Network


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

Hotspot


Dive into the research topics where angtao Li is active.

Publication


Featured researches published by angtao Li.


Cell | 1994

The FET3 gene of S. cerevisiae encodes a multicopper oxidase required for ferrous iron uptake.

Candice C. Askwith; David Eide; Anthony Van Ho; Philip S. Bernard; Liangtao Li; Sandra Davis-Kaplan; David Sipe; Jerry Kaplan

S. cerevisiae accumulate iron by a process requiring a ferrireductase and a ferrous transporter. We have isolated a mutant, fet3, defective for high affinity Fe(II) uptake. The wild-type FET3 gene was isolated by complementation of the mutant defect. Sequence analysis of the gene revealed the presence of an open reading frame coding for a protein with strong similarity to the family of blue multicopper oxidoreductases. Consistent with the role of copper in iron transport, growth of wild-type cells in copper-deficient media resulted in decreased ferrous iron transport. Addition of copper, but not other transition metals (manganese or zinc), to the assay media resulted in the recovery of Fe(II) transporter activity. We suggest that the catalytic activity of the Fet3 protein is required for cellular iron accumulation.


Science | 2006

Localization of Iron in Arabidopsis Seed Requires the Vacuolar Membrane Transporter VIT1

Sun A. Kim; Tracy Punshon; Antonio Lanzirotti; Liangtao Li; Jose M. Alonso; Joseph R. Ecker; Jerry Kaplan; Mary Lou Guerinot

Iron deficiency is a major human nutritional problem wherever plant-based diets are common. Using synchrotron x-ray fluorescence microtomography to directly visualize iron in Arabidopsis seeds, we show that iron is localized primarily to the provascular strands of the embryo. This localization is completely abolished when the vacuolar iron uptake transporter VIT1 is disrupted. Vacuolar iron storage is also critical for seedling development because vit1-1 seedlings grow poorly when iron is limiting. We have uncovered a fundamental aspect of seed biology that will ultimately aid the development of nutrient-rich seed, benefiting both human health and agricultural productivity.


Nature | 2006

Mitoferrin is essential for erythroid iron assimilation

George C. Shaw; John J. Cope; Liangtao Li; Kenneth Corson; Candace Hersey; Gabriele E. Ackermann; Babette Gwynn; Amy J. Lambert; Rebecca A. Wingert; David Traver; Nikolaus S. Trede; Bruce Barut; Yi Zhou; Emmanuel Minet; Adriana Donovan; Alison Brownlie; Rena Balzan; Mitchell J. Weiss; Luanne L. Peters; Jerry Kaplan; Leonard I. Zon; Barry H. Paw

Iron has a fundamental role in many metabolic processes, including electron transport, deoxyribonucleotide synthesis, oxygen transport and many essential redox reactions involving haemoproteins and Fe–S cluster proteins. Defective iron homeostasis results in either iron deficiency or iron overload. Precise regulation of iron transport in mitochondria is essential for haem biosynthesis, haemoglobin production and Fe–S cluster protein assembly during red cell development. Here we describe a zebrafish mutant, frascati (frs), that shows profound hypochromic anaemia and erythroid maturation arrest owing to defects in mitochondrial iron uptake. Through positional cloning, we show that the gene mutated in the frs mutant is a member of the vertebrate mitochondrial solute carrier family (SLC25) that we call mitoferrin (mfrn). mfrn is highly expressed in fetal and adult haematopoietic tissues of zebrafish and mouse. Erythroblasts generated from murine embryonic stem cells null for Mfrn (also known as Slc25a37) show maturation arrest with severely impaired incorporation of 55Fe into haem. Disruption of the yeast mfrn orthologues, MRS3 and MRS4, causes defects in iron metabolism and mitochondrial Fe–S cluster biogenesis. Murine Mfrn rescues the defects in frs zebrafish, and zebrafish mfrn complements the yeast mutant, indicating that the function of the gene may be highly conserved. Our data show that mfrn functions as the principal mitochondrial iron importer essential for haem biosynthesis in vertebrate erythroblasts.


The EMBO Journal | 2006

Ferroportin-mediated mobilization of ferritin iron precedes ferritin degradation by the proteasome

Ivana De Domenico; Michael B. Vaughn; Liangtao Li; Dustin Bagley; Giovanni Musci; Diane M. Ward; Jerry Kaplan

Ferritin is a cytosolic molecule comprised of subunits that self‐assemble into a nanocage capable of containing up to 4500 iron atoms. Iron stored within ferritin can be mobilized for use within cells or exported from cells. Expression of ferroportin (Fpn) results in export of cytosolic iron and ferritin degradation. Fpn‐mediated iron loss from ferritin occurs in the cytosol and precedes ferritin degradation by the proteasome. Depletion of ferritin iron induces the monoubiquitination of ferritin subunits. Ubiquitination is not required for iron release but is required for disassembly of ferritin nanocages, which is followed by degradation of ferritin by the proteasome. Specific mammalian machinery is not required to extract iron from ferritin. Iron can be removed from ferritin when ferritin is expressed in Saccharomyces cerevisiae, which does not have endogenous ferritin. Expressed ferritin is monoubiquitinated and degraded by the proteasome. Exposure of ubiquitination defective mammalian cells to the iron chelator desferrioxamine leads to degradation of ferritin in the lysosome, which can be prevented by inhibitors of autophagy. Thus, ferritin degradation can occur through two different mechanisms.


Journal of Biological Chemistry | 2008

Identification of FRA1 and FRA2 as Genes Involved in Regulating the Yeast Iron Regulon in Response to Decreased Mitochondrial Iron-Sulfur Cluster Synthesis

Attila Kumánovics; Opal S. Chen; Liangtao Li; Dustin Bagley; Erika M. Adkins; Huilan Lin; Nin N. Dingra; Caryn E. Outten; Greg Keller; Dennis R. Winge; Diane M. Ward; Jerry Kaplan

The nature of the connection between mitochondrial Fe-S cluster synthesis and the iron-sensitive transcription factor Aft1 in regulating the expression of the iron transport system in Saccharomyces cerevisiae is not known. Using a genetic screen, we identified two novel cytosolic proteins, Fra1 and Fra2, that are part of a complex that interprets the signal derived from mitochondrial Fe-S synthesis. We found that mutations in FRA1 (YLL029W) and FRA2 (YGL220W) led to an increase in transcription of the iron regulon. In cells incubated in high iron medium, deletion of either FRA gene results in the translocation of the low iron-sensing transcription factor Aft1 into the nucleus, where it occupies the FET3 promoter. Deletion of either FRA gene has the same effect on transcription as deletion of both genes and is not additive with activation of the iron regulon due to loss of mitochondrial Fe-S cluster synthesis. These observations suggest that the FRA proteins are in the same signal transduction pathway as Fe-S cluster synthesis. We show that Fra1 and Fra2 interact in the cytosol in an iron-independent fashion. The Fra1-Fra2 complex binds to Grx3 and Grx4, two cytosolic monothiol glutaredoxins, in an iron-independent fashion. These results show that the Fra-Grx complex is an intermediate between the production of mitochondrial Fe-S clusters and transcription of the iron regulon.


The Plant Cell | 2009

The Ferroportin Metal Efflux Proteins Function in Iron and Cobalt Homeostasis in Arabidopsis

Joe Morrissey; Ivan Baxter; Joohyun Lee; Liangtao Li; Brett Lahner; Natasha Grotz; Jerry Kaplan; David E. Salt; Mary Lou Guerinot

Relatively little is known about how metals such as iron are effluxed from cells, a necessary step for transport from the root to the shoot. Ferroportin (FPN) is the sole iron efflux transporter identified to date in animals, and there are two closely related orthologs in Arabidopsis thaliana, IRON REGULATED1 (IREG1/FPN1) and IREG2/FPN2. FPN1 localizes to the plasma membrane and is expressed in the stele, suggesting a role in vascular loading; FPN2 localizes to the vacuole and is expressed in the two outermost layers of the root in response to iron deficiency, suggesting a role in buffering metal influx. Consistent with these roles, fpn2 has a diminished iron deficiency response, whereas fpn1 fpn2 has an elevated iron deficiency response. Ferroportins also play a role in cobalt homeostasis; a survey of Arabidopsis accessions for ionomic phenotypes showed that truncation of FPN2 results in elevated shoot cobalt levels and leads to increased sensitivity to the metal. Conversely, loss of FPN1 abolishes shoot cobalt accumulation, even in the cobalt accumulating mutant frd3. Consequently, in the fpn1 fpn2 double mutant, cobalt cannot move to the shoot via FPN1 and is not sequestered in the root vacuoles via FPN2; instead, cobalt likely accumulates in the root cytoplasm causing fpn1 fpn2 to be even more sensitive to cobalt than fpn2 mutants.


Journal of Biological Chemistry | 1998

Defects in the yeast high affinity iron transport system result in increased metal sensitivity because of the increased expression of transporters with a broad transition metal specificity.

Liangtao Li; Jerry Kaplan

Yeast with defects in vacuolar pH show increased sensitivity to high concentrations of transition metals. This sensitivity has been presumed to result from defective metal storage. We demonstrate that mutations that result in a defective high affinity iron transport system, such as a deletion in the surface ferroxidaseFET3, also result in increased metal sensitivity independent of vacuolar function. Multiple copies of transition metal transporter resistance genes, such as COT1 orZRC1, do not reduce the metal sensitivity offet3 mutations. Increased metal sensitivity is because of an increased cellular accumulation of transition metals resulting from the increased activity of low affinity iron transporters, such asFET4, that mediates the transport of other transition metals. In cells lacking a high affinity iron transport system, the increased transition metal uptake can be prevented by increased extracellular iron. These results suggest that vacuolar function may not be required for transition metal sequestration.


FEBS Letters | 2002

Frataxin knockin mouse

Carlos J. Miranda; Manuela Santos; Keiichi Ohshima; Julie Smith; Liangtao Li; Michaeline Bunting; Mireille Cossée; Michael Koenig; Jorge Sequeiros; Jerry Kaplan; Massimo Pandolfo

Friedreich ataxia is the consequence of frataxin deficiency, most often caused by a GAA repeat expansion in intron 1 of the corresponding gene. Frataxin is a mitochondrial protein involved in iron homeostasis. As an attempt to generate a mouse model of the disease, we introduced a (GAA)230 repeat within the mouse frataxin gene by homologous recombination. GAA repeat knockin mice were crossed with frataxin knockout mice to obtain double heterozygous mice expressing 25–36% of wild‐type frataxin levels. These mice were viable and did not develop anomalies of motor coordination, iron metabolism or response to iron loading. Repeats were meiotically and mitotically stable.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Abcb10 physically interacts with mitoferrin-1 (Slc25a37) to enhance its stability and function in the erythroid mitochondria

Wen Chen; Prasad N. Paradkar; Liangtao Li; Eric L. Pierce; Nathaniel B. Langer; Naoko Takahashi-Makise; Brigham B. Hyde; Orian S. Shirihai; Diane M. Ward; Jerry Kaplan; Barry H. Paw

Mitoferrin-1 (Mfrn1; Slc25a37), a member of the solute carrier family localized in the mitochondrial inner membrane, functions as an essential iron importer for the synthesis of mitochondrial heme and iron–sulfur clusters in erythroblasts. The biochemistry of Mfrn1-mediated iron transport into the mitochondria, however, is poorly understood. Here, we used the strategy of in vivo epitope-tagging affinity purification and mass spectrometry to investigate Mfrn1-mediated mitochondrial iron homeostasis. Abcb10, a mitochondrial inner membrane ATP-binding cassette transporter highly induced during erythroid maturation in hematopoietic tissues, was found as one key protein that physically interacts with Mfrn1 during mouse erythroleukemia (MEL) cell differentiation. Mfrn1 was shown previously to have a longer protein half-life in differentiated MEL cells compared with undifferentiated cells. In this study, Abcb10 was found to enhance the stabilization of Mfrn1 protein in MEL cells and transfected heterologous COS7 cells. In undifferentiated MEL cells, cotransfected Abcb10 specifically interacts with Mfrn1 to enhance its protein stability and promote Mfrn1-dependent mitochondrial iron importation. The structural stabilization of the Mfrn1–Abcb10 complex demonstrates a previously uncharacterized function for Abcb10 in mitochondria. Furthermore, the binding domain of Mfrn1–Abcb10 interaction maps to the N terminus of Mfrn1. These results suggest the tight regulation of mitochondrial iron acquisition and heme synthesis in erythroblasts is mediated by both transcriptional and posttranslational mechanisms, whereby the high level of Mfrn1 is stabilized by oligomeric protein complexes.


Journal of Biological Chemistry | 1996

Characterization of Yeast Methyl Sterol Oxidase (ERG25) and Identification of a Human Homologue

Liangtao Li; Jerry Kaplan

A yeast mutant (LT06) was isolated that showed no growth on iron-limited medium but normal growth on iron-replete medium. A gene cloned from a genomic yeast library complemented the defect, allowing growth on low iron medium. Allelic segregation analysis demonstrated that the cloned gene was the normal allele rather than a high copy suppressor. A disruption mutant was nonviable, indicating that the gene was essential. Sequence analysis and functional assays indicated that the cloned gene was identical to ERG25, a gene that codes for methyl sterol oxidase. Incubation of LT06 in low iron medium resulted in marked changes in lipid metabolism, including the accumulation of fatty acids, triglycerides, methyl sterols, and other sterol precursors. A human homologue of ERG25 was cloned, sequenced, and mapped to human chromosome 4q32-34. Analysis of the data base with both ERG25 and the human homologue resulted in the identification of a putative set of metal binding motifs with similarity to that seen in a family of membrane desaturases-hydroxylases. Western analysis using antibodies to an Erg25-GST fusion protein detected two proteins of 34 and 75 kDa. Both proteins are membrane bound and contain one N-glycosyl unit. Immunofluorescence data suggest that the proteins are present in the endoplasmic reticulum and plasma membrane. Although ERG25 transcripts are not iron regulated, there is a large increase in the concentration of transcript in the mutant LT06 grown in low iron medium. These results suggest that the enzyme is regulated not by iron but by an end product of the ergosterol pathway.

Collaboration


Dive into the angtao Li's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Barry H. Paw

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Wen Chen

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Iman J. Schultz

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nathaniel B. Langer

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Caiyong Chen

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Dhvanit I. Shah

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jeffrey D. Cooney

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Eric L. Pierce

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge