GM1 gangliosidoses

Last updated
GM1 gangliosidoses
Other namesGM1 gangliosidosis
Specialty Endocrinology   OOjs UI icon edit-ltr-progressive.svg

The GM1 gangliosidoses, usually shortened to GM1, are gangliosidoses caused by mutation in the GLB1 gene resulting in a deficiency of beta-galactosidase. The deficiency causes abnormal storage of acidic lipid materials in cells of the central and peripheral nervous systems, but particularly in the nerve cells, resulting in progressive neurodegeneration. GM1 is a rare lysosomal storage disorder with a prevalence of 1 to every 100,000 to 200,000 live births worldwide, although rates are higher in some regions. [1] [2] [3] [4]

Contents

Cause

GM1 Gangliosidoses disorders are caused by mutations in the GLB1 gene, which codes for lysosomal hydrolase, acid beta-galactosidase (β-gal). Low levels of β-gal cause an accumulation of GM1 gangliosides. They are inherited, autosomal recessive sphingolipidoses, a class of lipid storage disorders. [5] [6]

Diagnosis

Diagnosis of GM1 can be obtained by genetic and enzymatic testing.

Types

GM1 has three forms classified by age of onset. [5]

Early infantile GM1

Symptoms of early infantile GM1 (the most severe subtype, with onset shortly after birth) may include neurodegeneration, seizures, liver enlargement (hepatomegaly), spleen enlargement (splenomegaly), coarsening of facial features, skeletal irregularities, joint stiffness, distended abdomen, muscle weakness, exaggerated startle response to sound, and problems with gait. [5] [6]

About half of affected patients develop cherry-red spots in the eye.

Children may be deaf and blind by age 1 and often die by age 3 from cardiac complications or pneumonia. [7]

  • Early psychomotor deterioration: decreased activity and lethargy in the first weeks; never sit; feeding problems - failure to thrive; visual failure (nystagmus noted) by 6 months; initial hypotonia; later spasticity with pyramidal signs; secondary microcephaly develops; decerebrate rigidity by 1 year and death by age 1–2 years (due to pneumonia and respiratory failure); some have hyperacusis.
  • Macular cherry-red spots in 50% by 6–10 months; corneal opacities in some
  • Facial dysmorphology: frontal bossing, wide nasal bridge, facial edema (puffy eyelids); peripheral edema, epicanthus, long upper lip, microretrognathia, gingival hypertrophy (thick alveolar ridges), macroglossia
  • Hepatomegaly by 6 months and splenomegaly later; some have cardiac failure
  • Skeletal deformities: flexion contractures noted by 3 months; early subperiosteal bone formation (may be present at birth); diaphyseal widening later; demineralization; thoracolumbar vertebral hypoplasia and beaking at age 3–6 months; kyphoscoliosis. *Dysostosis multiplex (as in the mucopolysaccharidoses)
  • 10–80% of peripheral lymphocytes are vacuolated; foamy histiocytes in bone marrow; visceral mucopolysaccharide storage similar to that in Hurler disease; GM1 storage in cerebral gray matter is 10-fold elevated (20–50-fold increased in viscera)
  • Galactose-containing oligosacchariduria and moderate keratan sulfaturia
  • Morquio disease Type B: Mutations with higher residual beta-galactosidase activity for the GM1 substrate than for keratan sulfate and other galactose-containing oligosaccharides have minimal neurologic involvement but severe dysostosis resembling Morquio disease type A (Mucopolysaccharidosis type 4). [8]

Late infantile/Juvenile GM1

Onset of late infantile GM1 is typically between ages 1 and 3 years. The juvenile form may be diagnosed into childhood. Some children live into adolescence or early adulthood. This subtype is characterized by a trajectory in which some developmental skills are gained, then they stabilize and delays occur, and these are followed by regression. Early symptoms include difficulty crawling and walking, hypotonia, speech and swallowing problems, and seizures. Neurological symptoms include ataxia, seizures, dementia, and difficulties with speech. [4] [5] [6]

Adult GM1

Onset of adult GM1 is typically in adolescence or adulthood and is the slowest progressing of the subtypes.

Symptoms include muscle atrophy, neurological complications that are less severe and progress at a slower rate than in other forms of the disorder, corneal clouding in some patients, and dystonia (sustained muscle contractions that cause twisting and repetitive movements or abnormal postures). Angiokeratomas may develop on the lower part of the trunk of the body. Most patients have a normal size liver and spleen. Prenatal diagnosis is possible by measurement of Acid Beta Galactosidase in cultured amniotic cells.[ citation needed ]

Treatment

Treatment for GM1 is symptom-based and palliative. There is no cure for GM1, although several gene therapy trials are underway. [9] More information for these can be found at ClinicalTrials.gov.

Related Research Articles

<span class="mw-page-title-main">Tay–Sachs disease</span> Human medical condition

Tay–Sachs disease is a genetic disorder that results in the destruction of nerve cells in the brain and spinal cord. The most common form is infantile Tay–Sachs disease, which becomes apparent around the age of three to six months of age, with the baby losing the ability to turn over, sit, or crawl. This is then followed by seizures, hearing loss, and inability to move, with death usually occurring by the age of three to five. Less commonly, the disease may occur in later childhood or adulthood. These forms tend to be less severe, but the juvenile form typically results in death by age 15.

<span class="mw-page-title-main">Mucopolysaccharidosis</span> Medical condition

Mucopolysaccharidoses are a group of metabolic disorders caused by the absence or malfunctioning of lysosomal enzymes needed to break down molecules called glycosaminoglycans (GAGs). These long chains of sugar carbohydrates occur within the cells that help build bone, cartilage, tendons, corneas, skin and connective tissue. GAGs are also found in the fluids that lubricate joints.

<span class="mw-page-title-main">Gaucher's disease</span> Medical condition

Gaucher's disease or Gaucher disease (GD) is a genetic disorder in which glucocerebroside accumulates in cells and certain organs. The disorder is characterized by bruising, fatigue, anemia, low blood platelet count and enlargement of the liver and spleen, and is caused by a hereditary deficiency of the enzyme glucocerebrosidase, which acts on glucocerebroside. When the enzyme is defective, glucocerebroside accumulates, particularly in white blood cells and especially in macrophages. Glucocerebroside can collect in the spleen, liver, kidneys, lungs, brain, and bone marrow.

<span class="mw-page-title-main">Lysosomal storage disease</span> Medical condition

Lysosomal storage diseases are a group of over 70 rare inherited metabolic disorders that result from defects in lysosomal function. Lysosomes are sacs of enzymes within cells that digest large molecules and pass the fragments on to other parts of the cell for recycling. This process requires several critical enzymes. If one of these enzymes is defective due to a mutation, the large molecules accumulate within the cell, eventually killing it.

<span class="mw-page-title-main">Sanfilippo syndrome</span> Genetic disorder

Sanfilippo syndrome, also known as mucopolysaccharidosis type III (MPS III), is a rare autosomal recessive lysosomal storage disease that primarily affects the brain and spinal cord. It is caused by a buildup of large sugar molecules called glycosaminoglycans (AKA GAGs, or mucopolysaccharides) in the body's lysosomes.

<span class="mw-page-title-main">Krabbe disease</span> Medical condition

Krabbe disease (KD) is a rare and often fatal lysosomal storage disease that results in progressive damage to the nervous system. KD involves dysfunctional metabolism of sphingolipids and is inherited in an autosomal recessive pattern. The disease is named after the Danish neurologist Knud Krabbe (1885–1961).

<span class="mw-page-title-main">Sandhoff disease</span> Medical condition

Sandhoff disease is a lysosomal genetic, lipid storage disorder caused by the inherited deficiency to create functional beta-hexosaminidases A and B. These catabolic enzymes are needed to degrade the neuronal membrane components, ganglioside GM2, its derivative GA2, the glycolipid globoside in visceral tissues, and some oligosaccharides. Accumulation of these metabolites leads to a progressive destruction of the central nervous system and eventually to death. The rare autosomal recessive neurodegenerative disorder is clinically almost indistinguishable from Tay–Sachs disease, another genetic disorder that disrupts beta-hexosaminidases A and S. There are three subsets of Sandhoff disease based on when first symptoms appear: classic infantile, juvenile and adult late onset.

<span class="mw-page-title-main">GM2-gangliosidosis, AB variant</span> Medical condition

GM2-gangliosidosis, AB variant is a rare, autosomal recessive metabolic disorder that causes progressive destruction of nerve cells in the brain and spinal cord. It has a similar pathology to Sandhoff disease and Tay–Sachs disease. The three diseases are classified together as the GM2 gangliosidoses, because each disease represents a distinct molecular point of failure in the activation of the same enzyme, beta-hexosaminidase. AB variant is caused by a failure in the gene that makes an enzyme cofactor for beta-hexosaminidase, called the GM2 activator.

<span class="mw-page-title-main">Morquio syndrome</span> Medical condition

Morquio syndrome, also known as Mucopolysaccharidosis Type IV (MPS IV), is a rare metabolic disorder in which the body cannot process certain types of sugar molecules called glycosaminoglycans (AKA GAGs, or mucopolysaccharides). In Morquio syndrome, the specific GAG which builds up in the body is called keratan sulfate. This birth defect, which is autosomal recessive, is a type of lysosomal storage disorder. The buildup of GAGs in different parts of the body causes symptoms in many different organ systems. In the US, the incidence rate for Morquio syndrome is estimated at between 1 in 200,000 and 1 in 300,000 live births.

A lipid storage disorder is any one of a group of inherited metabolic disorders in which harmful amounts of fats or lipids accumulate in some body cells and tissues. People with these disorders either do not produce enough of one of the enzymes needed to metabolize and break down lipids or, they produce enzymes that do not work properly. Over time, the buildup of fats may cause permanent cellular and tissue damage, particularly in the brain, peripheral nervous system, liver, spleen, and bone marrow.

<span class="mw-page-title-main">Sphingolipidoses</span> Medical condition

Sphingolipidoses are a class of lipid storage disorders or degenerative storage disorders caused by deficiency of an enzyme that is required for the catabolism of lipids that contain ceramide, also relating to sphingolipid metabolism. The main members of this group are Niemann–Pick disease, Fabry disease, Krabbe disease, Gaucher disease, Tay–Sachs disease and metachromatic leukodystrophy. They are generally inherited in an autosomal recessive fashion, but notably Fabry disease is X-linked recessive. Taken together, sphingolipidoses have an incidence of approximately 1 in 10,000, but substantially more in certain populations such as Ashkenazi Jews. Enzyme replacement therapy is available to treat mainly Fabry disease and Gaucher disease, and people with these types of sphingolipidoses may live well into adulthood. The other types are generally fatal by age 1 to 5 years for infantile forms, but progression may be mild for juvenile- or adult-onset forms.

The GM2 gangliosidoses are a group of three related genetic disorders that result from a deficiency of the enzyme beta-hexosaminidase. This enzyme catalyzes the biodegradation of fatty acid derivatives known as gangliosides. The diseases are better known by their individual names: Tay–Sachs disease, AB variant, and Sandhoff disease.

<span class="mw-page-title-main">HEXB</span> Protein-coding gene in the species Homo sapiens

Beta-hexosaminidase subunit beta is an enzyme that in humans is encoded by the HEXB gene.

<span class="mw-page-title-main">Cathepsin A</span>

Cathepsin A is an enzyme that is classified both as a cathepsin and a carboxypeptidase. In humans, it is encoded by the CTSA gene.

<span class="mw-page-title-main">GLB1</span> Protein

Galactosidase, beta 1, also known as GLB1, is a protein which in humans is encoded by the GLB1 gene.

<span class="mw-page-title-main">HEXA</span> Protein-coding gene in the species Homo sapiens

Hexosaminidase A (alpha polypeptide), also known as HEXA, is an enzyme that in humans is encoded by the HEXA gene, located on the 15th chromosome.

<span class="mw-page-title-main">Galactosialidosis</span> Rare disease

Galactosialidosis, also known as neuraminidase deficiency with beta-galactosidase deficiency, is a genetic lysosomal storage disease. It is caused by a mutation in the CTSA gene which leads to a deficiency of enzymes β-galactosidase and neuraminidase. This deficiency inhibits the lysosomes of cells from functioning properly, resulting in the accumulation of toxic matter within the cell. Hallmark symptoms include abnormal spinal structure, vision problems, coarse facial features, hearing impairment, and intellectual disability. Because galactosialidosis involves the lysosomes of all cells, it can affect various areas of the body, including the brain, eyes, bones, and muscles. Depending on the patient's age at the onset of symptoms, the disease consists of three subtypes: early infantile, late infantile, and juvenile/adult. This condition is considered rare, with most cases having been in the juvenile/adult group of patients.

<span class="mw-page-title-main">Schindler disease</span> Medical condition

Schindler disease, also known as Kanzaki disease and alpha-N-acetylgalactosaminidase deficiency is a rare disease found in humans. This lysosomal storage disorder is caused by a deficiency in the enzyme alpha-NAGA (alpha-N-acetylgalactosaminidase), attributable to mutations in the NAGA gene on chromosome 22, which leads to excessive lysosomal accumulation of glycoproteins. A deficiency of the alpha-NAGA enzyme leads to an accumulation of glycosphingolipids throughout the body. This accumulation of sugars gives rise to the clinical features associated with this disorder. Schindler disease is an autosomal recessive disorder, meaning that one must inherit an abnormal allele from both parents in order to have the disease.

<span class="mw-page-title-main">Coarse facial features</span> Medical condition

Coarse facial features is a constellation of facial features that are present in many inborn errors of metabolism.

<span class="mw-page-title-main">Glycoside hydrolase family 35</span>

In molecular biology, glycoside hydrolase family 35 is a family of glycoside hydrolases.

References

  1. Caciotti A, Garman SC, Rivera-Colón Y, Procopio E, Catarzi S, Ferri L, et al. (July 2011). "GM1 gangliosidosis and Morquio B disease: an update on genetic alterations and clinical findings". Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 1812 (7): 782–790. doi:10.1016/j.bbadis.2011.03.018. PMC   3210552 . PMID   21497194.
  2. Jarnes Utz JR, Kim S, King K, Ziegler R, Schema L, Redtree ES, Whitley CB (June 2017). "Infantile gangliosidoses: Mapping a timeline of clinical changes". Molecular Genetics and Metabolism. 121 (2): 170–179. doi:10.1016/j.ymgme.2017.04.011. PMC   5727905 . PMID   28476546.
  3. Regier DS, Kwon HJ, Johnston J, Golas G, Yang S, Wiggs E, et al. (March 2016). "MRI/MRS as a surrogate marker for clinical progression in GM1 gangliosidosis". American Journal of Medical Genetics. Part A. 170 (3): 634–644. doi:10.1002/ajmg.a.37468. PMID   26646981. S2CID   19466710.
  4. 1 2 Brunetti-Pierri N, Scaglia F (August 2008). "GM1 gangliosidosis: review of clinical, molecular, and therapeutic aspects". Molecular Genetics and Metabolism. 94 (4): 391–396. doi:10.1016/j.ymgme.2008.04.012. PMID   18524657.
  5. 1 2 3 4 Rha AK, Maguire AS, Martin DR (2021-04-09). "GM1 Gangliosidosis: Mechanisms and Management". The Application of Clinical Genetics. 14: 209–233. doi:10.2147/TACG.S206076. PMC   8044076 . PMID   33859490.
  6. 1 2 3 Nicoli ER, Annunziata I, d'Azzo A, Platt FM, Tifft CJ, Stepien KM (2021-09-03). "GM1 Gangliosidosis-A Mini-Review". Frontiers in Genetics. 12: 734878. doi: 10.3389/fgene.2021.734878 . PMC   8446533 . PMID   34539759.
  7. Bley AE, Giannikopoulos OA, Hayden D, Kubilus K, Tifft CJ, Eichler FS (November 2011). "Natural history of infantile G(M2) gangliosidosis". Pediatrics. 128 (5): e1233–e1241. doi:10.1542/peds.2011-0078. PMC   3208966 . PMID   22025593.
  8. Lyon G, Adams RD, Kolodny EH (1996). Neurology of Hereditary Metabolic Diseases of Children (2nd ed.). New York: McGraw-Hill, Health Professions Division. pp. 53–55. ISBN   978-0-07-000389-7.
  9. Soo, Sonja (November 22, 2022). "Tea party reunion marks milestone in treatment of rare genetic disorder". National Human Genome Research Institute.