Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mie Ichikawa is active.

Publication


Featured researches published by Mie Ichikawa.


The New England Journal of Medicine | 2014

Multiple phenotypes in phosphoglucomutase 1 deficiency

Laura C. Tegtmeyer; Stephan Rust; Monique van Scherpenzeel; Bobby G. Ng; Marie-Estelle Losfeld; Sharita Timal; Kimiyo Raymond; Ping He; Mie Ichikawa; Joris A. Veltman; Karin Huijben; Yoon S. Shin; Vandana Sharma; Maciej Adamowicz; Martin Lammens; Janine Reunert; Anika Witten; Esther Schrapers; Gert Matthijs; Jaak Jaeken; Daisy Rymen; Tanya Stojkovic; P. Laforêt; François Petit; Olivier Aumaître; Elżbieta Czarnowska; Monique Piraud; Teodor Podskarbi; Charles A. Stanley; Reuben Matalon

BACKGROUND Congenital disorders of glycosylation are genetic syndromes that result in impaired glycoprotein production. We evaluated patients who had a novel recessive disorder of glycosylation, with a range of clinical manifestations that included hepatopathy, bifid uvula, malignant hyperthermia, hypogonadotropic hypogonadism, growth retardation, hypoglycemia, myopathy, dilated cardiomyopathy, and cardiac arrest. METHODS Homozygosity mapping followed by whole-exome sequencing was used to identify a mutation in the gene for phosphoglucomutase 1 (PGM1) in two siblings. Sequencing identified additional mutations in 15 other families. Phosphoglucomutase 1 enzyme activity was assayed on cell extracts. Analyses of glycosylation efficiency and quantitative studies of sugar metabolites were performed. Galactose supplementation in fibroblast cultures and dietary supplementation in the patients were studied to determine the effect on glycosylation. RESULTS Phosphoglucomutase 1 enzyme activity was markedly diminished in all patients. Mass spectrometry of transferrin showed a loss of complete N-glycans and the presence of truncated glycans lacking galactose. Fibroblasts supplemented with galactose showed restoration of protein glycosylation and no evidence of glycogen accumulation. Dietary supplementation with galactose in six patients resulted in changes suggestive of clinical improvement. A new screening test showed good discrimination between patients and controls. CONCLUSIONS Phosphoglucomutase 1 deficiency, previously identified as a glycogenosis, is also a congenital disorder of glycosylation. Supplementation with galactose leads to biochemical improvement in indexes of glycosylation in cells and patients, and supplementation with complex carbohydrates stabilizes blood glucose. A new screening test has been developed but has not yet been validated. (Funded by the Netherlands Organization for Scientific Research and others.).


Journal of Biological Chemistry | 2011

Phosphomannose Isomerase Inhibitors Improve N-Glycosylation in Selected Phosphomannomutase-deficient Fibroblasts

Vandana Sharma; Mie Ichikawa; Ping He; David A. Scott; Yalda Bravo; Russell Dahl; Bobby G. Ng; Nicholas D. P. Cosford; Hudson H. Freeze

Background: Some glycosylation-deficient patients do not make enough mannose-1-P from mannose-6-P. Preventing mannose-6-P catabolism might improve glycosylation in cells and patients. Results: Inhibitors of mannose-6-P catabolism direct more mannose toward N-glycosylation in many cell types. Conclusion: Increasing exogenous mannose and blocking mannose-6-P catabolism can improve glycosylation in some glycosylation-deficient cells. Significance: Increasing activated mannose flux might benefit some glycosylation-deficient patients. Congenital disorders of glycosylation (CDG) are rare genetic disorders due to impaired glycosylation. The patients with subtypes CDG-Ia and CDG-Ib have mutations in the genes encoding phosphomannomutase 2 (PMM2) and phosphomannose isomerase (MPI or PMI), respectively. PMM2 (mannose 6-phosphate → mannose 1-phosphate) and MPI (mannose 6-phosphate ⇔ fructose 6-phosphate) deficiencies reduce the metabolic flux of mannose 6-phosphate (Man-6-P) into glycosylation, resulting in unoccupied N-glycosylation sites. Both PMM2 and MPI compete for the same substrate, Man-6-P. Daily mannose doses reverse most of the symptoms of MPI-deficient CDG-Ib patients. However, CDG-Ia patients do not benefit from mannose supplementation because >95% Man-6-P is catabolized by MPI. We hypothesized that inhibiting MPI enzymatic activity would provide more Man-6-P for glycosylation and possibly benefit CDG-Ia patients with residual PMM2 activity. Here we show that MLS0315771, a potent MPI inhibitor from the benzoisothiazolone series, diverts Man-6-P toward glycosylation in various cell lines including fibroblasts from CDG-Ia patients and improves N-glycosylation. Finally, we show that MLS0315771 increases mannose metabolic flux toward glycosylation in zebrafish embryos.


Biochemical and Biophysical Research Communications | 2014

Mannose metabolism: more than meets the eye.

Vandana Sharma; Mie Ichikawa; Hudson H. Freeze

Mannose is a simple sugar with a complex life. It is a welcome therapy for genetic and acquired human diseases, but it kills honeybees and blinds baby mice. It could cause diabetic complications. Mannose chemistry, metabolism, and metabolomics in cells, tissues and mammals can help explain these multiple systemic effects. Mannose has good, bad or ugly outcomes depending on its steady state levels and metabolic flux. This review describes the role of mannose at cellular level and its impact on organisms.


Journal of Medicinal Chemistry | 2011

Potent, Selective, and Orally Available Benzoisothiazolone Phosphomannose Isomerase Inhibitors as Probes for Congenital Disorder of Glycosylation Ia

Russell Dahl; Yalda Bravo; Vandana Sharma; Mie Ichikawa; Raveendra-Panickar Dhanya; Michael Hedrick; Brock Brown; Justin Rascon; Michael Vicchiarelli; Arianna Mangravita-Novo; Li Yang; Derek Stonich; Ying Su; Layton H. Smith; Eduard Sergienko; Hudson H. Freeze; Nicholas D. P. Cosford

We report the discovery and validation of a series of benzoisothiazolones as potent inhibitors of phosphomannose isomerase (PMI), an enzyme that converts mannose-6-phosphate (Man-6-P) into fructose-6-phosphate (Fru-6-P) and, more importantly, competes with phosphomannomutase 2 (PMM2) for Man-6-P, diverting this substrate from critical protein glycosylation events. In congenital disorder of glycosylation type Ia, PMM2 activity is compromised; thus, PMI inhibition is a potential strategy for the development of therapeutics. High-throughput screening (HTS) and subsequent chemical optimization led to the identification of a novel class of benzoisothiazolones as potent PMI inhibitors having little or no PMM2 inhibition. Two complementary synthetic routes were developed, enabling the critical structural requirements for activity to be determined, and the compounds were subsequently profiled in biochemical and cellular assays to assess efficacy. The most promising compounds were also profiled for bioavailability parameters, including metabolic stability, plasma stability, and permeability. The pharmacokinetic profile of a representative of this series (compound 19; ML089) was also assessed, demonstrating the potential of this series for in vivo efficacy when dosed orally in disease models.


Human Molecular Genetics | 2015

Biallelic mutations in CAD, impair de novo pyrimidine biosynthesis and decrease glycosylation precursors

Bobby G. Ng; Lynne A. Wolfe; Mie Ichikawa; Thomas C. Markello; Miao He; Cynthia J. Tifft; William A. Gahl; Hudson H. Freeze

In mitochondria, carbamoyl-phosphate synthetase 1 activity produces carbamoyl phosphate for urea synthesis, and deficiency results in hyperammonemia. Cytoplasmic carbamoyl-phosphate synthetase 2, however, is part of a tri-functional enzyme encoded by CAD; no human disease has been attributed to this gene. The tri-functional enzyme contains carbamoyl-phosphate synthetase 2 (CPS2), aspartate transcarbamylase (ATCase) and dihydroorotase (DHOase) activities, which comprise the first three of six reactions required for de novo pyrimidine biosynthesis. Here we characterize an individual who is compound heterozygous for mutations in different domains of CAD. One mutation, c.1843-1G>A, results in an in-frame deletion of exon 13. The other, c.6071G>A, causes a missense mutation (p.Arg2024Gln) in a highly conserved residue that is essential for carbamoyl-phosphate binding. Metabolic flux studies showed impaired aspartate incorporation into RNA and DNA through the de novo synthesis pathway. In addition, CTP, UTP and nearly all UDP-activated sugars that serve as donors for glycosylation were decreased. Uridine supplementation rescued these abnormalities, suggesting a potential therapy for this new glycosylation disorder.


The FASEB Journal | 2014

Mannose supplements induce embryonic lethality and blindness in phosphomannose isomerase hypomorphic mice

Vandana Sharma; Jonamani Nayak; Charles DeRossi; Adriana Charbono; Mie Ichikawa; Bobby G. Ng; Erika Grajales-Esquivel; Anand Srivastava; Ling Wang; Ping He; David A. Scott; Joseph Russell; Emily Contreras; Cherise M. Guess; Stan Krajewski; Katia Del Rio-Tsonis; Hudson H. Freeze

Patients with congenital disorder of glycosylation (CDG), type Ib (MPI‐CDG or CDG‐Ib) have mutations in phosphomannose isomerase (MPI) that impair glycosylation and lead to stunted growth, liver dysfunction, coagulopathy, hypoglycemia, and intestinal abnormalities. Mannose supplements correct hypo‐glycosylation and most symptoms by providing man‐nose‐6‐P (Man‐6‐P) via hexokinase. We generated viable Mpi hypomorphic mice with residual enzymatic activity comparable to that of patients, but surprisingly, these mice appeared completely normal except for modest (~15%) embryonic lethality. To overcome this lethality, pregnant dams were provided 1–2% mannose in their drinking water. However, mannose further reduced litter size and survival to weaning by 40 and 66%, respectively. Moreover, ~50% of survivors developed eye defects beginning around midgestation. Mannose started at birth also led to eye defects but had no effect when started after eye development was complete. Man‐6‐P and related metabolites accumulated in the affected adult eye and in developing embryos and placentas. Our results demonstrate that disturbing mannose metabolic flux in mice, especially during embryonic development, induces a highly specific, unanticipated pathological state. It is unknown whether mannose is harmful to human fetuses during gestation; however, mothers who are at risk for having MPI‐CDG children and who consume mannose during pregnancy hoping to benefit an affected fetus in utero should be cautious.—Sharma, V., Nayak, J., DeRossi, C., Charbono, A., Ichikawa, M., Ng, B. G., Grajales‐Esquivel, E., Srivastava, A., Wang, L., He, P., Scott, D. A., Russell, J., Contreras, E., Guess, C. M., Krajewski, S., Del Rio‐Tsonis, K., Freeze, H. H. Mannose supplements induce embryonic lethality and blindness in phosphomannose isomerase hypomorphic mice. FASEB J. 28, 1854–1869 (2014). www.fasebj.org


The Journal of Allergy and Clinical Immunology | 2014

Autosomal recessive phosphoglucomutase 3 (PGM3) mutations link glycosylation defects to atopy, immune deficiency, autoimmunity, and neurocognitive impairment.

Yu Zhang; Xiaomin Yu; Mie Ichikawa; Jonathan J. Lyons; Shrimati Datta; Ian T. Lamborn; Huie Jing; Emily S. Kim; Matthew Biancalana; Lynne A. Wolfe; Thomas DiMaggio; Helen F. Matthews; Sarah M. Kranick; Kelly D. Stone; Steven M. Holland; Daniel S. Reich; Jason D. Hughes; Huseyin Mehmet; Joshua McElwee; Alexandra F. Freeman; Hudson H. Freeze; Helen C. Su; Joshua D. Milner


/data/revues/00916749/unassign/S0091674914002620/ | 2014

Autosomal recessive phosphoglucomutase 3 ( PGM3 ) mutations link glycosylation defects to atopy, immune deficiency, autoimmunity, and neurocognitive impairment

Yu Zhang; Xiaomin Yu; Mie Ichikawa; Jonathan J. Lyons; Shrimati Datta; Ian T. Lamborn; Huie Jing; Emily S. Kim; Matthew Biancalana; Lynne A. Wolfe; Thomas DiMaggio; Helen F. Matthews; Sarah M. Kranick; Kelly D. Stone; Steven M. Holland; Daniel S. Reich; Jason D. Hughes; Huseyin Mehmet; Joshua McElwee; Alexandra F. Freeman; Hudson H. Freeze; Helen C. Su; Joshua D. Milner


Glycobiology | 2005

HYDROPHOBIC MAN-1-P DERIVATIVES CORRECT ABNORMAL GLYCOSYLATION IN TYPE I CONGENITAL DISORDER OF GLYCOSYLATION FIBROBLASTS

Erik A. Eklund; Nabyl Merbouh; Mie Ichikawa; Atsushi Nishikawa; Jessica M. Clima; James A. Dorman; Thomas Norberg; Hudson H. Freeze


Journal of Biological Chemistry | 2014

The Metabolic Origins of Mannose in Glycoproteins

Mie Ichikawa; David A. Scott; Marie-Estelle Losfeld; Hudson H. Freeze

Collaboration


Dive into the Mie Ichikawa's collaboration.

Top Co-Authors

Avatar

Lynne A. Wolfe

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alexandra F. Freeman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Daniel S. Reich

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Emily S. Kim

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Helen C. Su

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Helen F. Matthews

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Huie Jing

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ian T. Lamborn

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge