology: recent genetic findings and pathophysiological insig Ion channelopathies in endocrinology ... recapitulates the membrane action potential in endocrine cells and offers a short overview of known endocrine channelopathies ...
... the ion channelopathies and developmental epilepsy genes; and (5) for the first time in the Jaspers series describes the ...
Jurkat-Rott K, Holzherr B, Fauler M, Lehmann-Horn F. Sodium channelopathies of skeletal muscle result from gain or loss of ... Zimmer T, Surber R. SCN5A channelopathies-an update on mutations and mechanisms. Prog Biophys Mol Biol 2008, 98: 120-136. ... Painful Na-channelopathies: an expanding universe. Trends Mol Med 2013, 19: 406-409. ...
HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies and cardiomyopathies: This document ... HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies and cardiomyopathies: This document ...
HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies and cardiomyopathies: This document ... HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies and cardiomyopathies: This document ...
Patient-Specific Induced Pluripotent Stem Cell Models for Heritable Channelopathies and Cardiomyopathies. *. Location:. ... Patient-Specific Induced Pluripotent Stem Cell Models for Heritable Channelopathies and Cardiomyopathies. Rochester, MN ... study is to derive and characterize patient-specific disease models for sudden death-predisposing heritable channelopathies and ...
Sodium "channelopathies" and sudden death: must you be so sensitive? Balser JR. Balser JR. Circ Res. 1999 Oct 29;85(9):872-4. ... Inherited sodium channelopathies: models for acquired arrhythmias? Balser JR. Balser JR. Am J Physiol Heart Circ Physiol. 2002 ...

No FAQ available that match "channelopathies"

No images available that match "channelopathies"