fbpx
Wikipedia

GLUT1 deficiency


GLUT1 deficiency syndrome, also known as GLUT1-DS, De Vivo disease or Glucose transporter type 1 deficiency syndrome, is an autosomal dominant genetic metabolic disorder associated with a deficiency of GLUT1, the protein that transports glucose across the blood brain barrier.[1] Glucose Transporter Type 1 Deficiency Syndrome has an estimated birth incidence of 1 in 90,000[2] to 1 in 24,300.[3] This birth incidence translates to an estimated prevalence of 3,000 to 7,000 in the U.S.[2]

De Vivo disease
Other namesDe Vivo disease
De Vivo disease has an autosomal dominant pattern of inheritance
SpecialtyMedical genetics 

Presentation edit

GLUT1 deficiency is characterized by an array of signs and symptoms including mental and motor developmental delays, infantile seizures refractory to anticonvulsants, ataxia, dystonia, dysarthria, opsoclonus, spasticity, other paroxysmal neurologic phenomena and sometimes deceleration of head growth also known as microcephaly. The presence and severity of symptoms vary considerably between affected individuals. Individuals with the disorder generally have frequent seizures (epilepsy), often beginning in the first months of life. In newborns, the first sign of the disorder may be involuntary eye movements that are rapid and irregular.[4] Patients typically begin to experience seizures between three and six months of age, but some occur much later.[5] Other seizure types may occur, including generalized tonic clonic, focal, myoclonic, atypical absence, atonic or unclassified.[5]

Mothers of infants with this disorder usually have uneventful pregnancies and deliveries, with the child appearing normal and within typical birth weight and length ranges. Infants with GLUT1 deficiency syndrome have a normal head size at birth, but the growth of the brain and skull is slow, in severe cases resulting in an abnormally small head size (microcephaly).[4] Typically, seizures start between one and four months in 90% of cases with abnormal eye movements and apneic episodes preceding the onset of seizures in some cases.[6] Seizures usually are complex to begin with and later become more generalized. Seizure frequency is variable and a history of decreasing frequency during times of ketosis may prompt a diagnosis. It is estimated that 10% of individuals with Glut 1 Deficiency do not have seizures and symptoms are typically less severe in these cases.[7] Most of these non-epileptic cases will still have developmental delay, intellectual delays and movement disorders such as ataxia, alternating hemiplegia or dystonia.[7]

Some symptoms may be present all the time (like walking difficulties), while other signs may come and go (like seizures or poor balance).[8] These findings can be clustered under three major domains: cognition, behavior and movement.[8]

Effects edit

The syndrome can cause infantile seizures refractory to anticonvulsive drugs, developmental delay, acquired microcephaly and neurologic manifestations including spasticity, hypotonia and ataxia.[9] The frequency, severity and types of seizures may vary considerably among GLUT1 deficiency patients and do not necessarily correspond to the severity of other symptoms. Most seizures in GLUT1 deficiency patients are not easily treated with anti-seizure medications. A minority of GLUT1 deficiency patients (approximately 10%) do not experience seizures.[5] Cognitive symptoms often become apparent as developmental milestones are delayed. Cognitive deficits range from subtle learning difficulties to severe intellectual disabilities. Often speech and language are impaired.[5] Behavioral symptoms affect relations with other people and may include short attention span, intractability, and delays in achieving age-appropriate behaviors. Sociability with peers, however, is a strength in GLUT1 deficiency patients.[5] Movement symptoms relate to the quality of motor functions. Walking may be delayed or difficult because legs are stiff (spasticity), balance is poor (ataxia) or posture is twisted (dystonia). Fine motor deficits may affect speech quality and manipulative skills, such as writing. These abnormalities may be constant or intermittent (paroxysmal).[5] Paroxysmal exercise-induced dyskinesia (PED) may also be present.[10] Other intermittent symptoms may include headaches, confusion, and loss of energy. Episodes of confusion, lack of energy/stamina, and/or muscle twitches may occur; particularly during periods without food.[7] Some young patients experience occasional abnormal eye movements that may resemble opsoclonus or nystagmus.[5] The rapid eye movements that some Glut 1 patients exhibit are rapid, multidirectional, and there is often a head movement in the same direction as the eye movement.[11] These abnormal eye movements were recently named aberrant gaze saccades.[11] Hemiplegia or alternating intermittent hemiplegia may occur in some patients and mimic stroke-like symptoms.[12] Another characteristic of GLUT1 deficiency is that symptoms are sensitive to food (e.g. symptoms that can be temporarily improved by intake of carbohydrates), and symptoms may be worse in the morning upon and just after waking.[5] All symptoms may be aggravated or triggered by factors such as hunger, fatigue, heat, anxiety, and sickness. The symptom picture for each patient may evolve and change over time as children with GLUT1 deficiency grow and develop through adolescence and into adulthood.[5] Data on adult Glut1DS are just emerging.[13] Changes in symptomatology over time include a shift from infantile-childhood onset epilepsy to adolescent-adult onset movement disorders including PED.

Genetics edit

The GLUT1 protein that transports glucose across the blood brain barrier is made by the SLC2A1 gene, located on chromosome 1.[8]

In GLUT1 deficiency Syndrome one of the two genes is damaged by a mutation and insufficient protein is made. As a result, insufficient glucose is passing the blood brain barrier. Having less functional GLUT1 protein reduces the amount of glucose available to brain cells, which affects brain development and function.[14] Because glucose is the primary source of fuel for the brain, patients with GLUT1 deficiency have insufficient cellular energy to permit normal brain growth and function.[8]

Around 90% of cases of GLUT1 deficiency syndrome are de novo mutations of the SLC2A1 gene (a mutation not present in the parents, but present in one of the two copies of the gene in the baby), although it can be inherited.[15]

Glut 1 Deficiency can be inherited in an autosomal dominant manner. A person with GLUT1 deficiency syndrome has a 50% chance of passing along the altered SLC2A1 gene to his or her offspring.[16]


In a study focusing on GLUT1 mice model brain slides, physiological glucose concentration was found to be a modulator of frequency oscillations and less frequent 30–50 Hz or gamma oscillations.[17]

Diagnosis edit

Early diagnosis is crucial in order to initiate treatment during the important early stages of brain development. To make a proper diagnosis, it is important to know the various symptoms of GLUT1 deficiency and how those symptoms evolve with age.[18]

GLUT1 deficiency is diagnosed based on the clinical features in combination with determining the glucose concentration in the CSF and/or a genetic analysis through a lumbar puncture (spinal tap).[13] A low glucose value in CSF (<2.2 mmol/L) or lowered CSF/plasma glucose ratio (<0.4)are indicatieve of GLUT1 deficiency. A genetic mutation in the SLC2A1 gene also confirms the diagnosis, although mutations have not been identified in approximately 15% of GLUT1 deficiency patients.[19] A highly specialized lab test called the red blood cell uptake assay may confirm GLUT1 deficiency but is not commercially available.[20]

Management edit

Anti-seizure medications are generally not effective, since they do not provide nourishment to the starved brain.[8]

Once diagnosed, a medically supervised ketogenic diet is usually recommended as it can help to control seizures.[21] The ketogentic diet is the current standard of care treatment, with 80% of patients having >90% seizure reduction[13] and improving some movement disorders in approximately two thirds of GLUT1 deficiency patients.[18] There is also some evidence of some cognitive benefits for GLUT1 deficiency patients on a ketogenic diet, and most parents report improved energy, alertness, balance, coordination, and concentration,[18] especially when the diet is started early in childhood.

The ketogenic diet is a diet high in fat and low in protein and carbohydrates, with up to 90% of calories obtained from fat. Since the diet is low in carbohydrates, the body gets little glucose, normally the main energy source. The fat in the diet is converted by the liver in ketone bodies, which causes a build up of ketones in the blood stream, called ketosis. Ketone bodies are transported across the blood-brain barrier by other means than the GLUT1 protein and thus serve as an alternative fuel for the brain when glucose is not available.[22]

While ketogenic diets have been proven effective to control seizures and relieve some movement disorders in many GLUT1 deficiency patients, some patients do not respond as well as others. In addition, some critical symptoms, including cognitive deficits and certain movement difficulties, tend to persist in GLUT1 deficiency patients treated by a ketogenic diet, raising the question whether GLUT1 deficiency is caused simply by a lack of proper brain energy or if there are more complicated and widespread systems and processes affected.[18]

The ketogenic diet must be carefully crafted and tailored to meet the needs of each patient and reduce the risk of side effects. It should only be used under the care of medical professionals and dietitians, and it may take some time to establish the ideal ratio of fat versus proteins and carbohydrates and other diet variables for each individual patient to experience optimal tolerance and benefits. Variations on the ketogenic diet, including the Modified Atkins Diet, and diets based on MCT oil have also been shown to be beneficial for some GLUT1 deficiency patients.[18]

While the classic ketogenic diet is commonly used for younger children, compliance with the ketogenic diet can be difficult for older children and adults. In recent years, the Modified Atkins Diet, and MCT oil based diets, have gained increasing acceptance among doctors treating these groups and may be more feasible for quality of life and compliance.[13] There is growing empirical evidence that these diets can provide at least some of the benefits of the classical ketogenic diet for some GLUT1 deficiency patients.[18]

Ketone esters are an area of dietary therapy currently under investigation for potential treatment of GLUT1 deficiency and other medical conditions. Ketone esters are synthetic ketones that break down into natural ketones when metabolized. Ketone esters have been shown in recent research to improve seizures and movement disorders in GLUT1 deficient mice, but human studies have not yet been conducted.[18]

Triheptanoin (C7 oil), a triglyceride oil synthesized from castor beans.[18] is an investigational pharmaceutical-grade medical food that has shown potential as a treatment for a number of inherited metabolic diseases. When metabolized by the body, C7 oil produces ketones similar to those produced on a ketogenic diet in addition to other types of ketones that are thought to fulfill further metabolic requirements in the absence of sufficient glucose.[18] A phase 3 clinical trial however failed to find an improvement in patients with GLUT1 DS with disabling movement disorders.

The inhibition of insulin production to increase glucose in the blood with the medicine diazoxide, in combination with continuous glucose monitoring, has been successful in one adolescent. The increased blood glucose also increases the availability of glucose in the brain, through the increased transfer of more glucose through the GLUT1-protein. She became seizure-free, became more physically active and had improved cognition.[23]

Researchers are studying gene therapy as a possible effective treatment for Glut 1 Deficiency.[24][25]

Therapies and rehabilitative services are beneficial since most GLUT1 deficiency patients experience movement disturbances as well as speech and language disorders. Occupational, physical, and speech/language therapies are standard for most patients, especially in childhood.[18] Many families greatly benefit from other therapies such as aquatic therapy, hippotherapy, specific learning strategies, and behavioral therapy.[18]

Glut 1 patients Weak Areas are lowered IQ and adaptive behavior scores, expressive-language deficits, weakness in fine motor skills, limited visual attention to details, weakness in abstract analytical skills, and weakness in transfer of learning to new contexts.[citation needed]

Strong Areas include receptive language or understanding, social skills, fun-loving and empathetic personalities, perseverance.[18]

References edit

  1. ^ Todor, Arsov (2016). "Glut-1 deficiency: From Pathophysilogy ad genetics to abroad clinical spectrum". Sanamed. 11 (2): 151–155. doi:10.5937/sanamed1602151A.
  2. ^ a b "Understanding Glucose Transporter Type 1 Deficiency Syndrome (Glut1 DS): Current Management and Future Approaches". Epilepsy Foundation. Retrieved 2018-01-31.
  3. ^ Symonds (2019). "Incidence and phenotypes of childhood-onset genetic epilepsies: a prospective population-based national cohort". Brain. 142 (8): 2303–2318. doi:10.1093/brain/awz195. PMC 6658850. PMID 31302675.
  4. ^ a b "GLUT1 deficiency syndrome". Genetics Home Reference. Retrieved 10 October 2011.
  5. ^ a b c d e f g h i "Brochures". GLUT1 deficiency Foundation. Retrieved 2018-01-25.
  6. ^ Wang, Pascual, Vivo. "Glucose Transporter Type 1 Deficiency Syndrome". GeneReviews.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  7. ^ a b c Reference, Genetics Home. "GLUT1 deficiency syndrome". Genetics Home Reference. Retrieved 2018-01-25.
  8. ^ a b c d e (PDF). GLUT1 deficiency Foundation. Archived from the original (PDF) on 2015-10-02. Retrieved 19 June 2017.
  9. ^ Ticus I, Cano A, Villeneuve N, Milh M, Mancini J, Chabrol B (August 2008). "[GLUT-1 deficiency syndrome or De Vivo disease: a case report]". Arch Pediatr (in French). 15 (8): 1296–9. doi:10.1016/j.arcped.2008.04.024. PMID 18556184.
  10. ^ Wang, Dong; Pascual, Juan M.; De Vivo, Darryl (1993). "Glucose Transporter Type 1 Deficiency Syndrome". In Adam, Margaret P.; Ardinger, Holly H.; Pagon, Roberta A.; Wallace, Stephanie E.; Bean, Lora J.H.; Stephens, Karen; Amemiya, Anne (eds.). GeneReviews®. Seattle (WA): University of Washington, Seattle. PMID 20301603.
  11. ^ a b Pearson, Toni S.; Pons, Roser; Engelstad, Kristin; Kane, Steven A.; Goldberg, Michael E.; Vivo, Darryl C. De (2017-04-25). "Paroxysmal eye–head movements in GLUT1 deficiency syndrome". Neurology. 88 (17): 1666–1673. doi:10.1212/WNL.0000000000003867. ISSN 0028-3878. PMC 5405761. PMID 28341645.
  12. ^ Braakman, H.M.H.; Nicolai, J.; Willemsen, M.A.A.P. (2017-06-01). "Stroke-like episodes add to the phenotypic spectrum of GLUT1 deficiency syndrome". European Journal of Paediatric Neurology. 21: e176. doi:10.1016/j.ejpn.2017.04.995. ISSN 1090-3798.
  13. ^ a b c d Klepper, Joerg (2020). "Glut1 Deficiency Syndrome (Glut1DS): State of the art in 2020 and recommendations of the international Glut1DS study group". Epilepsia Open. 5 (3): 354–365. doi:10.1002/epi4.12414. PMC 7469861. PMID 32913944.
  14. ^ Reference, Genetics Home. "GLUT1 deficiency syndrome". Genetics Home Reference. Retrieved 2017-06-15.
  15. ^ (PDF). GLUT1 deficiency Foundation. Archived from the original (PDF) on 2015-10-03. Retrieved 19 June 2017.
  16. ^ . rarediseases.info.nih.gov. Archived from the original on 2018-02-01. Retrieved 2018-01-31.
  17. ^ Solis, Elysandra M.; Good, Levi B.; Vázquez, Rafael Granja; Patnaik, Sourav; Hernandez-Reynoso, Ana G.; Ma, Qian; Angulo, Gustavo; Dobariya, Aksharkumar; Cogan, Stuart F.; Pancrazio, Joseph J.; Pascual, Juan M.; Jakkamsetti, Vikram (2023). "Isolation of the murine Glut1 deficient thalamocortical circuit: wavelet characterization and reverse glucose dependence of low and gamma frequency oscillations". Frontiers in Neuroscience. 17: 1191492. doi:10.3389/fnins.2023.1191492. ISSN 1662-4548. PMC 10565352. PMID 37829723.
  18. ^ a b c d e f g h i j k l "Brochures". GLUT1 deficiency Foundation. Retrieved 2018-01-31.
  19. ^ . GLUT1 deficiency Foundation. Archived from the original on 2018-02-01. Retrieved 2018-01-31.
  20. ^ . www.metafora-biosystems.com (in French). Archived from the original on 2018-02-01. Retrieved 2018-01-31.
  21. ^ De Vivo, Darryl C.; Trifiletti, Rosario R.; Jacobson, Ronald I.; Ronen, Gabriel M.; Behmand, Ramin A.; Harik, Sami I. (5 September 1991). "Defective Glucose Transport across the Blood-Brain Barrier as a Cause of Persistent Hypoglycorrhachia, Seizures, and Developmental Delay". New England Journal of Medicine. 325 (10): 703–709. doi:10.1056/NEJM199109053251006. PMID 1714544.
  22. ^ Brockmann, Knut (2011-07-01). "Towards a more palatable treatment for GLUT1 deficiency syndrome". Developmental Medicine & Child Neurology. 53 (7): 580–581. doi:10.1111/j.1469-8749.2011.03946.x. ISSN 1469-8749. PMID 21585366.
  23. ^ Logel (2021). "Exploring diazoxide and continuous glucose monitoring as treatment for Glut1 deficiency syndrome". Ann Clin Transl Neurol. 8 (11): 2205–2209. doi:10.1002/acn3.51462. PMC 8607448. PMID 34612610.
  24. ^ "Sanofi Awarded New Funding to Monani Lab to Study Glucose Transporter-1 Deficiency Syndrome". Pathology. 2017-06-19. Retrieved 2018-01-31.
  25. ^ GLUT1 deficiency Foundation (2018-01-30), Dr. Umrao Monani, archived from the original on 2021-12-21, retrieved 2018-01-31{{citation}}: CS1 maint: numeric names: authors list (link)

External links edit

  • GeneReview/NIH/UW entry on Glucose Transporter Type 1 Deficiency Syndrome

glut1, deficiency, syndrome, also, known, glut1, vivo, disease, glucose, transporter, type, deficiency, syndrome, autosomal, dominant, genetic, metabolic, disorder, associated, with, deficiency, glut1, protein, that, transports, glucose, across, blood, brain, . GLUT1 deficiency syndrome also known as GLUT1 DS De Vivo disease or Glucose transporter type 1 deficiency syndrome is an autosomal dominant genetic metabolic disorder associated with a deficiency of GLUT1 the protein that transports glucose across the blood brain barrier 1 Glucose Transporter Type 1 Deficiency Syndrome has an estimated birth incidence of 1 in 90 000 2 to 1 in 24 300 3 This birth incidence translates to an estimated prevalence of 3 000 to 7 000 in the U S 2 De Vivo diseaseOther namesDe Vivo diseaseDe Vivo disease has an autosomal dominant pattern of inheritanceSpecialtyMedical genetics Contents 1 Presentation 1 1 Effects 2 Genetics 3 Diagnosis 4 Management 5 References 6 External linksPresentation editGLUT1 deficiency is characterized by an array of signs and symptoms including mental and motor developmental delays infantile seizures refractory to anticonvulsants ataxia dystonia dysarthria opsoclonus spasticity other paroxysmal neurologic phenomena and sometimes deceleration of head growth also known as microcephaly The presence and severity of symptoms vary considerably between affected individuals Individuals with the disorder generally have frequent seizures epilepsy often beginning in the first months of life In newborns the first sign of the disorder may be involuntary eye movements that are rapid and irregular 4 Patients typically begin to experience seizures between three and six months of age but some occur much later 5 Other seizure types may occur including generalized tonic clonic focal myoclonic atypical absence atonic or unclassified 5 Mothers of infants with this disorder usually have uneventful pregnancies and deliveries with the child appearing normal and within typical birth weight and length ranges Infants with GLUT1 deficiency syndrome have a normal head size at birth but the growth of the brain and skull is slow in severe cases resulting in an abnormally small head size microcephaly 4 Typically seizures start between one and four months in 90 of cases with abnormal eye movements and apneic episodes preceding the onset of seizures in some cases 6 Seizures usually are complex to begin with and later become more generalized Seizure frequency is variable and a history of decreasing frequency during times of ketosis may prompt a diagnosis It is estimated that 10 of individuals with Glut 1 Deficiency do not have seizures and symptoms are typically less severe in these cases 7 Most of these non epileptic cases will still have developmental delay intellectual delays and movement disorders such as ataxia alternating hemiplegia or dystonia 7 Some symptoms may be present all the time like walking difficulties while other signs may come and go like seizures or poor balance 8 These findings can be clustered under three major domains cognition behavior and movement 8 Effects edit The syndrome can cause infantile seizures refractory to anticonvulsive drugs developmental delay acquired microcephaly and neurologic manifestations including spasticity hypotonia and ataxia 9 The frequency severity and types of seizures may vary considerably among GLUT1 deficiency patients and do not necessarily correspond to the severity of other symptoms Most seizures in GLUT1 deficiency patients are not easily treated with anti seizure medications A minority of GLUT1 deficiency patients approximately 10 do not experience seizures 5 Cognitive symptoms often become apparent as developmental milestones are delayed Cognitive deficits range from subtle learning difficulties to severe intellectual disabilities Often speech and language are impaired 5 Behavioral symptoms affect relations with other people and may include short attention span intractability and delays in achieving age appropriate behaviors Sociability with peers however is a strength in GLUT1 deficiency patients 5 Movement symptoms relate to the quality of motor functions Walking may be delayed or difficult because legs are stiff spasticity balance is poor ataxia or posture is twisted dystonia Fine motor deficits may affect speech quality and manipulative skills such as writing These abnormalities may be constant or intermittent paroxysmal 5 Paroxysmal exercise induced dyskinesia PED may also be present 10 Other intermittent symptoms may include headaches confusion and loss of energy Episodes of confusion lack of energy stamina and or muscle twitches may occur particularly during periods without food 7 Some young patients experience occasional abnormal eye movements that may resemble opsoclonus or nystagmus 5 The rapid eye movements that some Glut 1 patients exhibit are rapid multidirectional and there is often a head movement in the same direction as the eye movement 11 These abnormal eye movements were recently named aberrant gaze saccades 11 Hemiplegia or alternating intermittent hemiplegia may occur in some patients and mimic stroke like symptoms 12 Another characteristic of GLUT1 deficiency is that symptoms are sensitive to food e g symptoms that can be temporarily improved by intake of carbohydrates and symptoms may be worse in the morning upon and just after waking 5 All symptoms may be aggravated or triggered by factors such as hunger fatigue heat anxiety and sickness The symptom picture for each patient may evolve and change over time as children with GLUT1 deficiency grow and develop through adolescence and into adulthood 5 Data on adult Glut1DS are just emerging 13 Changes in symptomatology over time include a shift from infantile childhood onset epilepsy to adolescent adult onset movement disorders including PED Genetics editThe GLUT1 protein that transports glucose across the blood brain barrier is made by the SLC2A1 gene located on chromosome 1 8 In GLUT1 deficiency Syndrome one of the two genes is damaged by a mutation and insufficient protein is made As a result insufficient glucose is passing the blood brain barrier Having less functional GLUT1 protein reduces the amount of glucose available to brain cells which affects brain development and function 14 Because glucose is the primary source of fuel for the brain patients with GLUT1 deficiency have insufficient cellular energy to permit normal brain growth and function 8 Around 90 of cases of GLUT1 deficiency syndrome are de novo mutations of the SLC2A1 gene a mutation not present in the parents but present in one of the two copies of the gene in the baby although it can be inherited 15 Glut 1 Deficiency can be inherited in an autosomal dominant manner A person with GLUT1 deficiency syndrome has a 50 chance of passing along the altered SLC2A1 gene to his or her offspring 16 In a study focusing on GLUT1 mice model brain slides physiological glucose concentration was found to be a modulator of frequency oscillations and less frequent 30 50 Hz or gamma oscillations 17 Diagnosis editEarly diagnosis is crucial in order to initiate treatment during the important early stages of brain development To make a proper diagnosis it is important to know the various symptoms of GLUT1 deficiency and how those symptoms evolve with age 18 GLUT1 deficiency is diagnosed based on the clinical features in combination with determining the glucose concentration in the CSF and or a genetic analysis through a lumbar puncture spinal tap 13 A low glucose value in CSF lt 2 2 mmol L or lowered CSF plasma glucose ratio lt 0 4 are indicatieve of GLUT1 deficiency A genetic mutation in the SLC2A1 gene also confirms the diagnosis although mutations have not been identified in approximately 15 of GLUT1 deficiency patients 19 A highly specialized lab test called the red blood cell uptake assay may confirm GLUT1 deficiency but is not commercially available 20 Management editAnti seizure medications are generally not effective since they do not provide nourishment to the starved brain 8 Once diagnosed a medically supervised ketogenic diet is usually recommended as it can help to control seizures 21 The ketogentic diet is the current standard of care treatment with 80 of patients having gt 90 seizure reduction 13 and improving some movement disorders in approximately two thirds of GLUT1 deficiency patients 18 There is also some evidence of some cognitive benefits for GLUT1 deficiency patients on a ketogenic diet and most parents report improved energy alertness balance coordination and concentration 18 especially when the diet is started early in childhood The ketogenic diet is a diet high in fat and low in protein and carbohydrates with up to 90 of calories obtained from fat Since the diet is low in carbohydrates the body gets little glucose normally the main energy source The fat in the diet is converted by the liver in ketone bodies which causes a build up of ketones in the blood stream called ketosis Ketone bodies are transported across the blood brain barrier by other means than the GLUT1 protein and thus serve as an alternative fuel for the brain when glucose is not available 22 While ketogenic diets have been proven effective to control seizures and relieve some movement disorders in many GLUT1 deficiency patients some patients do not respond as well as others In addition some critical symptoms including cognitive deficits and certain movement difficulties tend to persist in GLUT1 deficiency patients treated by a ketogenic diet raising the question whether GLUT1 deficiency is caused simply by a lack of proper brain energy or if there are more complicated and widespread systems and processes affected 18 The ketogenic diet must be carefully crafted and tailored to meet the needs of each patient and reduce the risk of side effects It should only be used under the care of medical professionals and dietitians and it may take some time to establish the ideal ratio of fat versus proteins and carbohydrates and other diet variables for each individual patient to experience optimal tolerance and benefits Variations on the ketogenic diet including the Modified Atkins Diet and diets based on MCT oil have also been shown to be beneficial for some GLUT1 deficiency patients 18 While the classic ketogenic diet is commonly used for younger children compliance with the ketogenic diet can be difficult for older children and adults In recent years the Modified Atkins Diet and MCT oil based diets have gained increasing acceptance among doctors treating these groups and may be more feasible for quality of life and compliance 13 There is growing empirical evidence that these diets can provide at least some of the benefits of the classical ketogenic diet for some GLUT1 deficiency patients 18 Ketone esters are an area of dietary therapy currently under investigation for potential treatment of GLUT1 deficiency and other medical conditions Ketone esters are synthetic ketones that break down into natural ketones when metabolized Ketone esters have been shown in recent research to improve seizures and movement disorders in GLUT1 deficient mice but human studies have not yet been conducted 18 Triheptanoin C7 oil a triglyceride oil synthesized from castor beans 18 is an investigational pharmaceutical grade medical food that has shown potential as a treatment for a number of inherited metabolic diseases When metabolized by the body C7 oil produces ketones similar to those produced on a ketogenic diet in addition to other types of ketones that are thought to fulfill further metabolic requirements in the absence of sufficient glucose 18 A phase 3 clinical trial however failed to find an improvement in patients with GLUT1 DS with disabling movement disorders The inhibition of insulin production to increase glucose in the blood with the medicine diazoxide in combination with continuous glucose monitoring has been successful in one adolescent The increased blood glucose also increases the availability of glucose in the brain through the increased transfer of more glucose through the GLUT1 protein She became seizure free became more physically active and had improved cognition 23 Researchers are studying gene therapy as a possible effective treatment for Glut 1 Deficiency 24 25 Therapies and rehabilitative services are beneficial since most GLUT1 deficiency patients experience movement disturbances as well as speech and language disorders Occupational physical and speech language therapies are standard for most patients especially in childhood 18 Many families greatly benefit from other therapies such as aquatic therapy hippotherapy specific learning strategies and behavioral therapy 18 Glut 1 patients Weak Areas are lowered IQ and adaptive behavior scores expressive language deficits weakness in fine motor skills limited visual attention to details weakness in abstract analytical skills and weakness in transfer of learning to new contexts citation needed Strong Areas include receptive language or understanding social skills fun loving and empathetic personalities perseverance 18 References edit Todor Arsov 2016 Glut 1 deficiency From Pathophysilogy ad genetics to abroad clinical spectrum Sanamed 11 2 151 155 doi 10 5937 sanamed1602151A a b Understanding Glucose Transporter Type 1 Deficiency Syndrome Glut1 DS Current Management and Future Approaches Epilepsy Foundation Retrieved 2018 01 31 Symonds 2019 Incidence and phenotypes of childhood onset genetic epilepsies a prospective population based national cohort Brain 142 8 2303 2318 doi 10 1093 brain awz195 PMC 6658850 PMID 31302675 a b GLUT1 deficiency syndrome Genetics Home Reference Retrieved 10 October 2011 a b c d e f g h i Brochures GLUT1 deficiency Foundation Retrieved 2018 01 25 Wang Pascual Vivo Glucose Transporter Type 1 Deficiency Syndrome GeneReviews a href Template Cite journal html title Template Cite journal cite journal a CS1 maint multiple names authors list link a b c Reference Genetics Home GLUT1 deficiency syndrome Genetics Home Reference Retrieved 2018 01 25 a b c d e Reaching for a brighter future PDF GLUT1 deficiency Foundation Archived from the original PDF on 2015 10 02 Retrieved 19 June 2017 Ticus I Cano A Villeneuve N Milh M Mancini J Chabrol B August 2008 GLUT 1 deficiency syndrome or De Vivo disease a case report Arch Pediatr in French 15 8 1296 9 doi 10 1016 j arcped 2008 04 024 PMID 18556184 Wang Dong Pascual Juan M De Vivo Darryl 1993 Glucose Transporter Type 1 Deficiency Syndrome In Adam Margaret P Ardinger Holly H Pagon Roberta A Wallace Stephanie E Bean Lora J H Stephens Karen Amemiya Anne eds GeneReviews Seattle WA University of Washington Seattle PMID 20301603 a b Pearson Toni S Pons Roser Engelstad Kristin Kane Steven A Goldberg Michael E Vivo Darryl C De 2017 04 25 Paroxysmal eye head movements in GLUT1 deficiency syndrome Neurology 88 17 1666 1673 doi 10 1212 WNL 0000000000003867 ISSN 0028 3878 PMC 5405761 PMID 28341645 Braakman H M H Nicolai J Willemsen M A A P 2017 06 01 Stroke like episodes add to the phenotypic spectrum of GLUT1 deficiency syndrome European Journal of Paediatric Neurology 21 e176 doi 10 1016 j ejpn 2017 04 995 ISSN 1090 3798 a b c d Klepper Joerg 2020 Glut1 Deficiency Syndrome Glut1DS State of the art in 2020 and recommendations of the international Glut1DS study group Epilepsia Open 5 3 354 365 doi 10 1002 epi4 12414 PMC 7469861 PMID 32913944 Reference Genetics Home GLUT1 deficiency syndrome Genetics Home Reference Retrieved 2017 06 15 Professional Guide PDF GLUT1 deficiency Foundation Archived from the original PDF on 2015 10 03 Retrieved 19 June 2017 Glucose transporter type 1 deficiency syndrome Genetic and Rare Diseases Information Center GARD an NCATS Program rarediseases info nih gov Archived from the original on 2018 02 01 Retrieved 2018 01 31 Solis Elysandra M Good Levi B Vazquez Rafael Granja Patnaik Sourav Hernandez Reynoso Ana G Ma Qian Angulo Gustavo Dobariya Aksharkumar Cogan Stuart F Pancrazio Joseph J Pascual Juan M Jakkamsetti Vikram 2023 Isolation of the murine Glut1 deficient thalamocortical circuit wavelet characterization and reverse glucose dependence of low and gamma frequency oscillations Frontiers in Neuroscience 17 1191492 doi 10 3389 fnins 2023 1191492 ISSN 1662 4548 PMC 10565352 PMID 37829723 a b c d e f g h i j k l Brochures GLUT1 deficiency Foundation Retrieved 2018 01 31 Professional Guide GLUT1 deficiency Foundation Archived from the original on 2018 02 01 Retrieved 2018 01 31 Metafora Diagnostics tests www metafora biosystems com in French Archived from the original on 2018 02 01 Retrieved 2018 01 31 De Vivo Darryl C Trifiletti Rosario R Jacobson Ronald I Ronen Gabriel M Behmand Ramin A Harik Sami I 5 September 1991 Defective Glucose Transport across the Blood Brain Barrier as a Cause of Persistent Hypoglycorrhachia Seizures and Developmental Delay New England Journal of Medicine 325 10 703 709 doi 10 1056 NEJM199109053251006 PMID 1714544 Brockmann Knut 2011 07 01 Towards a more palatable treatment for GLUT1 deficiency syndrome Developmental Medicine amp Child Neurology 53 7 580 581 doi 10 1111 j 1469 8749 2011 03946 x ISSN 1469 8749 PMID 21585366 Logel 2021 Exploring diazoxide and continuous glucose monitoring as treatment for Glut1 deficiency syndrome Ann Clin Transl Neurol 8 11 2205 2209 doi 10 1002 acn3 51462 PMC 8607448 PMID 34612610 Sanofi Awarded New Funding to Monani Lab to Study Glucose Transporter 1 Deficiency Syndrome Pathology 2017 06 19 Retrieved 2018 01 31 GLUT1 deficiency Foundation 2018 01 30 Dr Umrao Monani archived from the original on 2021 12 21 retrieved 2018 01 31 a href Template Citation html title Template Citation citation a CS1 maint numeric names authors list link External links editGeneReview NIH UW entry on Glucose Transporter Type 1 Deficiency Syndrome Retrieved from https en wikipedia org w index php title GLUT1 deficiency amp oldid 1187386261, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.