1
|
Baskar D, Christopher R, Arunachal G, Anudeep DDS, Mounika A, Sangeeth TA, Polavarapu K, Shalini BS, Joshi T, Sanka SB, Nashi S, Raja P, Chowdary RM, Yadav R, Nalini A, Vengalil S. Acute myeloneuropathy due to Glutaric aciduria-1: Expanding the phenotypic spectrum. Glob Med Genet 2025; 12:100036. [PMID: 40027240 PMCID: PMC11871448 DOI: 10.1016/j.gmg.2025.100036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/05/2025] [Indexed: 03/05/2025] Open
Abstract
Glutaric aciduria type-1 (GA-1) is a rare metabolic disorder due to mutation in GCDH gene resulting in varied clinical manifestations. Here we report a case of late-onset GA-1 with acute myelo-neuropathy and chronic renal failure. Institutional ethics committee approval was obtained and genetic analysis was done by clinical exome sequencing. Here we present 19 year-old-adolescent male with chronic renal disease for 2 years presented with 5 months history of sudden onset weakness of proximal and distal lower limbs and bladder retention. This was preceded by recurrent episodes of vomiting. On clinical examination he had features of myeloneuropathy. Laboratory evaluation showed significant elevation of blood glutaryl carnitine with very low free carnitine, while extensive white matter signal changes with diffusion restriction, subependymal nodules and involvement of internal capsule were evidenced on brain magnetic resonance imaging. Diagnosis was confirmed by clinical exome sequencing which showed a pathogenic homozygous missense mutation in exon 11 of GCDH gene (c .120 C>T, p.His403Tyr). This report expands phenotypic spectrum of GA-1 to include late onset acute myelo-neuropathy with chronic renal failure. A high index of suspicion is required since early treatment might decelerate further disease progression.
Collapse
Affiliation(s)
- Dipti Baskar
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Rita Christopher
- Integrative Medical Research, PES University Institute of Medical Sciences and Research, Bengaluru and Former Dean (Basic Sciences) & Senior Professor (Retired), Department of Neurochemistry National Institute of Mental Health & Neuro Sciences (NIMHANS) Bengaluru 560029, India
| | - Gautham Arunachal
- Department of Human Genetics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | | | - Ambati Mounika
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - TA Sangeeth
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Kiran Polavarapu
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa and Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - BS Shalini
- Department of Neuroradiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Tarachand Joshi
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Sai Bhargava Sanka
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Saraswati Nashi
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Pritam Raja
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | | | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| |
Collapse
|
2
|
Richard E, Marchuk H, Álvarez M, He W, Chen X, Desviat LR, Zhang GF. Metabolic flux analysis in hiPSC-CMs reveals insights into cardiac dysfunction in propionic acidemia. Cell Mol Life Sci 2025; 82:137. [PMID: 40172673 PMCID: PMC11965053 DOI: 10.1007/s00018-025-05661-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 04/04/2025]
Abstract
Propionic acidemia is an inborn error of metabolism caused by mutations in either the PCCA or PCCB genes. Patients with propionic acidemia experience a range of complications, including life-threatening cardiac dysfunctions. However, the pathological mechanisms underlying propionic acidemia-associated cardiac diseases remain largely unknown. To gain insights into the metabolic alterations in propionic acidemia, we studied human induced pluripotent stem cell-derived cardiomyocytes generated from a patient with propionic acidemia with two pathogenic PCCA mutations (p.Cys616_Val633del and p.Gly477Glufs9*) and from a healthy individual. Using stable isotope-based metabolic flux analysis, we confirmed that the PCCA mutations lead to impaired propionyl-CoA carboxylase activity in human induced pluripotent stem cell-derived cardiomyocytes. In addition to being converted to propionylcarnitine, the accumulated propionyl-CoA can also be hydrolyzed to propionate and exported out of the cell, serving as a secondary "pressure valve" to regulate cellular propionyl-CoA levels. Interestingly, the deficiency of propionyl-CoA carboxylase was found to shift fuel metabolism from fatty acid oxidation to increased glucose metabolism human in induced pluripotent stem cell-derived cardiomyocytes from patients with propionic acidemia. This metabolic switch is less energy-efficient and may contribute to the development of chronic cardiac dysfunction in patients with propionic acidemia.
Collapse
Affiliation(s)
- Eva Richard
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, CIBERER, IdiPaz, IUBM, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049, Madrid, Spain.
| | - Hannah Marchuk
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Mar Álvarez
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Wentao He
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Xiaoxin Chen
- Surgical Research Lab, Department of Surgery, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
- Coriell Institute for Medical Research, Camden, NJ, 08103, USA
- MD Anderson Cancer Center at Cooper, Camden, NJ, 08103, USA
| | - Lourdes R Desviat
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, CIBERER, IdiPaz, IUBM, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049, Madrid, Spain
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA.
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University Medical Center, Durham, NC, 27701, USA.
| |
Collapse
|
3
|
Martín-Hernández E, Bellusci M, Pérez-Mohand P, Correcher Medina P, Blasco-Alonso J, Morais-López A, de las Heras J, Meavilla Olivas SM, Dougherty-de Miguel L, Couce ML, Villarroya EC, García Jiménez MC, Moreno-Lozano PJ, Vives I, Gil-Campos M, Stanescu S, Ceberio-Hualde L, Camprodón M, Cortès-Saladelafont E, López-Urdiales R, Murray Hurtado M, Márquez Armenteros AM, Sierra Córcoles C, Peña-Quintana L, Ruiz-Pons M, Alcalde C, Castellanos-Pinedo F, Dios E, Barrio-Carreras D, Martín-Cazaña M, García-Peris M, Andrade JD, García-Volpe C, de los Santos M, García-Cazorla A, del Toro M, Felipe-Rucián A, Comino Monroy MJ, Sánchez-Pintos P, Matas A, Gil Ortega D, Martín-Rivada Á, Bergua A, Belanger-Quintana A, Vitoria I, Yahyaoui R, Pérez B, Morales-Conejo M, Quijada-Fraile P. Understanding the Natural History and the Effects of Current Therapeutic Strategies on Urea Cycle Disorders: Insights from the UCD Spanish Registry. Nutrients 2025; 17:1173. [PMID: 40218931 PMCID: PMC11990916 DOI: 10.3390/nu17071173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: The present study updates the Spanish registry of patients with urea cycle disorders (UCD), originally established in 2013, to provide comprehensive epidemiological data and evaluate the impact of therapeutic strategies and newborn screening (NBS) on clinical outcomes. Methods: This retrospective, multicenter study focuses on 255 Spanish UCD patients. It includes all living and deceased cases up to February 2024, analyzing demographic, clinical, and biochemical variables. Results: The incidence of UCD in Spain over the past decade was 1:36,063 births. The most common defects were ornithine transcarbamylase deficiency (OTCD) and argininosuccinate synthetase deficiency. Early-onset (EO) cases comprised 32.7%, and 10.6% were diagnosed through NBS. Global mortality was 14.9%, higher in carbamoylphosphate synthetase 1 deficiency (36.8%) and male OTCD patients (32.1%) compared to other defects (p = 0.013). EO cases presented a higher mortality rate (35.8%) than late-onset (LO) cases (7.1%) (p < 0.0001). The median ammonia level in deceased patients was higher at 1058 µmol/L (IQR 410-1793) than in survivors at 294 µmol/L (IQR 71-494) (p < 0.0001). Diagnosis through NBS improved survival and reduced neurological impairment compared to symptomatic diagnosis. Neurological impairment occurred in 44% of patients, with worse neurological outcomes observed in patients with argininosuccinate lyase deficiency, arginase 1 deficiency, hyperornithinemia-hyperammonemia-homocitrullinuria, EO presentations, pre-2014 diagnosis, and patients with higher levels of ammonia at diagnosis. Among transplanted patients (20.6%), survival was 95.2%, with no significant neurological differences compared to non-transplanted patients. Conclusions: This updated analysis highlights the positive impact of NBS and advanced treatments on mortality and neurologic outcomes. Persistent neurological challenges underscore the need for further therapeutic strategies.
Collapse
Affiliation(s)
- Elena Martín-Hernández
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Marcello Bellusci
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Patricia Pérez-Mohand
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Patricia Correcher Medina
- Unidad de Nutrición y Metabolopatías, Hospital Universitario La Fé, 46026 Valencia, Spain; (P.C.M.); (M.G.-P.); (I.V.)
| | - Javier Blasco-Alonso
- Unidad de Gastroenterología, Hepatología y Nutrición Pediátrica, Laboratorio de Bioquímica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.B.-A.); (R.Y.)
| | - Ana Morais-López
- Unidad de Nutrición Infantil y Enfermedades Metabólicas, Hospital Universitario La Paz, 28046 Madrid, Spain; (A.M.-L.); (J.D.A.); (A.B.)
| | - Javier de las Heras
- Hereditary Metabolic Diseases Unit, Hospital Universitario Cruces, MetabERN, Biobizkaia Health Research Institute, 48903 Bilbao, Spain; (J.d.l.H.); (L.C.-H.)
| | - Silvia María Meavilla Olivas
- Unidad de Enfermedades Metabólicas, Hospital San Joan de Déu, MetabERN, CIBERER, 08035 Barcelona, Spain; (S.M.M.O.); (C.G.-V.); (M.d.l.S.); (A.G.-C.)
| | - Lucy Dougherty-de Miguel
- Unidad de Enfermedades Metabólicas, Hospital Vall d’Hebrón, MetabERN, VHIR, 08035 Barcelona, Spain; (L.D.-d.M.); (M.C.); (M.d.T.); (A.F.-R.)
| | - Maria Luz Couce
- Unidad de Enfermedades Metabólicas, Hospital Clínico Universitario de Santiago, MetabERN, IDIS, 15706 Santiago de Compostela, Spain; (M.L.C.); (P.S.-P.)
| | - Elvira Cañedo Villarroya
- Unidad de Nutrición y Enfermedades Metabólicas, Hospital Universitario Niño Jesús, 28009 Madrid, Spain;
| | | | - Pedro Juan Moreno-Lozano
- Unidad de Errores Congénitos del Metabolismo en el Adulto, Medicina Interna, Hospital Clínic, 08036 Barcelona, Spain; (P.J.M.-L.); (A.M.)
| | - Inmaculada Vives
- Gastroenterología Pediátrica, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain; (I.V.); (D.G.O.)
| | - Mercedes Gil-Campos
- Unidad de Metabolismo, Hospital Universitario Reina Sofía, IMIBIC, UCO, 14004 Córdoba, Spain; (M.G.-C.); (M.J.C.M.)
| | - Sinziana Stanescu
- Unidad de Enfermedades Metabólicas, Hospital Ramón y Cajal, MetabERN, 28034 Madrid, Spain; (S.S.); (A.B.-Q.)
| | - Leticia Ceberio-Hualde
- Hereditary Metabolic Diseases Unit, Hospital Universitario Cruces, MetabERN, Biobizkaia Health Research Institute, 48903 Bilbao, Spain; (J.d.l.H.); (L.C.-H.)
| | - María Camprodón
- Unidad de Enfermedades Metabólicas, Hospital Vall d’Hebrón, MetabERN, VHIR, 08035 Barcelona, Spain; (L.D.-d.M.); (M.C.); (M.d.T.); (A.F.-R.)
| | - Elisenda Cortès-Saladelafont
- Unidad de Neurología Pediátrica y Enfermedades Metabólicas, Hospital Germans Trias i Pujol, 08916 Barcelona, Spain;
| | - Rafael López-Urdiales
- Departamento de Endocrinología y Nutrición, Hospital de Bellvitge, 08907 Barcelona, Spain;
| | - Mercedes Murray Hurtado
- Unidad de Nutrición y Metabolopatías, Pediatría, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain; (M.M.H.); (Á.M.-R.)
| | | | | | - Luis Peña-Quintana
- Gastroenterología y Nutrición Pediátrica, Complejo Hospitalario Universitario Insular-Materno Infantil, CIBEROBN-ISCIII, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain;
| | - Mónica Ruiz-Pons
- Unidad de Nutrición y Enfermedades Metabólicas, Pediatría, Hospital Universitario Virgen de la Candelaria, 38010 Tenerife, Spain;
| | - Carlos Alcalde
- Servicio de Pediatría, Hospital Universitario Río Ortega, 47012 Valladolid, Spain;
| | | | - Elena Dios
- Endocrinología y Enfermedades Metabólicas, MetabERN, Hospital Virgen del Rocío, 41013 Sevilla, Spain;
| | - Delia Barrio-Carreras
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - María Martín-Cazaña
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Mónica García-Peris
- Unidad de Nutrición y Metabolopatías, Hospital Universitario La Fé, 46026 Valencia, Spain; (P.C.M.); (M.G.-P.); (I.V.)
| | - José David Andrade
- Unidad de Nutrición Infantil y Enfermedades Metabólicas, Hospital Universitario La Paz, 28046 Madrid, Spain; (A.M.-L.); (J.D.A.); (A.B.)
| | - Camila García-Volpe
- Unidad de Enfermedades Metabólicas, Hospital San Joan de Déu, MetabERN, CIBERER, 08035 Barcelona, Spain; (S.M.M.O.); (C.G.-V.); (M.d.l.S.); (A.G.-C.)
| | - Mariela de los Santos
- Unidad de Enfermedades Metabólicas, Hospital San Joan de Déu, MetabERN, CIBERER, 08035 Barcelona, Spain; (S.M.M.O.); (C.G.-V.); (M.d.l.S.); (A.G.-C.)
| | - Angels García-Cazorla
- Unidad de Enfermedades Metabólicas, Hospital San Joan de Déu, MetabERN, CIBERER, 08035 Barcelona, Spain; (S.M.M.O.); (C.G.-V.); (M.d.l.S.); (A.G.-C.)
| | - Mireia del Toro
- Unidad de Enfermedades Metabólicas, Hospital Vall d’Hebrón, MetabERN, VHIR, 08035 Barcelona, Spain; (L.D.-d.M.); (M.C.); (M.d.T.); (A.F.-R.)
| | - Ana Felipe-Rucián
- Unidad de Enfermedades Metabólicas, Hospital Vall d’Hebrón, MetabERN, VHIR, 08035 Barcelona, Spain; (L.D.-d.M.); (M.C.); (M.d.T.); (A.F.-R.)
| | - María José Comino Monroy
- Unidad de Metabolismo, Hospital Universitario Reina Sofía, IMIBIC, UCO, 14004 Córdoba, Spain; (M.G.-C.); (M.J.C.M.)
| | - Paula Sánchez-Pintos
- Unidad de Enfermedades Metabólicas, Hospital Clínico Universitario de Santiago, MetabERN, IDIS, 15706 Santiago de Compostela, Spain; (M.L.C.); (P.S.-P.)
| | - Ana Matas
- Unidad de Errores Congénitos del Metabolismo en el Adulto, Medicina Interna, Hospital Clínic, 08036 Barcelona, Spain; (P.J.M.-L.); (A.M.)
| | - David Gil Ortega
- Gastroenterología Pediátrica, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain; (I.V.); (D.G.O.)
| | - Álvaro Martín-Rivada
- Unidad de Nutrición y Metabolopatías, Pediatría, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain; (M.M.H.); (Á.M.-R.)
| | - Ana Bergua
- Unidad de Nutrición Infantil y Enfermedades Metabólicas, Hospital Universitario La Paz, 28046 Madrid, Spain; (A.M.-L.); (J.D.A.); (A.B.)
| | - Amaya Belanger-Quintana
- Unidad de Enfermedades Metabólicas, Hospital Ramón y Cajal, MetabERN, 28034 Madrid, Spain; (S.S.); (A.B.-Q.)
| | - Isidro Vitoria
- Unidad de Nutrición y Metabolopatías, Hospital Universitario La Fé, 46026 Valencia, Spain; (P.C.M.); (M.G.-P.); (I.V.)
| | - Raquel Yahyaoui
- Unidad de Gastroenterología, Hepatología y Nutrición Pediátrica, Laboratorio de Bioquímica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.B.-A.); (R.Y.)
| | - Belén Pérez
- Centro de Diagnóstico de Enfermedades Moleculares, IdiPAZ, CIBERER, Universidad Autónoma Madrid, 28049 Madrid, Spain;
| | - Montserrat Morales-Conejo
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Pilar Quijada-Fraile
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| |
Collapse
|
4
|
Burkhardt T, Kastor KL, Christ S, Opladen T, Staufner C, Beck‐Wödl S, Haack T, Hempel M, Seitz A, Kölker S. An atypical presentation in a child with propionic acidemia? Better think twice! JIMD Rep 2025; 66:e12464. [PMID: 40078824 PMCID: PMC11897901 DOI: 10.1002/jmd2.12464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 03/14/2025] Open
Abstract
This report details the case of an infant with confirmed propionic acidemia who presented with progressive neurological deterioration and recurrent episodes of metabolic decompensation with elevated lactate levels, but without hyperammonemia. The child's clinical course and neuroradiological findings increasingly deviated from the known clinical and neuroradiological spectrum of propionic acidemia. A rapid trio exome sequencing identified SLC19A3-related thiamine metabolism dysfunction syndrome 2 as a second genetic disease. The pathomechanisms of both diseases synergize in the impairment of brain energy metabolism, and the associated clinical phenotypes partially overlap, which explains the severe and atypical course of propionic acidemia in the reported case.
Collapse
Affiliation(s)
- Tim Burkhardt
- Medical Faculty, Center for Pediatrics and Adolescent Medicine, Division of Pediatric Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Katharine L. Kastor
- Department of Pediatrics and Adolescent MedicineMarienhaus Klinikum St. Elisabeth NeuwiedNeuwiedGermany
| | - Stine Christ
- Medical Faculty, Center for Pediatrics and Adolescent Medicine, Division of Pediatric Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Thomas Opladen
- Medical Faculty, Center for Pediatrics and Adolescent Medicine, Division of Pediatric Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Christian Staufner
- Medical Faculty, Center for Pediatrics and Adolescent Medicine, Division of Pediatric Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| | - Stefanie Beck‐Wödl
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls University TübingenTübingenGermany
| | - Tobias Haack
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls University TübingenTübingenGermany
| | - Maja Hempel
- Medical Faculty, Institute of Human GeneticsHeidelberg UniversityHeidelbergGermany
| | - Angelika Seitz
- Medical Faculty, Department of NeuroradiologyHeidelberg UniversityHeidelbergGermany
| | - Stefan Kölker
- Medical Faculty, Center for Pediatrics and Adolescent Medicine, Division of Pediatric Neurology and Metabolic MedicineHeidelberg UniversityHeidelbergGermany
| |
Collapse
|
5
|
Richard E, Marchuk H, Álvarez M, He W, Chen X, Desviat LR, Zhang GF. Metabolic flux analysis in hiPSC-CMs reveals insights into cardiac dysfunction in propionic acidemia Eva Richard. RESEARCH SQUARE 2025:rs.3.rs-5874705. [PMID: 39975893 PMCID: PMC11838748 DOI: 10.21203/rs.3.rs-5874705/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Propionic acidemia is an inborn error of metabolism caused by mutations in either the PCCA or PCCB genes. Patients with propionic acidemia experience a range of complications, including life-threatening cardiac dysfunctions. However, the pathological mechanisms underlying propionic acidemia-associated cardiac diseases remain largely unknown. To gain insights into the metabolic alterations in propionic acidemia, we studied human induced pluripotent stem cell-derived cardiomyocytes generated from a patient with propionic acidemia with two pathogenic PCCA mutations (p.Cys616_Val633del and p.Gly477Glufs9*) and from a healthy individual. Using stable isotope-based metabolic flux analysis, we confirmed that the PCCA mutations lead to impaired propionyl-CoA carboxylase activity in human induced pluripotent stem cell-derived cardiomyocytes. In addition to being converted to propionylcarnitine, the accumulated propionyl-CoA can also be hydrolyzed to propionate and exported out of the cell, serving as a secondary "pressure valve" to regulate cellular propionyl-CoA levels. Interestingly, the deficiency of propionyl-CoA carboxylase was found to shift fuel metabolism from fatty acid oxidation to increased glucose metabolism human in induced pluripotent stem cell-derived cardiomyocytes from patients with propionic acidemia. This metabolic switch is less energy-efficient and may contribute to the development of chronic cardiac dysfunction in patients with propionic acidemia.
Collapse
Affiliation(s)
- Eva Richard
- Universidad Autónoma de Madrid: Universidad Autonoma de Madrid
| | | | - Mar Álvarez
- Universidad Autónoma de Madrid: Universidad Autonoma de Madrid
| | | | | | | | | |
Collapse
|
6
|
Luo Q, Liu C, Lin L, Liu X, Chen H. Caregiver's experiences with a mobile-based educational program and its impact on dietary treatment compliance of children with methylmalonic acidemia: an online survey. Orphanet J Rare Dis 2025; 20:31. [PMID: 39828698 PMCID: PMC11742810 DOI: 10.1186/s13023-025-03528-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Compliance to highly restrictive diets is critical for children with Methylmalonic Acidemia (MMA), and their caregivers play a prominent role in children's dietary treatment from early childhood through to adulthood. Despite lots of efforts by the multidisciplinary medical team to ensure the smooth implementation of dietary treatment, restricting dietary protein remains particularly challenging for children with MMA. This study aimed to assess dietary treatment compliance in children with MMA and evaluate the impact of WeChat-based parent education on compliance. METHODS A sample of 151 caregiver-child dyads was obtained through online recruitment using convenience sampling from February to March 2023. At least one month following the enrollment of MMA caregivers in the WeChat public account "Methylmalonic Acidemia Diet Manager", structured questionnaires were distributed to them through the electronic platform "Questionnaire Star" in collaboration with the Chinese National Alliance of Rare Diseases. Subsequently, the collected data was analyzed using quantitative methods. RESULTS Children with MMA aged over 5 years were more likely to present a lower level of dietary treatment compliance compared to those under 1 year old. Besides, the levels of children's dietary treatment compliance were higher when their caregivers had higher levels of satisfaction and benefit from using the public account. CONCLUSION Our findings highlighted the significance of age-related challenges in dietary treatment compliance among children with MMA and the promising impact of utilizing WeChat public accounts as a supportive education tool.
Collapse
Affiliation(s)
- Qing Luo
- School of Nursing, Guangzhou Medical University, Guangzhou, 510182, Guangdong, China
| | - Chunqin Liu
- School of Nursing, Guangzhou Medical University, Guangzhou, 510182, Guangdong, China
| | - Lizhou Lin
- Operating Theatre, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuehua Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Huifang Chen
- School of Nursing, Guangzhou Medical University, Guangzhou, 510182, Guangdong, China.
| |
Collapse
|
7
|
Uslu A, Çekmen N, Torgay A, Haberal M. Perioperative management in pediatric domino liver transplantation for metabolic disorders: A narrative review. Paediatr Anaesth 2024; 34:1107-1118. [PMID: 38980227 DOI: 10.1111/pan.14967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
Domino liver transplantation and domino-auxiliary partial orthotopic liver transplantation are emerging techniques that can expand the liver donor pool and provide hope for children with liver disease. The innovative technique of domino liver transplantation has emerged as a pioneering strategy, capitalizing on structurally preserved livers from donors exhibiting single enzymatic defects within a morphologically normal context, effectively broadening the donor pool. Concurrently, the increasingly prevalent domino-auxiliary partial orthotopic liver transplantation method assumes a critical role in bolstering available donor resources. These advanced transplantation methods present a unique opportunity for pediatric patients who, despite having structurally and functionally intact livers and lacking early signs of portal hypertension or extrahepatic involvement, do not attain priority on conventional transplant lists. Utilizing optimal clinical conditions enhances posttransplant outcomes, benefiting patients who would otherwise endure extended waiting periods for traditional transplantation. The perioperative management of children undergoing these procedures is complex and requires careful consideration of some factors, including clinical and metabolic conditions of the specific metabolic disorder, and the need for tailored perioperative management planning. Furthermore, the prudent consideration of de novo disease development in the recipient assumes paramount significance when selecting suitable donors for domino liver transplantation, as it profoundly influences prognosis, mortality, and morbidity. This narrative review of domino liver transplantation will discuss the pathophysiology, clinical evaluation, perioperative management, and prognostic expectations, focusing on perioperative anesthetic considerations for children undergoing domino liver transplantation.
Collapse
Affiliation(s)
- Ahmed Uslu
- Department of Surgical Sciences, Anesthesiology and Intensive Care Medicine, Başkent University, Ankara, Türkiye
| | - Nedim Çekmen
- Department of Surgical Sciences, Anesthesiology and Intensive Care Medicine, Başkent University, Ankara, Türkiye
| | - Adnan Torgay
- Department of Surgical Sciences, Anesthesiology and Intensive Care Medicine, Başkent University, Ankara, Türkiye
| | - Mehmet Haberal
- Department of Surgical Sciences, Organ and Tissue Transplantation Center, Başkent University, Ankara, Türkiye
| |
Collapse
|
8
|
Zielonka M, Kölker S, Garbade SF, Gleich F, Nagamani SCS, Gropman AL, Druck AC, Ramdhouni N, Göde L, Hoffmann GF, Posset R. Severity-adjusted evaluation of initial dialysis on short-term health outcomes in urea cycle disorders. Mol Genet Metab 2024; 143:108566. [PMID: 39299137 DOI: 10.1016/j.ymgme.2024.108566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE In individuals with urea cycle disorders (UCDs) and neonatal disease onset, extracorporeal detoxification by continuous kidney replacement therapy is considered the therapeutic method of choice in addition to metabolic emergency treatment to resolve hyperammonemic decompensation. However, the indications for the initiation of dialysis are heterogeneously implemented transnationally, thereby hampering our understanding of (optimal) short-term health outcomes. METHODS We performed a retrospective comparative analysis evaluating the therapeutic effects of initial dialysis on survival as well as neurocognitive outcome parameters in individuals with UCDs in comparison to a severity-adjusted non-dialyzed control cohort. Overall, 108 individuals with a severe phenotype of male ornithine transcarbamylase deficiency (mOTC-D), citrullinemia type 1 (CTLN1) and argininosuccinic aciduria (ASA) were investigated by stratification based on a recently established and validated genotype-specific disease prediction model. RESULTS Mortality is associated with the height of initial peak plasma ammonium concentration, but appears to be independent from treatment with initial dialysis in mOTC-D. However, improved survival after initial dialysis was observed in CTLN1, while there was a trend towards improved survival in ASA. In survivors, annual frequency of (subsequent) metabolic decompensations did not differ between the dialyzed and non-dialyzed cohorts. Moreover, treatment with initial dialysis was not associated with improved neurocognitive outcomes. INTERPRETATION The present severity-adjusted comparative analysis reveals that general practice of initial dialysis is neither associated with improved survival in individuals with mOTC-D nor does it differ with regard to the neurocognitive outcome for the investigated UCD subtypes. However, initial dialysis might potentially prove beneficial for survival in CTLN1 and ASA. CLINICAL TRIAL REGISTRATION The UCDC database is recorded at the US National Library of Medicine (https://clinicaltrials.gov).
Collapse
Affiliation(s)
- Matthias Zielonka
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Stefan Kölker
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Sven F Garbade
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Florian Gleich
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Andrea L Gropman
- Children's National Health System and The George Washington School of Medicine, Washington, DC, USA
| | - Ann-Catrin Druck
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Nesrine Ramdhouni
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Laura Göde
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Georg F Hoffmann
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Roland Posset
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| |
Collapse
|
9
|
Liu Y, Ma X, Kang L, Jin Y, Li M, Song J, Li H, Cao Y, Yang Y. The utility of methylmalonic acid, methylcitrate acid, and homocysteine in dried blood spots for therapeutic monitoring of three inherited metabolic diseases. Front Nutr 2024; 11:1414681. [PMID: 38966413 PMCID: PMC11222987 DOI: 10.3389/fnut.2024.1414681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024] Open
Abstract
Backgroud Routine metabolic assessments for methylmalonic acidemia (MMA), propionic acidemia (PA), and homocysteinemia involve detecting metabolites in dried blood spots (DBS) and analyzing specific biomarkers in serum and urine. This study aimed to establish a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the simultaneous detection of three specific biomarkers (methylmalonic acid, methylcitric acid, and homocysteine) in DBS, as well as to appraise the applicability of these three DBS metabolites in monitoring patients with MMA, PA, and homocysteinemia during follow-up. Methods A total of 140 healthy controls and 228 participants were enrolled, including 205 patients with MMA, 17 patients with PA, and 6 patients with homocysteinemia. Clinical data and DBS samples were collected during follow-up visits. Results The reference ranges (25th-95th percentile) for DBS methylmalonic acid, methylcitric acid, and homocysteine were estimated as 0.04-1.02 μmol/L, 0.02-0.27 μmol/L and 1.05-8.22 μmol/L, respectively. Following treatment, some patients achieved normal metabolite concentrations, but the majority still exhibited characteristic biochemical patterns. The concentrations of methylmalonic acid, methylcitric acid, and homocysteine in DBS showed positive correlations with urine methylmalonic acid (r = 0.849, p < 0.001), urine methylcitric acid (r = 0.693, p < 0.001), and serum homocysteine (r = 0.721, p < 0.001) concentrations, respectively. Additionally, higher levels of DBS methylmalonic acid and methylcitric acid may be associated with increased cumulative complication scores. Conclusion The LC-MS/MS method established in this study reliably detects methylmalonic acid, methylcitric acid, and homocysteine in DBS. These three DBS metabolites can be valuable for monitoring patients with MMA, PA, and homocysteinemia during follow-up. Further investigation is required to determine the significance of these DBS biomarkers in assessing disease burden over time.
Collapse
Affiliation(s)
- Yi Liu
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Xue Ma
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Lulu Kang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Mengqiu Li
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jinqing Song
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Haixia Li
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China
| | - Yongtong Cao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Yanling Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
10
|
López-Mejía LA, Vela-Amieva M, Guillén-López S, Mancera-Hernández D, Ibarra-González I, Medina-Torres EA, Espinosa-Padilla SE, Fernández-Lainez C. Hypogammaglobulinemia Class G Is Present in Compensated and Decompensated Patients with Propionate Defects, Independent of Their Nutritional Status. Nutrients 2024; 16:1775. [PMID: 38892708 PMCID: PMC11174734 DOI: 10.3390/nu16111775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Propionate defects (PDs) mainly include methylmalonic (MMA) and propionic acidemia (PA) defects. Lifelong PD patients progress from the compensated to the decompensated stages, the latter of which are characterized by life-threatening acidemia and hyperammonemia crises. PD patients can suffer immunocompromise, especially during the decompensation stage. There is a significant gap in the research regarding the humoral immune response in PD patients. Here, we analyzed serum immunoglobulin concentrations and hemograms across compensated and decompensated stages in PD patients. Nutritional status and crisis triggers of decompensation were also explored. Twenty patients were studied, and 25 decompensation events (DE) and 8 compensation events (CE) were recorded. Compared with those in the CE group, the IgG levels in the DE group (513.4 ± 244.5 mg/dL) were significantly lower than those in the CE group (860.8 ± 456.5 mg/dL) (p < 0.0087). The mean hemoglobin concentration was significantly lower in the DE group (11.8 g/dL) than in the CE group (13.4 g/dL) (p < 0.05). The most frequent (48%) possible decompensation trigger factor was infection. Most of the events were registered in eutrophic patients (87.9%), despite which 65.2% and 50% of patients who experienced decompensated and compensated events, respectively, presented with hypogammaglobulinemia G. These findings provide evidence of the immunodeficiency of PD patients, independent of their nutritional status. We suggest that PD patients be managed as immunocompromised independently of their nutritional status or metabolic state (compensated or decompensated).
Collapse
Affiliation(s)
- Lizbeth Alejandra López-Mejía
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Marcela Vela-Amieva
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Sara Guillén-López
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Daniela Mancera-Hernández
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
- Servicio Social Facultad de Medicina, Benemérita Universidad de Puebla, Puebla Pue 72570, Mexico
| | - Isabel Ibarra-González
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| | | | - Sara Elva Espinosa-Padilla
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Cynthia Fernández-Lainez
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| |
Collapse
|
11
|
Bretos-Azcona PE, Wallace M, Jootun M, Jin G, Agirrezabal I, Szende A. An Early Cost-Utility Model of mRNA-Based Therapies for the Treatment of Methylmalonic and Propionic Acidemia in the United Kingdom. Clin Drug Investig 2024; 44:399-412. [PMID: 38796677 DOI: 10.1007/s40261-024-01363-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND AND OBJECTIVE Novel messenger RNA (mRNA)-based therapies, currently in development, are emerging as a promising potential treatment modality for a broad range of life-threatening and life-limiting inherited liver diseases, including methylmalonic acidemia (MMA) and propionic acidemia (PA). However, owing in part to their complexity, they are likely to come at considerable financial cost to healthcare systems. The objective of this research was to synthesize available evidence on the costs and clinical consequences associated with MMA and PA for the purpose of exploratory economic evaluation of novel mRNA-based therapies using an early cost-utility model from the United Kingdom payer perspective. METHODS A Markov model was constructed to simulate the costs and outcomes associated with novel mRNA therapies, compared with a combination of dietary management and organ transplantation (standard of care) among hypothetical cohorts of new-born patients with MMA and PA. Key model drivers were identified, and a price threshold analysis was performed to estimate value-based price ranges for future mRNA therapies given willingness-to-pay thresholds for orphan diseases. RESULTS mRNA therapy was associated with an additional 5.7 and 1.3 quality-adjusted life-years (QALYs) gained per patient lifetime among patients with MMA and PA, respectively. Key drivers of cost-effectiveness were relative improvement in utility among patients who receive mRNA-based therapy and transplantation, and the cost of mRNA therapy. Assuming a willingness to pay range of £100,000-£300,000 per QALY gained, the model demonstrated mRNA therapy to be cost-effective in MMA and PA at an annual treatment cost of £70,452-£94,575 and £31,313-£36,695, respectively. CONCLUSIONS Despite the lack of a strong evidence base in MMA and PA, this model provides a useful tool to estimate the cost-effectiveness, and inform value-based pricing, of new mRNA-based therapies. Our analyses also identified areas for research that will have the greatest value in reducing uncertainty in future health economic evaluations of such treatments.
Collapse
Affiliation(s)
- Pablo E Bretos-Azcona
- Product Development and Market Access Consulting, Fortrea, 4 Maguire Street, London, SE1 2NQ, UK
| | - Matthew Wallace
- Product Development and Market Access Consulting, Fortrea, 4 Maguire Street, London, SE1 2NQ, UK
| | - Murvin Jootun
- Product Development and Market Access Consulting, Fortrea, 4 Maguire Street, London, SE1 2NQ, UK
| | - Guanyi Jin
- Product Development and Market Access Consulting, Fortrea, 4 Maguire Street, London, SE1 2NQ, UK
| | - Ion Agirrezabal
- Product Development and Market Access Consulting, Fortrea, 4 Maguire Street, London, SE1 2NQ, UK
| | - Agota Szende
- Product Development and Market Access Consulting, Fortrea, 4 Maguire Street, London, SE1 2NQ, UK.
| |
Collapse
|
12
|
Elola Pastor AI, Prieto García B, Díaz Martín JJ. Evaluation of the first 5 years of a glutaric aciduria type I neonatal screening programme in Asturias. An Pediatr (Barc) 2024; 100:318-324. [PMID: 38714461 DOI: 10.1016/j.anpede.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/29/2024] [Indexed: 05/10/2024] Open
Abstract
INTRODUCTION . Neonatal screening of glutaric aciduria type 1 (GA-1) has brought radical changes in the course and outcomes of this disease. This study analyses the outcomes of the first 5 years (2015-2019) of the AGA1 neonatal screening programme in our autonomous community. MATERIAL . We conducted an observational, descriptive and retrospective study. All neonates born between January 1, 2015 and December 31, 2019 that participated in the neonatal screening programme were included in the study. The glutarylcarnitine (C5DC) concentration in dry blood spot samples was measured by means of tandem mass spectrometry applying a cut-off point of 0.25 µmol/L. RESULTS . A total of 30 120 newborns underwent screening. We found differences in the C5DC concentration based on gestational age, type of feeding and hours of life at sample collection. These differences were not relevant for screening purposes. There were no differences between neonates with weights smaller and greater than 1500 g. Screening identified 2 affected patients and there were 3 false positives. There were no false negatives. The diagnosis was confirmed by genetic testing. Patients have been in treatment since diagnosis and have not developed encephalopathic crises in the first 4 years of life. CONCLUSIONS . Screening allowed early diagnosis of two cases of GA-1 in the first 5 years since its introduction in our autonomous community. Although there were differences in C5DC levels based on gestational age, type of feeding and hours of life at blood extraction, they were not relevant for screening.
Collapse
Affiliation(s)
| | - Belén Prieto García
- Unidad de Bioquímica Clínica, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Juan José Díaz Martín
- Sección de Gastroenterología y Nutrición Pediátrica, Hospital Universitario Central de Asturias, Universidad de Oviedo, Oviedo, Asturias, Spain
| |
Collapse
|
13
|
Shchelochkov OA, Farmer CA, Chlebowski C, Adedipe D, Ferry S, Manoli I, Pass A, McCoy S, Van Ryzin C, Sloan J, Thurm A, Venditti CP. Intellectual disability and autism in propionic acidemia: a biomarker-behavioral investigation implicating dysregulated mitochondrial biology. Mol Psychiatry 2024; 29:974-981. [PMID: 38200289 PMCID: PMC11176071 DOI: 10.1038/s41380-023-02385-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/13/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024]
Abstract
Propionic acidemia (PA) is an autosomal recessive condition (OMIM #606054), wherein pathogenic variants in PCCA and PCCB impair the activity of propionyl-CoA carboxylase. PA is associated with neurodevelopmental disorders, including intellectual disability (ID) and autism spectrum disorder (ASD); however, the correlates and mechanisms of these outcomes remain unknown. Using data from a subset of participants with PA enrolled in a dedicated natural history study (n = 33), we explored associations between neurodevelopmental phenotypes and laboratory parameters. Twenty (61%) participants received an ID diagnosis, and 12 of the 31 (39%) who were fully evaluated received the diagnosis of ASD. A diagnosis of ID, lower full-scale IQ (sample mean = 65 ± 26), and lower adaptive behavior composite scores (sample mean = 67 ± 23) were associated with several biomarkers. Higher concentrations of plasma propionylcarnitine, plasma total 2-methylcitrate, serum erythropoietin, and mitochondrial biomarkers plasma FGF21 and GDF15 were associated with a more severe ID profile. Reduced 1-13C-propionate oxidative capacity and decreased levels of plasma and urinary glutamine were also associated with a more severe ID profile. Only two parameters, increased serum erythropoietin and decreased plasma glutamine, were associated with ASD. Plasma glycine, one of the defining features of PA, was not meaningfully associated with either ID or ASD. Thus, while both ID and ASD were commonly observed in our PA cohort, only ID was robustly associated with metabolic parameters. Our results suggest that disease severity and associated mitochondrial dysfunction may play a role in CNS complications of PA and identify potential biomarkers and candidate surrogate endpoints.
Collapse
Affiliation(s)
- Oleg A Shchelochkov
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Cristan A Farmer
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Colby Chlebowski
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dee Adedipe
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Susan Ferry
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Irini Manoli
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alexandra Pass
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Samantha McCoy
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Carol Van Ryzin
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jennifer Sloan
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Charles P Venditti
- Organic Acid Research Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
14
|
Posset R, Garbade SF, Gleich F, Nagamani SCS, Gropman AL, Epp F, Ramdhouni N, Druck AC, Hoffmann GF, Kölker S, Zielonka M. Impact of supplementation with L-citrulline/arginine after liver transplantation in individuals with Urea Cycle Disorders. Mol Genet Metab 2024; 141:108112. [PMID: 38301530 DOI: 10.1016/j.ymgme.2023.108112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 02/03/2024]
Abstract
OBJECTIVE Liver transplantation (LTx) is an intervention when medical management is not sufficiently preventing individuals with urea cycle disorders (UCDs) from the occurrence of hyperammonemic events. Supplementation with L-citrulline/arginine is regularly performed prior to LTx to support ureagenesis and is often continued after the intervention. However, systematic studies assessing the impact of long-term L-citrulline/arginine supplementation in individuals who have undergone LTx is lacking to date. METHODS Using longitudinal data collected systematically, a comparative analysis was carried out by studying the effects of long-term L-citrulline/arginine supplementation vs. no supplementation on health-related outcome parameters (i.e., anthropometric, neurological, and cognitive outcomes) in individuals with UCDs who have undergone LTx. Altogether, 52 individuals with male ornithine transcarbamylase deficiency, citrullinemia type 1 and argininosuccinic aciduria and a pre-transplant "severe" disease course who have undergone LTx were investigated by using recently established and validated genotype-specific in vitro enzyme activities. RESULTS Long-term supplementation of individuals with L-citrulline/arginine who have undergone LTx (n = 16) does neither appear to alter anthropometric nor neurocognitive endpoints when compared to their severity-adjusted counterparts that were not supplemented (n = 36) after LTx with mean observation periods between four to five years. Moreover, supplementation with L-citrulline/arginine was not associated with an increase of disease-specific plasma arithmetic mean values for the respective amino acids when compared to the non-supplemented control cohort. CONCLUSION Although supplementation with L-citrulline/arginine is often continued after LTx, this pilot study does neither identify altered long-term anthropometric or neurocognitive health-related outcomes nor does it find an adequate biochemical response as reflected by the unaltered plasma arithmetic mean values for L-citrulline or L-arginine. Further prospective analyses in larger samples and even longer observation periods will provide more insight into the usefulness of long-term supplementation with L-citrulline/arginine for individuals with UCDs who have undergone LTx.
Collapse
Affiliation(s)
- Roland Posset
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Sven F Garbade
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Florian Gleich
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Andrea L Gropman
- Children's National Health System and The George Washington School of Medicine, Washington, DC, USA
| | - Friederike Epp
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Nesrine Ramdhouni
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Ann-Catrin Druck
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Georg F Hoffmann
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Stefan Kölker
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Matthias Zielonka
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| |
Collapse
|
15
|
Busiah K, Roda C, Crosnier AS, Brassier A, Servais A, Wicker C, Dubois S, Assoun M, Belloche C, Ottolenghi C, Pontoizeau C, Souberbielle JC, Piketty ML, Perin L, Le Bouc Y, Arnoux JB, Netchine I, Imbard A, de Lonlay P. Pubertal origin of growth retardation in inborn errors of protein metabolism: A longitudinal cohort study. Mol Genet Metab 2024; 141:108123. [PMID: 38219674 DOI: 10.1016/j.ymgme.2023.108123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/16/2024]
Abstract
OBJECTIVES Inherited amino-acid metabolism disorders (IAAMDs) require lifelong protein-restricted diet. We aimed to investigate: 1/ whether IAAMDs was associated with growth, pubertal, bone mineral apparent density (BMAD) or body composition impairments; 2/ associations linking height, amino-acid mixture (AAM), plasma amino-acids and IGF1 concentrations. DESIGN Retrospective longitudinal study of 213 patients with neonatal-onset urea cycle disorders (UCD,n = 77), organic aciduria (OA,n = 89), maple syrup urine disease (MSUD,n = 34), or tyrosinaemia type 1 (n = 13). METHODS We collected growth parameters, pubertal status, BMAD, body composition, protein-intake, and IGF1 throughout growth. RESULTS Overall final height (n = 69) was below target height (TH): -0.9(1.4) vs. -0.1(0.9) SD, p < 0.001. Final height was ≤ TH-2SD in 12 (21%) patients. Height ≤ - 2SD was more frequent during puberty than during early-infancy and pre-puberty: 23.5% vs. 6.9%, p = 0.002; and vs. 10.7%, p < 0.001. Pubertal delay was frequent (26.7%). Height (SD) was positively associated with isoleucine concentration: β, 0.008; 95%CI, 0.003 to 0.012; p = 0.001. In the pubertal subgroup, height (SD) was lower in patients with vs. without AAM supplementation: -1.22 (1.40) vs. -0.63 (1.46) (p = 0.02). In OA, height and median (IQR) isoleucine and valine concentrations(μmol/L) during puberty were lower in patients with vs. without AAM supplementation: -1.75 (1.30) vs. -0.33 (1.55) SD, p < 0.001; and 40 (23) vs. 60 (25) (p = 0.02) and 138 (92) vs. 191 (63) (p = 0.01), respectively. No correlation was found with IGF1. Lean-mass index was lower than fat-mass index: -2.03 (1.15) vs. -0.44 (0.89), p < 0.001. CONCLUSIONS In IAAMDs, growth retardation worsened during puberty which was delayed in all disease subgroups. Height seems linked to the disease, AAM composition and lower isoleucine concentration, independently of the GH-IGF1 pathway. We recommend close monitoring of diet during puberty.
Collapse
Affiliation(s)
- Kanetee Busiah
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France; Paediatric endocrinology, diabetology and obesity unit, Women-Mothers-Children Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.
| | - Célina Roda
- Université Paris Cité, HERA Team, CRESS, INSERM, INRAE, F-75004 Paris, France
| | - Anne-Sophie Crosnier
- Endocrine function testing department, Assistance Publique-Hôpitaux de Paris, Trousseau University Hospital, Paris, France
| | - Anaïs Brassier
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France
| | - Aude Servais
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France
| | - Camille Wicker
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France; Pediatric Inherited Metabolic Diseases department, University Hospital of Strasbourg- Hautepierre, Strasbourg, France
| | - Sandrine Dubois
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France
| | - Murielle Assoun
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France
| | - Claire Belloche
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France
| | - Chris Ottolenghi
- Metabolic biochemistry, Necker Enfants-Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Imagine Institute, Filière G2M, MetabERN, Medical School, Université Paris Cité, Paris, France
| | - Clément Pontoizeau
- Metabolic biochemistry, Necker Enfants-Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Imagine Institute, Filière G2M, MetabERN, Medical School, Université Paris Cité, Paris, France
| | - Jean-Claude Souberbielle
- Hormonology laboratory, Physiology department, Necker-Enfants Malades Teaching Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marie-Liesse Piketty
- Hormonology laboratory, Physiology department, Necker-Enfants Malades Teaching Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Laurence Perin
- Endocrine function testing department, Assistance Publique-Hôpitaux de Paris, Trousseau University Hospital, Paris, France
| | - Yves Le Bouc
- Endocrine function testing department, Assistance Publique-Hôpitaux de Paris, Trousseau University Hospital, Paris, France; Sorbonne University, INSERM, Saint Antoine research centre, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Baptiste Arnoux
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France
| | - Irène Netchine
- Endocrine function testing department, Assistance Publique-Hôpitaux de Paris, Trousseau University Hospital, Paris, France; Sorbonne University, INSERM, Saint Antoine research centre, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Apolline Imbard
- Metabolic biochemistry, Necker Enfants-Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Imagine Institute, Filière G2M, MetabERN, Medical School, Université Paris Cité, Paris, France
| | - Pascale de Lonlay
- Inherited Metabolic Diseases Reference Center, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Filière G2M, MetabERN, Université Paris Cité, Paris, France; INSERM U1151, Necker-Enfants Malades Institute (INEM), Paris, France
| |
Collapse
|
16
|
Tejero J, Lazure F, Gomes AP. Methylmalonic acid in aging and disease. Trends Endocrinol Metab 2024; 35:188-200. [PMID: 38030482 PMCID: PMC10939937 DOI: 10.1016/j.tem.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023]
Abstract
Metabolic byproducts have conventionally been disregarded as waste products without functions. In this opinion article, we bring to light the multifaceted role of methylmalonic acid (MMA), a byproduct of the propionate metabolism pathway mostly commonly known as a clinical biomarker of vitamin B12 deficiency. MMA is normally present at low levels in the body, but increased levels can come from different sources, such as vitamin B12 deficiency, genetic mutations in enzymes related to the propionate pathway, the gut microbiota, and aggressive cancers. Here, we describe the diverse metabolic and signaling functions of MMA and discuss the consequences of increased MMA levels, such as during the aging process, for several age-related human pathologies.
Collapse
Affiliation(s)
- Joanne Tejero
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Felicia Lazure
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ana P Gomes
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
17
|
Manoli I, Sysol JR, Head PE, Epping MW, Gavrilova O, Crocker MK, Sloan JL, Koutsoukos SA, Wang C, Ktena YP, Mendelson S, Pass AR, Zerfas PM, Hoffmann V, Vernon HJ, Fletcher LA, Reynolds JC, Tsokos MG, Stratakis CA, Voss SD, Chen KY, Brown RJ, Hamosh A, Berry GT, Chen XS, Yanovski JA, Venditti CP. Lipodystrophy in methylmalonic acidemia associated with elevated FGF21 and abnormal methylmalonylation. JCI Insight 2024; 9:e174097. [PMID: 38271099 DOI: 10.1172/jci.insight.174097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
A distinct adipose tissue distribution pattern was observed in patients with methylmalonyl-CoA mutase deficiency, an inborn error of branched-chain amino acid (BCAA) metabolism, characterized by centripetal obesity with proximal upper and lower extremity fat deposition and paucity of visceral fat, that resembles familial multiple lipomatosis syndrome. To explore brown and white fat physiology in methylmalonic acidemia (MMA), body composition, adipokines, and inflammatory markers were assessed in 46 patients with MMA and 99 matched controls. Fibroblast growth factor 21 levels were associated with acyl-CoA accretion, aberrant methylmalonylation in adipose tissue, and an attenuated inflammatory cytokine profile. In parallel, brown and white fat were examined in a liver-specific transgenic MMA mouse model (Mmut-/- TgINS-Alb-Mmut). The MMA mice exhibited abnormal nonshivering thermogenesis with whitened brown fat and had an ineffective transcriptional response to cold stress. Treatment of the MMA mice with bezafibrates led to clinical improvement with beiging of subcutaneous fat depots, which resembled the distribution seen in the patients. These studies defined what we believe to be a novel lipodystrophy phenotype in patients with defects in the terminal steps of BCAA oxidation and demonstrated that beiging of subcutaneous adipose tissue in MMA could readily be induced with small molecules.
Collapse
Affiliation(s)
- Irini Manoli
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Justin R Sysol
- Metabolic Medicine Branch, National Human Genome Research Institute
| | | | | | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Melissa K Crocker
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | - Jennifer L Sloan
- Metabolic Medicine Branch, National Human Genome Research Institute
| | | | - Cindy Wang
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Yiouli P Ktena
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Sophia Mendelson
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | - Alexandra R Pass
- Metabolic Medicine Branch, National Human Genome Research Institute
| | - Patricia M Zerfas
- Office of Research Services, Division of Veterinary Resources, NIH, Bethesda, Maryland, USA
| | - Victoria Hoffmann
- Office of Research Services, Division of Veterinary Resources, NIH, Bethesda, Maryland, USA
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Laura A Fletcher
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | | | - Maria G Tsokos
- Ultrastructural Pathology Section, Center for Cancer Research; and
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Stephan D Voss
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kong Y Chen
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Rebecca J Brown
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Ada Hamosh
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gerard T Berry
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoyuan Shawn Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, Maryland, USA
| | - Jack A Yanovski
- Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
| | | |
Collapse
|
18
|
Gurung S, Timmermand OV, Perocheau D, Gil-Martinez AL, Minnion M, Touramanidou L, Fang S, Messina M, Khalil Y, Spiewak J, Barber AR, Edwards RS, Pinto PL, Finn PF, Cavedon A, Siddiqui S, Rice L, Martini PGV, Ridout D, Heywood W, Hargreaves I, Heales S, Mills PB, Waddington SN, Gissen P, Eaton S, Ryten M, Feelisch M, Frassetto A, Witney TH, Baruteau J. mRNA therapy corrects defective glutathione metabolism and restores ureagenesis in preclinical argininosuccinic aciduria. Sci Transl Med 2024; 16:eadh1334. [PMID: 38198573 PMCID: PMC7615535 DOI: 10.1126/scitranslmed.adh1334] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 10/06/2023] [Indexed: 01/12/2024]
Abstract
The urea cycle enzyme argininosuccinate lyase (ASL) enables the clearance of neurotoxic ammonia and the biosynthesis of arginine. Patients with ASL deficiency present with argininosuccinic aciduria, an inherited metabolic disease with hyperammonemia and a systemic phenotype coinciding with neurocognitive impairment and chronic liver disease. Here, we describe the dysregulation of glutathione biosynthesis and upstream cysteine utilization in ASL-deficient patients and mice using targeted metabolomics and in vivo positron emission tomography (PET) imaging using (S)-4-(3-18F-fluoropropyl)-l-glutamate ([18F]FSPG). Up-regulation of cysteine metabolism contrasted with glutathione depletion and down-regulated antioxidant pathways. To assess hepatic glutathione dysregulation and liver disease, we present [18F]FSPG PET as a noninvasive diagnostic tool to monitor therapeutic response in argininosuccinic aciduria. Human hASL mRNA encapsulated in lipid nanoparticles improved glutathione metabolism and chronic liver disease. In addition, hASL mRNA therapy corrected and rescued the neonatal and adult Asl-deficient mouse phenotypes, respectively, enhancing ureagenesis. These findings provide mechanistic insights in liver glutathione metabolism and support clinical translation of mRNA therapy for argininosuccinic aciduria.
Collapse
Affiliation(s)
- Sonam Gurung
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | | | - Dany Perocheau
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ana Luisa Gil-Martinez
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Magdalena Minnion
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Loukia Touramanidou
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sherry Fang
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Martina Messina
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Youssef Khalil
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Justyna Spiewak
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Abigail R Barber
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Richard S Edwards
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Patricia Lipari Pinto
- Santa Maria's Hospital, Lisbon North University Hospital Center, 1649-028 Lisbon, Portugal
| | | | | | | | - Lisa Rice
- Moderna Inc., Cambridge, MA 02139, USA
| | | | - Deborah Ridout
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Wendy Heywood
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ian Hargreaves
- Pharmacy and Biomolecular Sciences, Liverpool John Moore University, Liverpool L3 5UG, UK
| | - Simon Heales
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Philippa B Mills
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Simon N Waddington
- EGA Institute for Women's Health, University College London, London WC1E 6HX, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of Witswatersrand, Braamfontein, 2000 Johannesburg, South Africa
| | - Paul Gissen
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Mina Ryten
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | | | - Timothy H Witney
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Julien Baruteau
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK
| |
Collapse
|
19
|
Moritz L, Schumann A, Pohl M, Köttgen A, Hannibal L, Spiekerkoetter U. A systematic review of metabolomic findings in adult and pediatric renal disease. Clin Biochem 2024; 123:110703. [PMID: 38097032 DOI: 10.1016/j.clinbiochem.2023.110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023]
Abstract
Chronic kidney disease (CKD) affects over 0.5 billion people worldwide across their lifetimes. Despite a growingly ageing world population, an increase in all-age prevalence of kidney disease persists. Adult-onset forms of kidney disease often result from lifestyle-modifiable metabolic illnesses such as type 2 diabetes. Pediatric and adolescent forms of renal disease are primarily caused by morphological abnormalities of the kidney, as well as immunological, infectious and inherited metabolic disorders. Alterations in energy metabolism are observed in CKD of varying causes, albeit the molecular mechanisms underlying pathology are unclear. A systematic indexing of metabolites identified in plasma and urine of patients with kidney disease alongside disease enrichment analysis uncovered inborn errors of metabolism as a framework that links features of adult and pediatric kidney disease. The relationship of genetics and metabolism in kidney disease could be classified into three distinct landscapes: (i) Normal genotypes that develop renal damage because of lifestyle and / or comorbidities; (ii) Heterozygous genetic variants and polymorphisms that result in unique metabotypes that may predispose to the development of kidney disease via synergistic heterozygosity, and (iii) Homozygous genetic variants that cause renal impairment by perturbing metabolism, as found in children with monogenic inborn errors of metabolism. Interest in the identification of early biomarkers of onset and progression of CKD has grown steadily in the last years, though it has not translated into clinical routine yet. This systematic review indexes findings of differential concentration of metabolites and energy pathway dysregulation in kidney disease and appraises their potential use as biomarkers.
Collapse
Affiliation(s)
- Lennart Moritz
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Anke Schumann
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Martin Pohl
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Luciana Hannibal
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany.
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
20
|
Seidl MJ, Scharre S, Posset R, Druck AC, Epp F, Okun JG, Dimitrov B, Hoffmann GF, Kölker S, Zielonka M. ASS1 deficiency is associated with impaired neuronal differentiation in zebrafish larvae. Mol Genet Metab 2024; 141:108097. [PMID: 38113552 DOI: 10.1016/j.ymgme.2023.108097] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Citrullinemia type 1 (CTLN1) is a rare autosomal recessive urea cycle disorder caused by deficiency of the cytosolic enzyme argininosuccinate synthetase 1 (ASS1) due to pathogenic variants in the ASS1 gene located on chromosome 9q34.11. Even though hyperammenomia is considered the major pathomechanistic factor for neurological impairment and cognitive dysfunction, a relevant subset of individuals presents with a neurodegenerative course in the absence of hyperammonemic decompensations. Here we show, that ASS1 deficiency induced by antisense-mediated knockdown of the zebrafish ASS1 homologue is associated with defective neuronal differentiation ultimately causing neuronal cell loss and consecutively decreased brain size in zebrafish larvae in vivo. Whereas ASS1-deficient zebrafish larvae are characterized by markedly elevated concentrations of citrulline - the biochemical hallmark of CTLN1, accumulation of L-citrulline, hyperammonemia or therewith associated secondary metabolic alterations did not account for the observed phenotype. Intriguingly, coinjection of the human ASS1 mRNA not only normalized citrulline concentration but also reversed the morphological cerebral phenotype and restored brain size, confirming conserved functional properties of ASS1 across species. The results of the present study imply a novel, potentially non-enzymatic (moonlighting) function of the ASS1 protein in neurodevelopment.
Collapse
Affiliation(s)
- Marie J Seidl
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Svenja Scharre
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Roland Posset
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Ann-Catrin Druck
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Friederike Epp
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen G Okun
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Bianca Dimitrov
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Georg F Hoffmann
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Kölker
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Zielonka
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany; Heidelberg Research Center for Molecular Medicine (HRCMM), Heidelberg, Germany.
| |
Collapse
|
21
|
Posset R, Zielonka M, Gleich F, Garbade SF, Hoffmann GF, Kölker S. The challenge of understanding and predicting phenotypic diversity in urea cycle disorders. J Inherit Metab Dis 2023; 46:1007-1016. [PMID: 37702610 DOI: 10.1002/jimd.12678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023]
Abstract
The Urea Cycle Disorders Consortium (UCDC) and the European registry and network for Intoxication type Metabolic Diseases (E-IMD) are the worldwide largest databases for individuals with urea cycle disorders (UCDs) comprising longitudinal data from more than 1100 individuals with an overall long-term follow-up of approximately 25 years. However, heterogeneity of the clinical phenotype as well as different diagnostic and therapeutic strategies hamper our understanding on the predictors of phenotypic diversity and the impact of disease-immanent and interventional variables (e.g., diagnostic and therapeutic interventions) on the long-term outcome. A new strategy using combined and comparative data analyses helped overcome this challenge. This review presents the mechanisms and relevant principles that are necessary for the identification of meaningful clinical associations by combining data from different data sources, and serves as a blueprint for future analyses of rare disease registries.
Collapse
Affiliation(s)
- Roland Posset
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Zielonka
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
- Heidelberg Research Center for Molecular Medicine (HRCMM), Heidelberg, Germany
| | - Florian Gleich
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sven F Garbade
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Georg F Hoffmann
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Kölker
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
22
|
Schumann A, Schultheiss UT, Ferreira CR, Blau N. Clinical and biochemical footprints of inherited metabolic diseases. XIV. Metabolic kidney diseases. Mol Genet Metab 2023; 140:107683. [PMID: 37597335 PMCID: PMC11747985 DOI: 10.1016/j.ymgme.2023.107683] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
Kidney disease is a global health burden with high morbidity and mortality. Causes of kidney disease are numerous, extending from common disease groups like diabetes and arterial hypertension to rare conditions including inherited metabolic diseases (IMDs). Given its unique anatomy and function, the kidney is a target organ in about 10% of known IMDs, emphasizing the relevant contribution of IMDs to kidney disease. The pattern of injury affects all segments of the nephron including glomerular disease, proximal and distal tubular damage, kidney cyst formation, built-up of nephrocalcinosis and stones as well as severe malformations. We revised and updated the list of known metabolic etiologies associated with kidney involvement and found 190 relevant IMDs. This represents the 14th of a series of educational articles providing a comprehensive and revised list of metabolic differential diagnoses according to system involvement.
Collapse
Affiliation(s)
- Anke Schumann
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany.
| | - Ulla T Schultheiss
- Department of Medicine IV, Nephrology and Primary Care, Faculty of Medicine, and Medical Center, University of Freiburg, Institute of Genetic Epidemiology, Freiburg, Germany.
| | - Carlos R Ferreira
- National Human Genome Research Institute, National Institutes of Health, Bethesda, USA.
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital, Zürich, Switzerland.
| |
Collapse
|
23
|
Baker PR. Recognizing and Managing a Metabolic Crisis. Pediatr Clin North Am 2023; 70:979-993. [PMID: 37704355 DOI: 10.1016/j.pcl.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
In some relatively common inborn errors of metabolism there can be the accumulation of toxic compounds including ammonia and organic acids such as lactate and ketoacids, as well as energy deficits at the cellular level. The clinical presentation is often referred to as a metabolic emergency or crisis. Fasting and illness can result in encephalopathy within hours, and without appropriate recognition and intervention, the outcome may be permanent disability or death. This review outlines easy and readily available means of recognizing and diagnosing a metabolic emergency as well as general guidelines for management. Disease-specific interventions focus on parenteral nutrition to reverse catabolism, toxin removal strategies, and vitamin/nutrition supplementation.
Collapse
Affiliation(s)
- Peter R Baker
- University of Colorado, Children's Hospital Colorado, 13123 East 16th Avenue, Box 300, Aurora, CO 80045, USA.
| |
Collapse
|
24
|
Marchuk H, Wang Y, Ladd ZA, Chen X, Zhang GF. Pathophysiological mechanisms of complications associated with propionic acidemia. Pharmacol Ther 2023; 249:108501. [PMID: 37482098 PMCID: PMC10529999 DOI: 10.1016/j.pharmthera.2023.108501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/06/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Propionic acidemia (PA) is a genetic metabolic disorder caused by mutations in the mitochondrial enzyme, propionyl-CoA carboxylase (PCC), which is responsible for converting propionyl-CoA to methylmalonyl-CoA for further metabolism in the tricarboxylic acid cycle. When this process is disrupted, propionyl-CoA and its metabolites accumulate, leading to a variety of complications including life-threatening cardiac diseases and other metabolic strokes. While the clinical symptoms and diagnosis of PA are well established, the underlying pathophysiological mechanisms of PA-induced diseases are not fully understood. As a result, there are currently few effective therapies for PA beyond dietary restriction. This review focuses on the pathophysiological mechanisms of the various complications associated with PA, drawing on extensive research and clinical reports. Most research suggests that propionyl-CoA and its metabolites can impair mitochondrial energy metabolism and cause cellular damage by inducing oxidative stress. However, direct evidence from in vivo studies is still lacking. Additionally, elevated levels of ammonia can be toxic, although not all PA patients develop hyperammonemia. The discovery of pathophysiological mechanisms underlying various complications associated with PA can aid in the development of more effective therapeutic treatments. The consequences of elevated odd-chain fatty acids in lipid metabolism and potential gene expression changes mediated by histone propionylation also warrant further investigation.
Collapse
Affiliation(s)
- Hannah Marchuk
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA
| | - You Wang
- Jining Key Laboratory of Pharmacology, Jining Medical University, Shandong 272067, China.; School of Basic Medicine, Jining Medical University, Shandong 272067, China
| | - Zachary Alec Ladd
- Surgical Research Lab, Department of Surgery, Cooper University Healthcare and Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Xiaoxin Chen
- Surgical Research Lab, Department of Surgery, Cooper University Healthcare and Cooper Medical School of Rowan University, Camden, NJ 08103, USA; Coriell Institute for Medical Research, Camden, NJ 08103, USA; MD Anderson Cancer Center at Cooper, Camden, NJ 08103, USA.
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, and Metabolism Nutrition, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
25
|
Lucienne M, Gerlini R, Rathkolb B, Calzada-Wack J, Forny P, Wueest S, Kaech A, Traversi F, Forny M, Bürer C, Aguilar-Pimentel A, Irmler M, Beckers J, Sauer S, Kölker S, Dewulf JP, Bommer GT, Hoces D, Gailus-Durner V, Fuchs H, Rozman J, Froese DS, Baumgartner MR, de Angelis MH. Insights into energy balance dysregulation from a mouse model of methylmalonic aciduria. Hum Mol Genet 2023; 32:2717-2734. [PMID: 37369025 PMCID: PMC10460489 DOI: 10.1093/hmg/ddad100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/25/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
Inherited disorders of mitochondrial metabolism, including isolated methylmalonic aciduria, present unique challenges to energetic homeostasis by disrupting energy-producing pathways. To better understand global responses to energy shortage, we investigated a hemizygous mouse model of methylmalonyl-CoA mutase (Mmut)-type methylmalonic aciduria. We found Mmut mutant mice to have reduced appetite, energy expenditure and body mass compared with littermate controls, along with a relative reduction in lean mass but increase in fat mass. Brown adipose tissue showed a process of whitening, in line with lower body surface temperature and lesser ability to cope with cold challenge. Mutant mice had dysregulated plasma glucose, delayed glucose clearance and a lesser ability to regulate energy sources when switching from the fed to fasted state, while liver investigations indicated metabolite accumulation and altered expression of peroxisome proliferator-activated receptor and Fgf21-controlled pathways. Together, these shed light on the mechanisms and adaptations behind energy imbalance in methylmalonic aciduria and provide insight into metabolic responses to chronic energy shortage, which may have important implications for disease understanding and patient management.
Collapse
Affiliation(s)
- Marie Lucienne
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
- radiz – Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare Diseases, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Raffaele Gerlini
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Birgit Rathkolb
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Julia Calzada-Wack
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Patrick Forny
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology and Children’s Research Center, University Children's Hospital, University of Zurich, 8032 Zurich, Switzerland
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Florian Traversi
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
| | - Merima Forny
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
| | - Céline Bürer
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
| | - Antonio Aguilar-Pimentel
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sven Sauer
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital, Heidelberg, Germany
| | - Stefan Kölker
- Division of Pediatric Neurology and Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital, Heidelberg, Germany
| | - Joseph P Dewulf
- Department of Biochemistry, de Duve Institute, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium
- Department of Laboratory Medicine, Cliniques universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Guido T Bommer
- Department of Biochemistry, de Duve Institute, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium
| | - Daniel Hoces
- Institute of Food, Nutrition and Health, D-HEST, ETH Zurich, Zurich, Switzerland
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jan Rozman
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - D Sean Froese
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
- radiz – Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare Diseases, University of Zurich, Zurich, Switzerland
| | - Matthias R Baumgartner
- Division of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
- radiz – Rare Disease Initiative Zurich, Clinical Research Priority Program for Rare Diseases, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics and German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Freising, Germany
| |
Collapse
|
26
|
Vockley J, Burton B, Jurecka A, Ganju J, Leiro B, Zori R, Longo N. Challenges and strategies for clinical trials in propionic and methylmalonic acidemias. Mol Genet Metab 2023; 139:107612. [PMID: 37245378 DOI: 10.1016/j.ymgme.2023.107612] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 05/30/2023]
Abstract
Clinical trial development in rare diseases poses significant study design and methodology challenges, such as disease heterogeneity and appropriate patient selection, identification and selection of key endpoints, decisions on study duration, choice of control groups, selection of appropriate statistical analyses, and patient recruitment. Therapeutic development in organic acidemias (OAs) shares many challenges with other inborn errors of metabolism, such as incomplete understanding of natural history, heterogenous disease presentations, requirement for sensitive outcome measures and difficulties recruiting a small sample of participants. Here, we review strategies for the successful development of a clinical trial to evaluate treatment response in propionic and methylmalonic acidemias. Specifically, we discuss crucial decisions that may significantly impact success of the study, including patient selection, identification and selection of endpoints, determination of the study duration, consideration of control groups including natural history controls, and selection of appropriate statistical analyses. The significant challenges associated with designing a clinical trial in rare disease can sometimes be successfully met through strategic engagement with experts in the rare disease, seeking regulatory and biostatistical guidance, and early involvement of patients and families.
Collapse
Affiliation(s)
- Jerry Vockley
- Division Medical Genetics, Department of Pediatrics, University of Pittsburgh, School of Medicine, Center for Rare Disease Therapy, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Barbara Burton
- Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | - Agnieszka Jurecka
- CoA Therapeutics, Inc., a BridgeBio company, San Francisco, CA, USA.
| | - Jitendra Ganju
- Independent Consultant to BridgeBio, San Francisco, CA, USA
| | - Beth Leiro
- Independent Consultant to BridgeBio, San Francisco, CA, USA
| | - Roberto Zori
- Department of Pediatrics, Division of Genetics and Metabolism, University of Florida, Gainesville, FL, USA
| | - Nicola Longo
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
27
|
Manoli I, Gebremariam A, McCoy S, Pass AR, Gagné J, Hall C, Ferry S, Van Ryzin C, Sloan JL, Sacchetti E, Catesini G, Rizzo C, Martinelli D, Spada M, Dionisi-Vici C, Venditti CP. Biomarkers to predict disease progression and therapeutic response in isolated methylmalonic acidemia. J Inherit Metab Dis 2023; 46:554-572. [PMID: 37243446 PMCID: PMC10330948 DOI: 10.1002/jimd.12636] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/28/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023]
Abstract
Methylmalonic Acidemia (MMA) is a heterogenous group of inborn errors of metabolism caused by a defect in the methylmalonyl-CoA mutase (MMUT) enzyme or the synthesis and transport of its cofactor, 5'-deoxy-adenosylcobalamin. It is characterized by life-threatening episodes of ketoacidosis, chronic kidney disease, and other multiorgan complications. Liver transplantation can improve patient stability and survival and thus provides clinical and biochemical benchmarks for the development of hepatocyte-targeted genomic therapies. Data are presented from a US natural history protocol that evaluated subjects with different types of MMA including mut-type (N = 91), cblB-type (15), and cblA-type MMA (17), as well as from an Italian cohort of mut-type (N = 19) and cblB-type MMA (N = 2) subjects, including data before and after organ transplantation in both cohorts. Canonical metabolic markers, such as serum methylmalonic acid and propionylcarnitine, are variable and affected by dietary intake and renal function. We have therefore explored the use of the 1-13 C-propionate oxidation breath test (POBT) to measure metabolic capacity and the changes in circulating proteins to assess mitochondrial dysfunction (fibroblast growth factor 21 [FGF21] and growth differentiation factor 15 [GDF15]) and kidney injury (lipocalin-2 [LCN2]). Biomarker concentrations are higher in patients with the severe mut0 -type and cblB-type MMA, correlate with a decreased POBT, and show a significant response postliver transplant. Additional circulating and imaging markers to assess disease burden are necessary to monitor disease progression. A combination of biomarkers reflecting disease severity and multisystem involvement will be needed to help stratify patients for clinical trials and assess the efficacy of new therapies for MMA.
Collapse
Affiliation(s)
- Irini Manoli
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Abigael Gebremariam
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Samantha McCoy
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Alexandra R. Pass
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jack Gagné
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Camryn Hall
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Susan Ferry
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Carol Van Ryzin
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jennifer L. Sloan
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Elisa Sacchetti
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Giulio Catesini
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Cristiano Rizzo
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Diego Martinelli
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Marco Spada
- Division of Hepatobiliopancreatic Surgery, Liver and Kidney Tranplantation, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- European Research Network TransplantChild
| | - Carlo Dionisi-Vici
- Division of Metabolic Diseases, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Charles P. Venditti
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
28
|
Schumann A, Brutsche M, Havermans M, Grünert SC, Kölker S, Groß O, Hannibal L, Spiekerkoetter U. The impact of metabolic stressors on mitochondrial homeostasis in a renal epithelial cell model of methylmalonic aciduria. Sci Rep 2023; 13:7677. [PMID: 37169781 PMCID: PMC10175303 DOI: 10.1038/s41598-023-34373-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023] Open
Abstract
Methylmalonic aciduria (MMA-uria) is caused by deficiency of the mitochondrial enzyme methylmalonyl-CoA mutase (MUT). MUT deficiency hampers energy generation from specific amino acids, odd-chain fatty acids and cholesterol. Chronic kidney disease (CKD) is a well-known long-term complication. We exposed human renal epithelial cells from healthy controls and MMA-uria patients to different culture conditions (normal treatment (NT), high protein (HP) and isoleucine/valine (I/V)) to test the effect of metabolic stressors on renal mitochondrial energy metabolism. Creatinine levels were increased and antioxidant stress defense was severely comprised in MMA-uria cells. Alterations in mitochondrial homeostasis were observed. Changes in tricarboxylic acid cycle metabolites and impaired energy generation from fatty acid oxidation were detected. Methylcitrate as potentially toxic, disease-specific metabolite was increased by HP and I/V load. Mitophagy was disabled in MMA-uria cells, while autophagy was highly active particularly under HP and I/V conditions. Mitochondrial dynamics were shifted towards fission. Sirtuin1, a stress-resistance protein, was down-regulated by HP and I/V exposure in MMA-uria cells. Taken together, both interventions aggravated metabolic fingerprints observed in MMA-uria cells at baseline. The results point to protein toxicity in MMA-uria and lead to a better understanding, how the accumulating, potentially toxic organic acids might trigger CKD.
Collapse
Affiliation(s)
- Anke Schumann
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Mathildenstr. 1, 79106, Freiburg, Germany.
| | - Marion Brutsche
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Mathildenstr. 1, 79106, Freiburg, Germany
| | - Monique Havermans
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sarah C Grünert
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Mathildenstr. 1, 79106, Freiburg, Germany
| | - Stefan Kölker
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Luciana Hannibal
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Laboratory of Clinical Biochemistry and Metabolism, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Ute Spiekerkoetter
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Mathildenstr. 1, 79106, Freiburg, Germany
| |
Collapse
|
29
|
Maines E, Moretti M, Vitturi N, Gugelmo G, Fasan I, Lenzini L, Piccoli G, Gragnaniello V, Maiorana A, Soffiati M, Burlina A, Franceschi R. Understanding the Pathogenesis of Cardiac Complications in Patients with Propionic Acidemia and Exploring Therapeutic Alternatives for Those Who Are Not Eligible or Are Waiting for Liver Transplantation. Metabolites 2023; 13:563. [PMID: 37110221 PMCID: PMC10143878 DOI: 10.3390/metabo13040563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The guidelines for the management of patients affected by propionic acidemia (PA) recommend standard cardiac therapy in the presence of cardiac complications. A recent revision questioned the impact of high doses of coenzyme Q10 on cardiac function in patients with cardiomyopathy (CM). Liver transplantation is a therapeutic option for several patients since it may stabilize or reverse CM. Both the patients waiting for liver transplantation and, even more, the ones not eligible for transplant programs urgently need therapies to improve cardiac function. To this aim, the identification of the pathogenetic mechanisms represents a key point. Aims: This review summarizes: (1) the current knowledge of the pathogenetic mechanisms underlying cardiac complications in PA and (2) the available and potential pharmacological options for the prevention or the treatment of cardiac complications in PA. To select articles, we searched the electronic database PubMed using the Mesh terms "propionic acidemia" OR "propionate" AND "cardiomyopathy" OR "Long QT syndrome". We selected 77 studies, enlightening 12 potential disease-specific or non-disease-specific pathogenetic mechanisms, namely: impaired substrate delivery to TCA cycle and TCA dysfunction, secondary mitochondrial electron transport chain dysfunction and oxidative stress, coenzyme Q10 deficiency, metabolic reprogramming, carnitine deficiency, cardiac excitation-contraction coupling alteration, genetics, epigenetics, microRNAs, micronutrients deficiencies, renin-angiotensin-aldosterone system activation, and increased sympathetic activation. We provide a critical discussion of the related therapeutic options. Current literature supports the involvement of multiple cellular pathways in cardiac complications of PA, indicating the growing complexity of their pathophysiology. Elucidating the mechanisms responsible for such abnormalities is essential to identify therapeutic strategies going beyond the correction of the enzymatic defect rather than engaging the dysregulated mechanisms. Although these approaches are not expected to be resolutive, they may improve the quality of life and slow the disease progression. Available pharmacological options are limited and tested in small cohorts. Indeed, a multicenter approach is mandatory to strengthen the efficacy of therapeutic options.
Collapse
Affiliation(s)
- Evelina Maines
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Michele Moretti
- Division of Cardiology, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Nicola Vitturi
- Division of Metabolic Diseases, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Giorgia Gugelmo
- Division of Clinical Nutrition, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Ilaria Fasan
- Division of Clinical Nutrition, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Livia Lenzini
- Emergency Medicine Unit, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Giovanni Piccoli
- CIBIO, Department of Cellular, Computational and Integrative Biology, Italy & Dulbecco Telethon Institute, Università degli Studi di Trento, 38123 Trento, Italy
| | - Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, Department of Women’s and Children’s Health, University Hospital, 35128 Padova, Italy
| | - Arianna Maiorana
- Division of Metabolism and Research Unit of Metabolic Biochemistry, Bambino Gesù Children’s Hospital-IRCCS, 00165 Rome, Italy
| | - Massimo Soffiati
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, Department of Women’s and Children’s Health, University Hospital, 35128 Padova, Italy
| | - Roberto Franceschi
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| |
Collapse
|
30
|
Mütze U, Gleich F, Barić I, Baumgartner M, Burlina A, Chapman KA, Chien Y, Cortès‐Saladelafont E, De Laet C, Dobbelaere D, Eysken F, Gautschi M, Santer R, Häberle J, Joaquín C, Karall D, Lindner M, Lund AM, Mühlhausen C, Murphy E, Roland D, Ruiz Gomez A, Skouma A, Grünert SC, Wagenmakers M, Garbade SF, Kölker S, Boy N. Impact of the SARS-CoV-2 pandemic on the health of individuals with intoxication-type metabolic diseases-Data from the E-IMD consortium. J Inherit Metab Dis 2023; 46:220-231. [PMID: 36266255 PMCID: PMC9874390 DOI: 10.1002/jimd.12572] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/20/2022] [Accepted: 10/19/2022] [Indexed: 01/27/2023]
Abstract
The SARS-CoV-2 pandemic challenges healthcare systems worldwide. Within inherited metabolic disorders (IMDs) the vulnerable subgroup of intoxication-type IMDs such as organic acidurias (OA) and urea cycle disorders (UCD) show risk for infection-induced morbidity and mortality. This study (observation period February 2020 to December 2021) evaluates impact on medical health care as well as disease course and outcome of SARS-CoV-2 infections in patients with intoxication-type IMDs managed by participants of the European Registry and Network for intoxication type metabolic diseases Consortium (E-IMD). Survey's respondents managing 792 patients (n = 479 pediatric; n = 313 adult) with intoxication-type IMDs (n = 454 OA; n = 338 UCD) in 14 countries reported on 59 (OA: n = 36; UCD: n = 23), SARS-CoV-2 infections (7.4%). Medical services were increasingly requested (95%), mostly alleviated by remote technologies (86%). Problems with medical supply were scarce (5%). Regular follow-up visits were reduced in 41% (range 10%-50%). Most infected individuals (49/59; 83%) showed mild clinical symptoms, while 10 patients (17%; n = 6 OA including four transplanted MMA patients; n = 4 UCD) were hospitalized (metabolic decompensation in 30%). ICU treatment was not reported. Hospitalization rate did not differ for diagnosis or age group (p = 0.778). Survival rate was 100%. Full recovery was reported for 100% in outpatient care and 90% of hospitalized individuals. SARS-CoV-2 impacts health care of individuals with intoxication-type IMDs worldwide. Most infected individuals, however, showed mild symptoms and did not require hospitalization. SARS-CoV-2-induced metabolic decompensations were usually mild without increased risk for ICU treatment. Overall prognosis of infected individuals is very promising and IMD-specific or COVID-19-related complications have not been observed.
Collapse
Affiliation(s)
- Ulrike Mütze
- Centre for Child and Adolescent Medicine, Division of Neuropaediatrics and Metabolic Medicine, Department of General PaediatricsUniversity Hospital HeidelbergHeidelbergGermany
| | - Florian Gleich
- Centre for Child and Adolescent Medicine, Division of Neuropaediatrics and Metabolic Medicine, Department of General PaediatricsUniversity Hospital HeidelbergHeidelbergGermany
| | - Ivo Barić
- University of Zagreb, School of Medicine and Department of PediatricsUniversity Hospital Center ZagrebZagrebCroatia
| | - Mathias Baumgartner
- University Children's Hospital and Children's Research Center, University of ZurichZurichSwitzerland
| | - Alberto Burlina
- Division of Inherited Metabolic DiseaseUniversity Hospital PadovaPadovaItaly
| | | | - Yin‐Hsiu Chien
- Department of Medical Genetics and PediatricsNational Taiwan University HospitalTaipeiTaiwan
| | - Elisenda Cortès‐Saladelafont
- University Hospital Germans Trias i Pujol, Department of Paediatrics, Unit of Inherited Metabolic Diseases and NeuropediatricsUniversitat Autònoma de BarcelonaBadalonaSpain
| | - Corinne De Laet
- Hôpital Universitaire des Enfants Reine Fabiola‐Université Libre de BruxellesBrusselsBelgium
| | - Dries Dobbelaere
- Medical Reference Center for Inherited Metabolic DiseasesJeanne de Flandre University Hospital and RADEME Research Team for Rare Metabolic and Developmental Diseases, CHRU LilleLilleFrance
| | - Francois Eysken
- Department of Pediatric Inherited Metabolic DiseasesUniversity Hospital of Antwerp UZAAntwerpBelgium
| | - Matthias Gautschi
- Department of Paediatrics, Division of Paediatric Endocrinology, Diabetology and Metabolism, InselspitalUniversity Hospital BernBernSwitzerland
| | - Rene Santer
- University Children's Hospital, University Medical Centre Hamburg‐EppendorfHamburgGermany
| | - Johannes Häberle
- University Children's Hospital and Children's Research Center, University of ZurichZurichSwitzerland
| | - Clara Joaquín
- Endocrinology and Nutrition DepartmentHospital Universitari Germans Trias i PujolBadalonaSpain
| | - Daniela Karall
- Clinic for PediatricsMedical University of InnsbruckInnsbruckAustria
| | - Martin Lindner
- Goethe University Frankfurt/Main, Uni.‐Children's HospitalFrankfurt/MainGermany
| | - Allan M. Lund
- Centre Inherited Metabolic Diseases, Departments of Paediatrics and Clinical GeneticsCopenhagen University HospitalCopenhagenDenmark
| | - Chris Mühlhausen
- Universitätsmedizin Göttingen, Georg‐August‐UniversitätGöttingenGermany
| | - Elaine Murphy
- National Hospital for Neurology and Neurosurgery, Charles Dent Metabolic UnitLondonUK
| | - Dominique Roland
- Institute of Pathology and Human Genetics (IPG)CharleroisBelgium
| | | | - Anastasia Skouma
- Institouto Ygeias Tou Paidiou, Institute of Child HealthAthensGreece
| | - Sarah C. Grünert
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical CentreUniversity of Freiburg, Faculty of MedicineFreiburgGermany
| | | | - Sven F. Garbade
- Centre for Child and Adolescent Medicine, Division of Neuropaediatrics and Metabolic Medicine, Department of General PaediatricsUniversity Hospital HeidelbergHeidelbergGermany
| | - Stefan Kölker
- Centre for Child and Adolescent Medicine, Division of Neuropaediatrics and Metabolic Medicine, Department of General PaediatricsUniversity Hospital HeidelbergHeidelbergGermany
| | - Nikolas Boy
- Centre for Child and Adolescent Medicine, Division of Neuropaediatrics and Metabolic Medicine, Department of General PaediatricsUniversity Hospital HeidelbergHeidelbergGermany
| |
Collapse
|
31
|
Chen T, Gao Y, Zhang S, Wang Y, Sui C, Yang L. Methylmalonic acidemia: Neurodevelopment and neuroimaging. Front Neurosci 2023; 17:1110942. [PMID: 36777632 PMCID: PMC9909197 DOI: 10.3389/fnins.2023.1110942] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Methylmalonic acidemia (MMA) is a genetic disease of abnormal organic acid metabolism, which is one of the important factors affecting the survival rate and quality of life of newborns or infants. Early detection and diagnosis are particularly important. The diagnosis of MMA mainly depends on clinical symptoms, newborn screening, biochemical detection, gene sequencing and neuroimaging diagnosis. The accumulation of methylmalonic acid and other metabolites in the body of patients causes brain tissue damage, which can manifest as various degrees of intellectual disability and severe neurological dysfunction. Neuroimaging examination has important clinical significance in the diagnosis and prognosis of MMA. This review mainly reviews the etiology, pathogenesis, and nervous system development, especially the neuroimaging features of MMA.
Collapse
Affiliation(s)
- Tao Chen
- Department of Clinical Laboratory, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yian Gao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shengdong Zhang
- Department of Radiology, Shandong Yinan People’s Hospital, Linyi, Shandong, China
| | - Yuanyuan Wang
- Department of Radiology, Binzhou Medical University, Yantai, Shandong, China
| | - Chaofan Sui
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Linfeng Yang
- Department of Radiology, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China,*Correspondence: Linfeng Yang,
| |
Collapse
|
32
|
Kovacevic A, Garbade SF, Hörster F, Hoffmann GF, Gorenflo M, Mereles D, Kölker S, Staufner C. Evaluation of Right Ventricular Function in Patients with Propionic Acidemia-A Cross-Sectional Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10010113. [PMID: 36670663 PMCID: PMC9856918 DOI: 10.3390/children10010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023]
Abstract
(1) Background: In propionic acidemia (PA), myocardial involvement often leads to progressive cardiac dysfunction of the left ventricle (LV). Cardiomyopathy (CM) is an important contributor to mortality. Although known to be of prognostic value in CM, there are no published data on right ventricular (RV) function in PA patients. (2) Methods: In this cross-sectional single-center study, systolic and diastolic RV function of PA patients was assessed by echocardiography, including frequency, onset, and combinations of echocardiographic parameters, as well as correlations to LV size and function. (3) Results: N = 18 patients were enrolled. Tricuspid annulus S' was abnormal in 16.7%, RV-longitudinal strain in 11.1%, tricuspid annular plane systolic excursion (TAPSE) in 11.1%, Tricuspid valve (TV) E/e' in 33.3%, and TV E/A in 16.7%. The most prevalent combinations of pathological parameters were TV E/A + TV E/e' and TAPSE + TV S'. With age, the probability of developing abnormal RV function increases according to age-dependent normative data. There is a significant correlation between TAPSE and mitral annular plane systolic excursion (MAPSE), and RV/LV-longitudinal strain (p ≤ 0.05). N = 5 individuals died 1.94 years (mean) after cardiac evaluation for this study, and all had abnormal RV functional parameters. (4) Conclusions: Signs of diastolic RV dysfunction can be found in up to one third of individuals, and systolic RV dysfunction in 16.7% of individuals in our cohort. RV function is impaired in PA patients with a poor outcome. RV functional parameters should be used to complement clinical and left ventricular echocardiographic findings.
Collapse
Affiliation(s)
- Alexander Kovacevic
- Department of Pediatric and Congenital Cardiology, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Sven F. Garbade
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Friederike Hörster
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Georg F. Hoffmann
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Matthias Gorenflo
- Department of Pediatric and Congenital Cardiology, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Derliz Mereles
- Department of Cardiology, Angiology and Pulmology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Stefan Kölker
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Christian Staufner
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
33
|
Forny P, Hörster F, Baumgartner MR, Kölker S, Boy N. How guideline development has informed clinical research for organic acidurias (et vice versa). J Inherit Metab Dis 2023; 46:520-535. [PMID: 36591944 DOI: 10.1002/jimd.12586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/22/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Organic acidurias, such as glutaric aciduria type 1 (GA1), methylmalonic (MMA), and propionic aciduria (PA) are a prominent group of inherited metabolic diseases involving accumulation of eponymous metabolites causing endogenous intoxication. For all three conditions, guidelines for diagnosis and management have been developed and revised over the last years, resulting in three revisions for GA1 and one revision for MMA/PA. The process of clinical guideline development in rare metabolic disorders is challenged by the scarcity and limited quality of evidence available. The body of literature is often fragmentary and where information is present, it is usually derived from small sample sizes. Therefore, the development of guidelines for GA1 and MMA/PA was initially confronted with a poor evidence foundation that hindered formulation of concrete recommendations in certain contexts, triggering specific research projects and initiation of longitudinal, prospective observational studies using patient registries. Reversely, these observational studies contributed to evaluate the value of newborn screening, phenotypic diversities, and treatment effects, thus significantly improving the quality of evidence and directly influencing formulation and evidence levels of guideline recommendations. Here, we present insights into interactions between guideline development and (pre)clinical research for GA1 and MMA/PA, and demonstrate how guidelines gradually improved from revision to revision. We describe how clinical studies help to unravel the relative impact of therapeutic interventions on outcome and conclude that despite new and better quality of research data over the last decades, significant shortcomings of evidence regarding prognosis and treatment remain. It appears that development of clinical guidelines can directly help to guide research, and vice versa.
Collapse
Affiliation(s)
- Patrick Forny
- Division of Metabolism and Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Friederike Hörster
- Division of Neuropaediatrics and Metabolic Medicine, Department of General Paediatrics, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Stefan Kölker
- Division of Neuropaediatrics and Metabolic Medicine, Department of General Paediatrics, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Nikolas Boy
- Division of Neuropaediatrics and Metabolic Medicine, Department of General Paediatrics, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
34
|
Identification of potential interferents of methylmalonic acid: A previously unrecognized pitfall in clinical diagnostics and newborn screening. Clin Biochem 2023; 111:72-80. [PMID: 36202155 DOI: 10.1016/j.clinbiochem.2022.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Determination of methylmalonic acid (MMA) from dried blood spots (DBS) is commonly performed in clinical diagnostics and newborn screening for propionic acidemia (PA) and methylmalonic acidemia. Isobaric compounds of MMA having the same mass can affect diagnostic reliability and quantitative results, which represents a previously unrecognized pitfall in clinical assays for MMA. We set out to identify interfering substances of MMA in DBS, serum and urine samples from confirmed patients with PA and methylmalonic acidemia. METHODS Techniques included quadrupole time-of-flight high-resolution mass spectrometry (QTOF HR-MS), nuclear magnetic resonance (NMR) spectroscopy, liquid chromatography (LC) and tandem mass spectrometry (MS/MS). RESULTS The five isobaric metabolites detected in DBS, serum and urine from PA and methylmalonic acidemia patients were confirmed as 2-methyl-3-hydroxybutyrate, 3-hydroxyisovalerate, 2-hydroxyisovalerate, 3-hydroxyvalerate and succinate using a series of experiments. An additional unknown substance with low abundance remained unidentified. CONCLUSIONS The presented results facilitate the diagnostic and quantitative reliability of the MMA determination in clinical assays. Isobaric species should be investigated in assays for MMA to eliminate possible interference in a wide range of conditions including PA, methylmalonic acidemia, a vitamin B12 deficiency, ketosis and lactic acidosis.
Collapse
|
35
|
Kovacevic A, Garbade SF, Hörster F, Hoffmann GF, Gorenflo M, Mereles D, Kölker S, Staufner C. Detection of early cardiac disease manifestation in propionic acidemia - Results of a monocentric cross-sectional study. Mol Genet Metab 2022; 137:349-358. [PMID: 36395710 DOI: 10.1016/j.ymgme.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND In propionic acidemia (PA) myocardial involvement is common and includes development of cardiomyopathy, life-threatening acute heart failure, and acquired long-QT syndrome. We sought to investigate which echocardiographic parameters of left ventricular systolic and diastolic function indicate early cardiac disease manifestation in PA. METHODS This is a prospective observational study (cross-sectional design) in a Tertiary Medical Care Center. Individuals with confirmed PA were enrolled and the following cardiac investigations were performed in all study individuals: echocardiographic measurements of systolic and diastolic left ventricular (LV) function (LV fractional shortening (LV-FS), LV ejection fraction by biplane modified Simpson's (LV-EF), mitral annular plane systolic excursion (MAPSE), LV global longitudinal strain (LV-GLS) by speckle tracking echocardiography (STE), pulsed Doppler analyses of mitral valve (MV) inflow velocities (MV E/A) and MV deceleration time (DT-E), tissue doppler imaging (TDI) of the mitral annulus (MV E/e'), and LV myocardial performance index (LV-MPI)). LV and left atrial (LA) diameters were assessed. 12‑lead electrocardiograms (ECG) were recorded and corrected QT intervals (QTc) calculated. Clinical phenotype and laboratory parameters at the time of cardiac investigation were assessed. Besides descriptive analyses we analyzed frequency, onset, and combinations of echocardiographic and ECG data as well as their correlations with clinical and biochemical findings. The effects of 'age at visit' and LV functional parameters on QTc were analyzed with multiple regression. RESULTS A total of 18 patients with confirmed PA were enrolled. Median age at PA onset was 6 days (range 1-357 days). Median age at visit for cardiac evaluation was 13.1 years (range 0.6-28.1 years). LV-GLS was abnormal in 72.2%, LV-EF in 61.1%, MAPSE in 50%, MV E/e' in 44.4%, LV-MPI in 33.3%, LV-FS in 33.3%, and MV E/A in 27.8%. In cases with normal or near normal LV-FS, LV-GLS was pathological in 5/10, LV-EF in 4/10, and MAPSE in 3/10. The probability of developing LV dysfunction - systolic and diastolic - increases with age. LV-MPI is a reliable parameter to indicate systolic LV-dysfunction in combination with a dilated LV, i. e. dilated cardiomyopathy (DCM) in PA. Multiple regression reveals a significant positive association between LV diameters and QTc. Abnormal LV-GLS significantly correlates with reduced muscle strength, muscle tone and/or abnormal gross motor function. CONCLUSIONS Our data suggests a high prevalence of cardiac disease manifestation in PA, considerably higher than in previous studies, where only LV-FS was used to assess LV function. Usage of advanced echocardiographic techniques, such as LV-GLS assessment, may allow for early detection of subtle LV dysfunction in PA, and may lead to timely cardiac treatment but also consideration of liver transplantation to prevent development of manifest cardiac complications.
Collapse
Affiliation(s)
- Alexander Kovacevic
- Department of Pediatric and Congenital Cardiology, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Sven F Garbade
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Friederike Hörster
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Georg F Hoffmann
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Matthias Gorenflo
- Department of Pediatric and Congenital Cardiology, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Derliz Mereles
- Department of Cardiology, Angiology and Pulmology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Stefan Kölker
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Christian Staufner
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| |
Collapse
|
36
|
Scharre S, Posset R, Garbade SF, Gleich F, Seidl MJ, Druck A, Okun JG, Gropman AL, Nagamani SCS, Hoffmann GF, Kölker S, Zielonka M. Predicting the disease severity in male individuals with ornithine transcarbamylase deficiency. Ann Clin Transl Neurol 2022; 9:1715-1726. [PMID: 36217298 PMCID: PMC9639638 DOI: 10.1002/acn3.51668] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVE Ornithine transcarbamylase deficiency (OTC-D) is an X-linked metabolic disease and the most common urea cycle disorder. Due to high phenotypic heterogeneity, ranging from lethal neonatal hyperammonemic events to moderate symptoms and even asymptomatic individuals, the prediction of the disease course at an early disease stage is very important to individually adjust therapies such as medical treatment or liver transplantation. In this translational study, we developed a severity-adjusted classification system based on in vitro residual enzymatic OTC activity. METHODS Applying a cell-based expression system, residual enzymatic OTC activities of 71 pathogenic OTC variants were spectrophotometrically determined and subsequently correlated with clinical and biochemical outcome parameters of 119 male individuals with OTC-D (mOTC-D) as reported in the UCDC and E-IMD registries. RESULTS Integration of multiple data sources enabled the establishment of a robust disease prediction model for mOTC-D. Residual enzymatic OTC activity not only correlates with age at first symptoms, initial peak plasma ammonium concentration and frequency of metabolic decompensations but also predicts mortality. The critical threshold of 4.3% residual enzymatic activity distinguishes a severe from an attenuated phenotype. INTERPRETATION Residual enzymatic OTC activity reliably predicts the disease severity in mOTC-D and could thus serve as a tool for severity-adjusted evaluation of therapeutic strategies and counselling patients and parents.
Collapse
Affiliation(s)
- Svenja Scharre
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Roland Posset
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Sven F. Garbade
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Florian Gleich
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Marie J. Seidl
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Ann‐Catrin Druck
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Jürgen G. Okun
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Andrea L. Gropman
- Division of Neurodevelopmental Pediatrics and Neurogenetics, Children's National Health System and The George Washington School of MedicineWashingtonDistrict of ColumbiaUSA
| | - Sandesh C. S. Nagamani
- Department of Molecular and Human GeneticsBaylor College of Medicine and Texas Children's HospitalHoustonTexasUSA
| | - Georg F. Hoffmann
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Stefan Kölker
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
| | - Matthias Zielonka
- Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent MedicineUniversity Hospital HeidelbergHeidelbergGermany
- Heidelberg Research Center for Molecular Medicine (HRCMM)HeidelbergGermany
| |
Collapse
|
37
|
Healy L, O'Shea M, McNulty J, King G, Twomey E, Treacy E, Crushell E, Hughes J, Knerr I, Monavari AA. Glutaric aciduria type 1: Diagnosis, clinical features and long-term outcome in a large cohort of 34 Irish patients. JIMD Rep 2022; 63:379-387. [PMID: 35822093 PMCID: PMC9259398 DOI: 10.1002/jmd2.12302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/08/2022] Open
Abstract
Glutaric aciduria type 1 (GA1) is a rare neurometabolic disorder that can lead to encephalopathic crises and severe dystonic movement disorders. Adherence to strict dietary restriction, in particular a diet low in lysine, carnitine supplementation and emergency treatment in pre-symptomatic patients diagnosed by high-risk screen (HRS) or newborn screen (NBS) leads to a favourable outcome. We present biochemical and clinical characteristics and long-term outcome data of 34 Irish patients with GA1 aged 1-40 years. Sixteen patients were diagnosed clinically, and 17 patients by HRS, prior to introduction of NBS for GA1 in the Republic of Ireland in 2018. One patient was diagnosed by NBS. Clinical diagnosis was at a median of 1 year (range 1 month to 8 years) and by HRS was at a median of 4 days (range 3 days to 11 years). 14/18 (77.8%) diagnosed by HRS or NBS had neither clinical manifestations nor radiological features of GA1, or had radiological features only, compared to 0/16 (0%) diagnosed clinically (p < 0.001). Patients diagnosed clinically who survived to school-age were more likely to have significant cerebral palsy and dystonia (7/11; 63.6% vs. 0/13; 0%, p < 0.001). They were less likely to be in mainstream school versus the HRS group (5/10; 50% vs. 12/13; 92.3%; p = 0.012). Clinical events occurring after 6 years of age were unusual, but included spastic diplegia, thalamic haemorrhage, Chiari malformation, pituitary hormone deficiency and epilepsy. The exact aetiology of these events is unclear.
Collapse
Affiliation(s)
- Lydia Healy
- National Centre of Inherited Metabolic Disorders, Children's Health Ireland at Temple StreetDublinRepublic of Ireland
| | - Meabh O'Shea
- National Centre of Inherited Metabolic Disorders, Children's Health Ireland at Temple StreetDublinRepublic of Ireland
- European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN)European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN), National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street and Mater Misericordiae University HospitalDublinRepublic of Ireland
| | - Jennifer McNulty
- National Centre of Inherited Metabolic Disorders, Children's Health Ireland at Temple StreetDublinRepublic of Ireland
- European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN)European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN), National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street and Mater Misericordiae University HospitalDublinRepublic of Ireland
| | - Graham King
- National Centre of Inherited Metabolic Disorders, Children's Health Ireland at Temple StreetDublinRepublic of Ireland
| | - Eilish Twomey
- Department of RadiologyChildren's Health Ireland at Temple StreetDublinRepublic of Ireland
| | - Eileen Treacy
- European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN)European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN), National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street and Mater Misericordiae University HospitalDublinRepublic of Ireland
- National Adult Centre for Inherited Metabolic DisordersMater Misericordiae University HospitalDublinRepublic of Ireland
- University College DublinDublinRepublic of Ireland
- University of DublinDublinRepublic of Ireland
| | - Ellen Crushell
- National Centre of Inherited Metabolic Disorders, Children's Health Ireland at Temple StreetDublinRepublic of Ireland
- European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN)European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN), National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street and Mater Misericordiae University HospitalDublinRepublic of Ireland
- University College DublinDublinRepublic of Ireland
| | - Joanne Hughes
- National Centre of Inherited Metabolic Disorders, Children's Health Ireland at Temple StreetDublinRepublic of Ireland
- European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN)European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN), National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street and Mater Misericordiae University HospitalDublinRepublic of Ireland
| | - Ina Knerr
- National Centre of Inherited Metabolic Disorders, Children's Health Ireland at Temple StreetDublinRepublic of Ireland
- European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN)European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN), National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street and Mater Misericordiae University HospitalDublinRepublic of Ireland
- University College DublinDublinRepublic of Ireland
| | - Ahmad Ardeshir Monavari
- National Centre of Inherited Metabolic Disorders, Children's Health Ireland at Temple StreetDublinRepublic of Ireland
- European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN)European Reference Network for Rare Hereditary Metabolic Disorders (MetabERN), National Centre for Inherited Metabolic Disorders, Children's Health Ireland at Temple Street and Mater Misericordiae University HospitalDublinRepublic of Ireland
- University College DublinDublinRepublic of Ireland
| |
Collapse
|
38
|
Ding S, Liang L, Qiu W, Zhang H, Xiao B, Dong L, Ji W, Xu F, Gong Z, Gu X, Wang L, Han L. Prenatal Diagnosis of Isovaleric Acidemia From Amniotic Fluid Using Genetic and Biochemical Approaches. Front Genet 2022; 13:898860. [PMID: 35846131 PMCID: PMC9280075 DOI: 10.3389/fgene.2022.898860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Isovaleric acidemia (IVA) is an inborn error of leucine metabolism and different approaches have been applied to its prenatal diagnosis. However, systemic application of a biochemical strategy is rare. To evaluate its reliability and validity, we conducted a retrospective study of our experience with metabolite measurement together with genetic analysis in IVA prenatal diagnosis at a single center. Methods: A total of eight pregnancies whose probands were diagnosed as IVA were referred to our center for prenatal diagnosis. Prenatal data of genetic analysis and metabolite measurement using tandem mass spectrometry (MS/MS) and gas chromatography/mass spectrometry (GC/MS) in amniotic fluid (AF) samples were retrospectively reviewed. Results: Genetic and biochemical results were both available in these eight at-risk fetuses. Among them, two fetuses had higher levels of isovalerylcarnitine (C5) and C5/acetylcarnitine (C2) in AF compared with normal reference range and, thus, were determined to be affected, both of whom were found to carry compound heterogeneous mutations according to genetic analysis. The remaining six fetuses were determined to be unaffected based on a normal AF metabolite profile, except one showed slightly elevated C5 and they were found to be carriers according to genetic analysis. However, the level of isovalerylglycine (IVG) could not be detected at all in both groups. Conclusion: The biochemical analysis, as a quick and convenient method, could be an additional reliable option for the prenatal diagnosis of IVA, especially in families with inconclusive genetic results, and can achieve a more precise diagnosis in conjunction with mutation analysis.
Collapse
Affiliation(s)
- Si Ding
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lili Liang
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjuan Qiu
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiwen Zhang
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Xiao
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Dong
- Neonatal Disease Screening Center, Zibo Maternal and Child Health Hospital, Zibo, China
| | - Wenjun Ji
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Xu
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuwen Gong
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuefan Gu
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wang
- Center for Prenatal Diagnosis, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Lei Wang, ; Lianshu Han,
| | - Lianshu Han
- Department of Pediatric Endocrinology and Genetic Metabolism, Xinhua Hospital, Shanghai Institute of Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Lei Wang, ; Lianshu Han,
| |
Collapse
|
39
|
The Regulation and Characterization of Mitochondrial-Derived Methylmalonic Acid in Mitochondrial Dysfunction and Oxidative Stress: From Basic Research to Clinical Practice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7043883. [PMID: 35656023 PMCID: PMC9155905 DOI: 10.1155/2022/7043883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/16/2022] [Accepted: 04/23/2022] [Indexed: 01/11/2023]
Abstract
Methylmalonic acid (MMA) can act as a diagnosis of hereditary methylmalonic acidemia and assess the status of vitamin B12. Moreover, as a new potential biomarker, it has been widely reported to be associated with the progression and prognosis of chronic diseases such as cardiovascular events, renal insufficiency, cognitive impairment, and cancer. MMA accumulation may cause oxidative stress and impair mitochondrial function, disrupt cellular energy metabolism, and trigger cell death. This review primarily focuses on the mechanisms and epidemiology or progression in the clinical study on MMA.
Collapse
|
40
|
Scharping M, Brennenstuhl H, Garbade SF, Wild B, Posset R, Zielonka M, Kölker S, Haun MW, Opladen T. Unmet Needs of Parents of Children with Urea Cycle Disorders. CHILDREN 2022; 9:children9050712. [PMID: 35626889 PMCID: PMC9140128 DOI: 10.3390/children9050712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 12/26/2022]
Abstract
(1) Background: Phenotypic diversity and long-term health outcomes of individuals with urea cycle disorders (UCDs) have been described in detail. However, there is limited information on the burden on affected families. (2) Methods: To evaluate the family burden in parents with children suffering from UCDs, we used validated questionnaires. Socio-demographic characteristics were evaluated, and an adapted version of the Parental Need Scale for Rare Diseases questionnaire was used. The survey was conducted in families of UCD patients cared for at the University Children’s Hospital Heidelberg. (3) Results: From April to November 2021, 59 participants were interviewed (mothers n = 34, fathers n = 25). The affected patients most frequently suffered from ornithine transcarbamylase deficiency (OTC-D) (female n = 12, male n = 12), followed by argininosuccinate synthetase deficiency (ASS-D, n = 13) and argininosuccinate lyase deficiency (ASL-D, n = 8). About one-third of the participants were “dissatisfied” or “extremely dissatisfied” with health professionals’ disease knowledge. In addition, 30% of the participants reported a medium or high need for “additional information on the development of their children”, and 44% reported a medium or high need “for information on available services”. A majority of 68% reported a need for additional support regarding services such as support groups (42%) or psychological counseling (29%). (4) Conclusions: Our study indicates that there is an unmet need for sufficient information about the development of children with UCDs, as well as for information about available support services for families with UCD patients. Furthermore, the results highlight the importance of establishing or improving family-centered care approaches. This pilot study may serve as a template for the assessment of the family burden associated with other inherited metabolic diseases.
Collapse
Affiliation(s)
- Mara Scharping
- Division of Child Neurology and Metabolic Medicine, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (H.B.); (S.F.G.); (R.P.); (M.Z.); (S.K.)
| | - Heiko Brennenstuhl
- Division of Child Neurology and Metabolic Medicine, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (H.B.); (S.F.G.); (R.P.); (M.Z.); (S.K.)
| | - Sven F. Garbade
- Division of Child Neurology and Metabolic Medicine, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (H.B.); (S.F.G.); (R.P.); (M.Z.); (S.K.)
| | - Beate Wild
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, 69120 Heidelberg, Germany; (B.W.); (M.W.H.)
| | - Roland Posset
- Division of Child Neurology and Metabolic Medicine, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (H.B.); (S.F.G.); (R.P.); (M.Z.); (S.K.)
| | - Matthias Zielonka
- Division of Child Neurology and Metabolic Medicine, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (H.B.); (S.F.G.); (R.P.); (M.Z.); (S.K.)
| | - Stefan Kölker
- Division of Child Neurology and Metabolic Medicine, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (H.B.); (S.F.G.); (R.P.); (M.Z.); (S.K.)
| | - Markus W. Haun
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, 69120 Heidelberg, Germany; (B.W.); (M.W.H.)
| | - Thomas Opladen
- Division of Child Neurology and Metabolic Medicine, Centre for Child and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (M.S.); (H.B.); (S.F.G.); (R.P.); (M.Z.); (S.K.)
- Correspondence:
| |
Collapse
|
41
|
Liu Y, Chen Z, Dong H, Ding Y, He R, Kang L, Li D, Shen M, Jin Y, Zhang Y, Song J, Tian Y, Cao Y, Liang D, Yang Y. Analysis of the relationship between phenotypes and genotypes in 60 Chinese patients with propionic acidemia: a fourteen-year experience at a tertiary hospital. Orphanet J Rare Dis 2022; 17:135. [PMID: 35331292 PMCID: PMC8944130 DOI: 10.1186/s13023-022-02271-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/20/2022] [Indexed: 11/21/2022] Open
Abstract
Background Propionic acidemia is a severe inherited metabolic disorder, caused by the deficiency of propionyl-CoA carboxylase which encoded by the PCCA and PCCB genes. The aim of the study was to investigate the clinical features and outcomes, molecular epidemiology and phenotype-genotype relationship in Chinese population. Methods We conducted a retrospective study of 60 Chinese patients diagnosed at Peking University First Hospital from 2007 to 2020. Their clinical and laboratory data were reviewed. The next-generation sequencing was conducted on blood samples from 58 patients. Results Only 5 (8.3%) patients were identified by newborn screening. In the rest 55 patients, 25 had early-onset (≤ 3 months) disease and 30 had late-onset (> 3 months) disease. Neurological abnormalities were the most frequent complications. Five cases detected by newborn screening had basically normal development. Nine (15%) cases died in our cohort. 24 patients (41.4%) harbored PCCA variants, and 34 (58.6%) harbored PCCB variants. 30 (11 reported and 19 novel) variants in PCCA and 28 (18 reported and 10 novel) variants in PCCB mere identified. c.2002G>A and c.937C>T in PCCA, and c.838dupC in PCCB were the most common variants in this cohort, with the frequency of 13.9% (6/44 alleles), 13.9% (6/44 alleles) and 12.5% (8/64 alleles), respectively. There was no difference in clinical features and outcomes between patients with PCCA and PCCB variants. Certain variants with high frequencies and homozygotes may be associated with early-onset or late-onset propionic acidemia. Conclusions Although the genotype–phenotype correlation is still unclear, certain variants seemed to be related to early-onset or late-onset propionic acidemia. Our study further delineated the complex clinical manifestations of propionic acidemia and expanded the spectrum of gene variants associated with propionic acidemia. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02271-3.
Collapse
Affiliation(s)
- Yi Liu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.,Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Zhehui Chen
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Hui Dong
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yuan Ding
- Department of Endocrinology and Genetic, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
| | - Ruxuan He
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Lulu Kang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dongxiao Li
- Department of Endocrinology and Genetic, Henan Children's Hospital, Zhengzhou, 450053, China
| | - Ming Shen
- Translational Medicine Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Ying Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yao Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Jinqing Song
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yaping Tian
- Translational Medicine Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yongtong Cao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Desheng Liang
- School of Life Sciences, Central South University, Changsha, 410000, China.
| | - Yanling Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
42
|
Guilder LL, Kronick JB. Organic Acidemias. Pediatr Rev 2022; 43:123-134. [PMID: 35229111 DOI: 10.1542/pir.2020-000562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Laura L Guilder
- Clinical and Metabolic Genetics, Department of Pediatrics, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jonathan B Kronick
- Clinical and Metabolic Genetics, Department of Pediatrics, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Altun I, Kiykim A, Zubarioglu T, Burtecene N, Hopurcuoglu D, Topcu B, Cansever MS, Kiykim E, Cokugras HC, Aktuglu Zeybek AC. Altered immune response in organic acidemia. Pediatr Int 2022; 64:e15082. [PMID: 34861062 DOI: 10.1111/ped.15082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/17/2021] [Accepted: 11/26/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Most patients with organic acidemia suffer from recurrent infections. Although neutropenia has been reported in multiple studies, other components of the immune system have not been evaluated thoroughly. This study was conducted to assess the immune status of patients with organic acidemia (OA). METHODS Thirty-three patients with OA who were followed up in Istanbul University-Cerrahpasa, Cerrahpasa School of Medicine, Nutrition and Metabolism Department and a total of 32 age- and sex-matched healthy controls were enrolled to the study. The demographic and clinical data were recorded retrospectively from patient files. Complete blood counts, immunoglobulins, and lymphocyte immunophenotyping were recorded prospectively in a symptom- (infection-) free period. RESULTS Of the 33 patients enrolled to the study, 21 (88%) were diagnosed with methylmalonic acidemia, 10 (33%) with propionic acidemia, and two (6.6%) with isovaleric acidemia. The mean age of the patients with OA and healthy subjects were 5.89 ± 4.11 years and 5.34 ± 4.36, respectively (P = 0.602). Twenty-nine (88%) of the patients had experienced frequent hospital admission, 13 (39%) were admitted to pediatric intensive care unit, and 18 (55%) suffered from sepsis. Naïve helper T cells and recent thymic emigrants were significantly lower in OAs (P < 0.001). Various defects in humoral immunity have also been documented including memory B cells and immunoglobulins. CONCLUSIONS Patients with OAs may show adaptive immune defects rendering them susceptible to infections. Metabolic reprogramming based on nutritional modifications may be a promising therapeutic option in the future.
Collapse
Affiliation(s)
- Ilayda Altun
- Department of Pediatric of Child Health and Diseases, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| | - Ayca Kiykim
- Department of Pediatric Allergy and Immunology Istanbul, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| | - Tanyel Zubarioglu
- Department of Pediatric Nutrition and Metabolism, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| | - Nihan Burtecene
- Department of Pediatric Allergy and Immunology Istanbul, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| | - Duhan Hopurcuoglu
- Department of Pediatric Nutrition and Metabolism, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| | - Birol Topcu
- Department of Biostatistics, Faculty of Medicine, Tekirdag Namik Kemal University, Tekirdag, Turkey
| | - Mehmet Serif Cansever
- Department of Pediatric Nutrition and Metabolism, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| | - Ertugrul Kiykim
- Department of Pediatric Nutrition and Metabolism, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| | - Haluk Cezmi Cokugras
- Department of Pediatric Allergy and Immunology Istanbul, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| | - Ayse Cigdem Aktuglu Zeybek
- Department of Pediatric Nutrition and Metabolism, Istanbul University-Cerrahpasa Cerrahpasa Medical School, Istanbul, Turkey
| |
Collapse
|
44
|
Sasai H, Goto H, Kawashiri M, Kuwahara T. Long QT as a first sign for propionic acidemia in a 10-year-old girl. Pediatr Int 2022; 64:e14752. [PMID: 34747083 DOI: 10.1111/ped.14752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/09/2021] [Accepted: 04/16/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Hideo Sasai
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan.,Clinical Genetics Center, Gifu University Hospital, Gifu, Japan
| | - Hiroko Goto
- Department of Pediatric Cardiology, Gifu Prefectural General Medical Center, Gifu, Japan
| | - Miwa Kawashiri
- Department of Pediatrics, Japanese Red Cross Takayama Hospital, Takayama, Japan
| | - Takashi Kuwahara
- Department of Pediatric Cardiology, Gifu Prefectural General Medical Center, Gifu, Japan
| |
Collapse
|
45
|
Gonzalez Melo M, Fontana AO, Viertl D, Allenbach G, Prior JO, Rotman S, Feichtinger RG, Mayr JA, Costanzo M, Caterino M, Ruoppolo M, Braissant O, Barbey F, Ballhausen D. A knock-in rat model unravels acute and chronic renal toxicity in glutaric aciduria type I. Mol Genet Metab 2021; 134:287-300. [PMID: 34799272 DOI: 10.1016/j.ymgme.2021.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 01/14/2023]
Abstract
Glutaric aciduria type I (GA-I, OMIM # 231670) is an autosomal recessive inborn error of metabolism caused by deficiency of the mitochondrial enzyme glutaryl-CoA dehydrogenase (GCDH). The principal clinical manifestation in GA-I patients is striatal injury most often triggered by catabolic stress. Early diagnosis by newborn screening programs improved survival and reduced striatal damage in GA-I patients. However, the clinical phenotype is still evolving in the aging patient population. Evaluation of long-term outcome in GA-I patients recently identified glomerular filtration rate (GFR) decline with increasing age. We recently created the first knock-in rat model for GA-I harboring the mutation p.R411W (c.1231 C>T), corresponding to the most frequent GCDH human mutation p.R402W. In this study, we evaluated the effect of an acute metabolic stress in form of high lysine diet (HLD) on young Gcdhki/ki rats. We further studied the chronic effect of GCDH deficiency on kidney function in a longitudinal study on a cohort of Gcdhki/ki rats by repetitive 68Ga-EDTA positron emission tomography (PET) renography, biochemical and histological analyses. In young Gcdhki/ki rats exposed to HLD, we observed a GFR decline and biochemical signs of a tubulopathy. Histological analyses revealed lipophilic vacuoles, thinning of apical brush border membranes and increased numbers of mitochondria in proximal tubular (PT) cells. HLD also altered OXPHOS activities and proteome in kidneys of Gcdhki/ki rats. In the longitudinal cohort, we showed a progressive GFR decline in Gcdhki/ki rats starting at young adult age and a decline of renal clearance. Histopathological analyses in aged Gcdhki/ki rats revealed tubular dilatation, protein accumulation in PT cells and mononuclear infiltrations. These observations confirm that GA-I leads to acute and chronic renal damage. This raises questions on indication for follow-up on kidney function in GA-I patients and possible therapeutic interventions to avoid renal damage.
Collapse
Affiliation(s)
- Mary Gonzalez Melo
- Pediatric Metabolic Unit, Pediatrics, Woman-Mother-Child Department, University of Lausanne and University Hospital of Lausanne, Switzerland.
| | - Andrea Orlando Fontana
- Department of Nuclear Medicine and Molecular Imaging, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland.
| | - David Viertl
- Department of Nuclear Medicine and Molecular Imaging, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland.
| | - Gilles Allenbach
- Department of Nuclear Medicine and Molecular Imaging, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland.
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland.
| | - Samuel Rotman
- Service of Clinical Pathology, University of Lausanne and University Hospital of Lausanne, Switzerland.
| | - René Günther Feichtinger
- Department of Pediatrics, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Johannes Adalbert Mayr
- Department of Pediatrics, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; CEINGE - Biotecnologie, Avanzate s.c.ar.l., 80145 Naples, Italy.
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; CEINGE - Biotecnologie, Avanzate s.c.ar.l., 80145 Naples, Italy.
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; CEINGE - Biotecnologie, Avanzate s.c.ar.l., 80145 Naples, Italy.
| | - Olivier Braissant
- Service of Clinical Chemistry, University of Lausanne and University Hospital of Lausanne, Switzerland.
| | - Frederic Barbey
- Department of Immunology, University of Lausanne and University Hospital of Lausanne, Switzerland.
| | - Diana Ballhausen
- Pediatric Metabolic Unit, Pediatrics, Woman-Mother-Child Department, University of Lausanne and University Hospital of Lausanne, Switzerland.
| |
Collapse
|
46
|
Timmer C, Davids M, Nieuwdorp M, Levels JHM, Langendonk JG, Breederveld M, Ahmadi Mozafari N, Langeveld M. Differences in faecal microbiome composition between adult patients with UCD and PKU and healthy control subjects. Mol Genet Metab Rep 2021; 29:100794. [PMID: 34527515 PMCID: PMC8433284 DOI: 10.1016/j.ymgmr.2021.100794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 01/07/2023] Open
Abstract
Urea cycle disorders (UCDs) are a group of rare inherited metabolic diseases causing hyperammonemic encephalopathy. Despite intensive dietary and pharmacological therapy, outcome is poor in a subset of UCD patients. Reducing ammonia production by changing faecal microbiome in UCD is an attractive treatment approach. We compared faecal microbiome composition of 10 UCD patients, 10 healthy control subjects and 10 phenylketonuria (PKU) patients. PKU patients on a low protein diet were included to differentiate between the effect of a low protein diet and the UCD itself on microbial composition. Participants were asked to collect a faecal sample and to fill out a 24 h dietary journal. DNA was extracted from faecal material, taxonomy was assigned and microbiome data was analyzed, with a focus on microbiota involved in ammonia metabolism.In this study we show an altered faecal microbiome in UCD patients, different from both PKU and healthy controls. UCD patients on dietary and pharmacological treatment had a less diverse faecal microbiome, and the faecal microbiome of PKU patients on a protein restricted diet with amino acid supplementation showed reduced richness compared to healthy adults without a specific diet. The differences in the microbiome composition of UCD patients compared to healthy controls were in part related to lactulose use. Other genomic process encodings involved in ammonia metabolism, did not seem to differ. Since manipulation of the microbiome is possible, this could be a potential treatment modality. We propose as a first next step, to study the impact of these faecal microbiome alterations on metabolic stability. TAKE HOME MESSAGE The faecal microbiome of UCD patients was less diverse compared to PKU patients and even more compared to healthy controls.
Collapse
Key Words
- 16S rRNA, taxonomic marker genes, common to all bacteria
- ADI, Arginine Deimination. Bacteria derive energy from the deamination of arginine to citrulline and citrulline cleavage to ornithine plus carbamoyl phosphate. The latter is then converted into ATP and carbon dioxide, or used for pyrimidine biosynthesis. This route also generates two moles of ammonia (one from the arginine-citrulline conversion, the second from carbamoyl phosphate hydrolysis)
- ARG1d, arginase 1 (ARG1) deficiency
- ASLd, argininosuccinate lyase (ASL) deficiency
- ASSd, argininosuccinate synthetase (ASS) deficiency
- ASV, Amplified Sequence Variant. A specific nucleotide sequence representing a bacterial lineage
- Alpha Diversity, the species diversity in a microbial sample. Used to represent the taxonomic diversities of individual samples
- Ammonium scavengers, agents developed for the reduction of blood ammonia concentration used for the treatment of patients with urea cycle disorders. Sodiumbenzoate and phenylbutyrate are ammonium scavengers
- BCAA, branched chain amino acids: isoleucine, leucine and valine
- DEGs, differentially expressed genes
- DESeq, an R package to analyse count data from high-throughput sequencing assays such as RNA-Seq and test for differential expression
- EAA supplement, essential amino acids supplement containing L-histidine, L-isoleucine, L-leucine, l-lysine, L-methionine, L-phenylalanine, L-threonine, L-tryptofaan and L-valine with optional L-cystine and L-tyrosine added (depending on what product is used)
- FPD, Faiths Phylogenetic Diversity, alpha diversity metric accounting for genetic diversity
- Faecal
- Genus, a taxonomic rank
- Gut
- Hyperammonemia
- Metagenome, microbiome collective genome
- Microbiome
- OTCd, ornithine transcarbamylase deficiency
- PCoA, Principal Coordinate Analysis. PCoA is aimed at graphically representing a resemblance matrix between p elements (individuals, variables, objects, among others). By using PCoA we can visualize individual and/or group differences. Individual differences can be used to show outliers
- PFAA, precursor free amino acid supplement, in this case phenylalanine free
- PKU, Phenylketonuria
- Phenylketonuria
- Proteolytic capacity, the capacity to break proteins down into smaller polypeptides or amino acids. In this study: enzymes involved in protein degradation
- RT-qPCR, real-time quantitative polymerase chain reaction
- Sodium BPA, sodium phenylbutyrate
- UCD, urea cycle defect
- Urea cycle defect
Collapse
Affiliation(s)
- C Timmer
- Department of Dietetics and Nutritional science and Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - M Davids
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - M Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - J H M Levels
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - J G Langendonk
- Department of Dietetics and Department of Internal Medicine, Center of Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Erasmus MC, Rotterdam, the Netherlands
| | - M Breederveld
- Department of Dietetics and Department of Internal Medicine, Center of Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Erasmus MC, Rotterdam, the Netherlands
| | - N Ahmadi Mozafari
- Department of Dietetics and Department of Internal Medicine, Center of Lysosomal and Metabolic Diseases, Erasmus University Medical Center, Erasmus MC, Rotterdam, the Netherlands
| | - M Langeveld
- Department of Dietetics and Nutritional science and Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| |
Collapse
|
47
|
Kido J, Matsumoto S, Häberle J, Inomata Y, Kasahara M, Sakamoto S, Horikawa R, Tanemura A, Okajima H, Suzuki T, Nakamura K. Role of liver transplantation in urea cycle disorders: Report from a nationwide study in Japan. J Inherit Metab Dis 2021; 44:1311-1322. [PMID: 34232532 DOI: 10.1002/jimd.12415] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022]
Abstract
Urea cycle disorders (UCDs) are inherited metabolic diseases causing hyperammonemia by defects in urea cycle enzymes or transporters. Liver transplantation (LT) currently is the only curative treatment option until novel therapies become available. We performed a nationwide questionnaire-based study between January 2000 and March 2018 to investigate the effect of LT in patients with UCDs in Japan. A total of 231 patients with UCDs were enrolled in this study. Of them, a total of 78 patients with UCDs (30 male and 16 female ornithine transcarbamylase deficiency (OTCD), 21 carbamoyl phosphate synthetase 1 deficiency (CPSD), 10 argininosuccinate synthetase deficiency (ASSD) and 1 arginase 1 deficiency (ARGD)) had undergone LT. Concerning the maximum blood ammonia levels at the onset time in the transplanted male OTCD (N = 28), female OTCD (N = 15), CPSD (N = 21) and ASSD (N = 10), those were median 634 (IQR: 277-1172), 268 (211-352), 806 (535-1382), and 628 (425-957) μmol/L, respectively. The maximum blood ammonia levels in female OTCD were thus significantly lower than in the other UCDs (all P < .01). LT was effective for long-term survival, prevented recurrent hyperammonemia attack, and lowered baseline blood ammonia levels in patients with UCDs. LT had limited effect for ameliorating neurodevelopmental outcome in patients with severe disease because hyperammonemia at the onset time already had a significant impact on the brain. Patients with ASSD may be more likely to survive without cognitive impairment by receiving early LT despite severe neonatal hyperammonemia ≥ 360 μmol/L. In patients with neonatal onset OTCD or CPSD, there may be additional factors with adverse effects on the brain that are not improved by LT.
Collapse
Affiliation(s)
- Jun Kido
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shirou Matsumoto
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Johannes Häberle
- University Children's Hospital Zurich and Children's Research Centre, Zurich, Switzerland
| | - Yukihiro Inomata
- Department of Transplantation and Pediatric Surgery, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Labor Welfare Corporation, Kumamoto Rosai Hospital, Yatsushiro, Japan
| | - Mureo Kasahara
- Organ Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | - Seisuke Sakamoto
- Organ Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | - Reiko Horikawa
- Division of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo, Japan
| | - Akihiro Tanemura
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hideaki Okajima
- Department of Pediatric Surgery, Kanazawa Medical University, Kanazawa, Japan
| | - Tatsuya Suzuki
- Department of Pediatric Surgery, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
48
|
Kılavuz S, Bulut D, Kor D, Şeker-Yılmaz B, Özcan N, Incecik F, Onan B, Ceylaner G, Önenli-Mungan N. The outcome of 41 Late-Diagnosed Turkish GA-1 Patients: A Candidate for the Turkish NBS. Neuropediatrics 2021; 52:358-369. [PMID: 33578440 DOI: 10.1055/s-0040-1722691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Glutaric aciduria type 1(GA-1) is an inherited cerebral organic aciduria. Untreated patients with GA-1 have a risk of acute encephalopathic crises during the first 6 years of life. In so far as GA-1 desperately does not exist in Turkish newborn screening (NBS) program, most patients in our study were late-diagnosed. METHOD This study included 41 patients diagnosed with acylcarnitine profile, urinary organic acids, mutation analyses in the symptomatic period. We presented with clinical, neuroradiological, and molecular data of our 41 patients. RESULTS The mean age at diagnosis was 14.8 ± 13.9 (15 days to 72 months) and, high blood glutaconic acid, glutarylcarnitine and urinary glutaric acid (GA) levels in 41 patients were revealed. Seventeen different mutations in the glutaryl-CoA dehydrogenase gene were identified, five of which were novel. The patients, most of whom were late-diagnosed, had a poor neurological outcome. Treatment strategies made a little improvement in dystonia and the frequency of encephalopathic attacks. CONCLUSION All GA-1 patients in our study were severely affected since they were late-diagnosed, while others show that GA-1 is a treatable metabolic disorder if it is diagnosed with NBS. This study provides an essential perspective of the severe impact on GA-1 patients unless it is diagnosed with NBS. We immediately advocate GA-1 to be included in the Turkish NBS.
Collapse
Affiliation(s)
- Sebile Kılavuz
- Division of Pediatric Nutrition and Metabolism, Department of Pediatrics, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Derya Bulut
- Division of Pediatric Nutrition and Metabolism, Department of Pediatrics, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Deniz Kor
- Division of Pediatric Nutrition and Metabolism, Department of Pediatrics, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Berna Şeker-Yılmaz
- Division of Pediatric Nutrition and Metabolism, Department of Pediatrics, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Neslihan Özcan
- Division of Pediatric Neurology, Department of Pediatrics, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Faruk Incecik
- Division of Pediatric Neurology, Department of Pediatrics, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Bilen Onan
- Department of Radiology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Gülay Ceylaner
- Department of Medical Genetics, Intergen Genetics Centre, Ankara, Turkey
| | - Neslihan Önenli-Mungan
- Division of Pediatric Nutrition and Metabolism, Department of Pediatrics, Çukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
49
|
Guerreiro G, Faverzani J, Moura AP, Volfart V, Gome Dos Reis B, Sitta A, Gonzalez EA, de Lima Rosa G, Coitinho AS, Baldo G, Wajner M, Vargas CR. Protective effects of L-carnitine on behavioral alterations and neuroinflammation in striatum of glutaryl-COA dehydrogenase deficient mice. Arch Biochem Biophys 2021; 709:108970. [PMID: 34181873 DOI: 10.1016/j.abb.2021.108970] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/06/2021] [Accepted: 06/15/2021] [Indexed: 12/18/2022]
Abstract
Glutaric acidemia type 1 (GA1) is caused by glutaryl-CoA dehydrogenase deficiency that leads to a blockage in the metabolic route of the amino acids lysine and tryptophan and subsequent accumulation of glutaric acid (GA), 3-hydroxyglutaric acids and glutarylcarnitine (C5DC). Patients predominantly manifest neurological symptoms, associated with acute striatal degeneration, as well as progressive cortical and striatum injury whose pathogenesis is not yet fully established. Current treatment includes protein/lysine restriction and l-carnitine supplementation of (L-car). The aim of this work was to evaluate behavior parameters and pro-inflammatory factors (cytokines IL-1β, TNF-α and cathepsin-D levels), as well as the anti-inflammatory cytokine IL10 in striatum of knockout mice (Gcdh-/-) and wild type (WT) mice submitted to a normal or a high Lys diet. The potential protective effects of L-car treatment on these parameters were also evaluated. Gcdh-/- mice showed behavioral changes, including lower motor activity (decreased number of crossings) and exploratory activity (reduced number of rearings). Also, Gcdh-/- mice had significantly higher concentrations of glutarylcarnitine (C5DC) in blood and cathepsin-D (CATD), interleukin IL-1β and tumor factor necrosis alpha (TNF-α) in striatum than WT mice. Noteworthy, L-car treatment prevented most behavioral alterations, normalized CATD levels and attenuated IL-1β levels in striatum of Gcdh-/- mice. Finally, IL-1β was positively correlated with CATD and C5DC levels and L-car was negatively correlated with CATD. Our results demonstrate behavioral changes and a pro-inflammatory status in striatum of the animal model of GA1 and, most importantly, L-car showed important protective effects on these alterations.
Collapse
Affiliation(s)
- Gilian Guerreiro
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil; Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035- 903, Porto Alegre, RS, Brazil.
| | - Jéssica Faverzani
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil; Programa de Pós-Graduação Em Ciências Farmacêuticas, UFRGS, Av. Ipiranga, 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Alana Pimentel Moura
- Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035- 903, Porto Alegre, RS, Brazil
| | - Vitoria Volfart
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Bianca Gome Dos Reis
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil
| | - Angela Sitta
- Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035- 903, Porto Alegre, RS, Brazil
| | - Esteban Alberto Gonzalez
- Programa de Pós-Graduação Em Fisiologia, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil; Departamento de Microbiologia, Imunologia e Parasitologia, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Gabriel de Lima Rosa
- Programa de Pós-Graduação Em Fisiologia, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil; Departamento de Microbiologia, Imunologia e Parasitologia, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Adriana Simon Coitinho
- Programa de Pós-Graduação Em Fisiologia, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil; Departamento de Microbiologia, Imunologia e Parasitologia, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Guilherme Baldo
- Programa de Pós-Graduação Em Fisiologia, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil; Departamento de Microbiologia, Imunologia e Parasitologia, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035- 903, Porto Alegre, RS, Brazil; Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, UFRGS, Rua Ramiro Barcelos, 2600, 90035 000, Porto Alegre, RS, Brazil
| | - Carmen Regla Vargas
- Faculdade de Farmácia, UFRGS, Av. Ipiranga 2752, 90610-000, Porto Alegre, RS, Brazil; Serviço de Genética Médica, HCPA, UFRGS, Rua Ramiro Barcelos, 2350, 90035- 903, Porto Alegre, RS, Brazil; Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, UFRGS, Rua Ramiro Barcelos, 2600, 90035 000, Porto Alegre, RS, Brazil; Programa de Pós-Graduação Em Ciências Farmacêuticas, UFRGS, Av. Ipiranga, 2752, 90610-000, Porto Alegre, RS, Brazil.
| |
Collapse
|
50
|
Luciani A, Denley MCS, Govers LP, Sorrentino V, Froese DS. Mitochondrial disease, mitophagy, and cellular distress in methylmalonic acidemia. Cell Mol Life Sci 2021; 78:6851-6867. [PMID: 34524466 PMCID: PMC8558192 DOI: 10.1007/s00018-021-03934-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/18/2021] [Accepted: 08/30/2021] [Indexed: 01/09/2023]
Abstract
Mitochondria—the intracellular powerhouse in which nutrients are converted into energy in the form of ATP or heat—are highly dynamic, double-membraned organelles that harness a plethora of cellular functions that sustain energy metabolism and homeostasis. Exciting new discoveries now indicate that the maintenance of this ever changing and functionally pleiotropic organelle is particularly relevant in terminally differentiated cells that are highly dependent on aerobic metabolism. Given the central role in maintaining metabolic and physiological homeostasis, dysregulation of the mitochondrial network might therefore confer a potentially devastating vulnerability to high-energy requiring cell types, contributing to a broad variety of hereditary and acquired diseases. In this Review, we highlight the biological functions of mitochondria-localized enzymes from the perspective of understanding—and potentially reversing—the pathophysiology of inherited disorders affecting the homeostasis of the mitochondrial network and cellular metabolism. Using methylmalonic acidemia as a paradigm of complex mitochondrial dysfunction, we discuss how mitochondrial directed-signaling circuitries govern the homeostasis and physiology of specialized cell types and how these may be disturbed in disease. This Review also provides a critical analysis of affected tissues, potential molecular mechanisms, and novel cellular and animal models of methylmalonic acidemia which are being used to develop new therapeutic options for this disease. These insights might ultimately lead to new therapeutics, not only for methylmalonic acidemia, but also for other currently intractable mitochondrial diseases, potentially transforming our ability to regulate homeostasis and health.
Collapse
Affiliation(s)
- Alessandro Luciani
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8032, Zurich, Switzerland.
| | - Matthew C S Denley
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, 8032, Zurich, Switzerland
| | - Larissa P Govers
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8032, Zurich, Switzerland
| | - Vincenzo Sorrentino
- Department of Musculo-Skeletal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1015, Lausanne, Switzerland.
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, 8032, Zurich, Switzerland.
| |
Collapse
|