Network


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

Hotspot


Dive into the research topics where Xiaojing Yuan is active.

Publication


Featured researches published by Xiaojing Yuan.


PLOS Pathogens | 2012

Heme Uptake by Leishmania amazonensis Is Mediated by the Transmembrane Protein LHR1

Chau Huynh; Xiaojing Yuan; Danilo C. Miguel; Rebecca L. Renberg; Olga Protchenko; Caroline C. Philpott; Iqbal Hamza; Norma W. Andrews

Trypanosomatid protozoan parasites lack a functional heme biosynthetic pathway, so must acquire heme from the environment to survive. However, the molecular pathway responsible for heme acquisition by these organisms is unknown. Here we show that L. amazonensis LHR1, a homolog of the C. elegans plasma membrane heme transporter HRG-4, functions in heme transport. Tagged LHR1 localized to the plasma membrane and to endocytic compartments, in both L. amazonensis and mammalian cells. Heme deprivation in L. amazonensis increased LHR1 transcript levels, promoted uptake of the fluorescent heme analog ZnMP, and increased the total intracellular heme content of promastigotes. Conversely, deletion of one LHR1 allele reduced ZnMP uptake and the intracellular heme pool by approximately 50%, indicating that LHR1 is a major heme importer in L. amazonensis. Viable parasites with correct replacement of both LHR1 alleles could not be obtained despite extensive attempts, suggesting that this gene is essential for the survival of promastigotes. Notably, LHR1 expression allowed Saccharomyces cerevisiae to import heme from the environment, and rescued growth of a strain deficient in heme biosynthesis. Syntenic genes with high sequence identity to LHR1 are present in the genomes of several species of Leishmania and also Trypanosoma cruzi and Trypanosoma brucei, indicating that therapeutic agents targeting this transporter could be effective against a broad group of trypanosomatid parasites that cause serious human disease.


Journal of Biological Chemistry | 2012

Topologically Conserved Residues Direct Heme Transport in HRG-1-related Proteins

Xiaojing Yuan; Olga Protchenko; Caroline C. Philpott; Iqbal Hamza

Background: HRG-1-related proteins have low sequence homology but are functionally conserved. Results: Conserved residues within the transmembrane domain and the C terminus dictate the function of HRG-1-related proteins. Conclusion: Heme transport is mediated by topologically conserved residues implying an evolutionary ancient transport mechanism. Significance: Understanding the mechanism of HRG-1-related protein function will provide novel therapeutic insights into human hematological disorders and parasites, which rely on host heme for survival. Caenorhabditis elegans and human HRG-1-related proteins are conserved, membrane-bound permeases that bind and translocate heme in metazoan cells via a currently uncharacterized mechanism. Here, we show that cellular import of heme by HRG-1-related proteins from worms and humans requires strategically located amino acids that are topologically conserved across species. We exploit a heme synthesis-defective Saccharomyces cerevisiae mutant to model the heme auxotrophy of C. elegans and demonstrate that, under heme-deplete conditions, the endosomal CeHRG-1 requires both a specific histidine in the predicted second transmembrane domain (TMD2) and the FARKY motif in the C terminus tail for heme transport. By contrast, the plasma membrane CeHRG-4 transports heme by utilizing a histidine in the exoplasmic (E2) loop and the FARKY motif. Optimal activity under heme-limiting conditions, however, requires histidine in the E2 loop of CeHRG-1 and tyrosine in TMD2 of CeHRG-4. An analogous system exists in humans, because mutation of the synonymous histidine in TMD2 of hHRG-1 eliminates heme transport activity, implying an evolutionary conserved heme transport mechanism that predates vertebrate origins. Our results support a model in which heme is translocated across membranes facilitated by conserved amino acids positioned on the exoplasmic, cytoplasmic, and transmembrane regions of HRG-1-related proteins. These findings may provide a framework for understanding the structural basis of heme transport in eukaryotes and human parasites, which rely on host heme for survival.


Current Opinion in Chemical Biology | 2013

Heme transport and erythropoiesis

Xiaojing Yuan; Mark D. Fleming; Iqbal Hamza

In humans, systemic heme homeostasis is achieved via coordinated regulation of heme synthesis, transport and degradation. Although the heme biosynthesis and degradation pathways have been well characterized, the pathways for heme trafficking and incorporation into hemoproteins remain poorly understood. In the past few years, researchers have exploited genetic, cellular and biochemical tools, to identify heme transporters and, in the process, reveal unexpected functions for this elusive group of proteins. However, given the complexity of heme trafficking pathways, current knowledge of heme transporters is fragmented and sometimes contradictory. This review seeks to focus on recent studies on heme transporters with specific emphasis on their functions during erythropoiesis.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Heme dynamics and trafficking factors revealed by genetically encoded fluorescent heme sensors.

David A. Hanna; Raven M. Harvey; Osiris Martinez-Guzman; Xiaojing Yuan; Bindu Chandrasekharan; Gheevarghese Raju; F. Wayne Outten; Iqbal Hamza; Amit R. Reddi

Significance All heme-dependent functions require the mobilization of labile heme (LH), of which there is little understanding of its nature and dynamics. To probe LH pools, we developed genetically encoded fluorescent heme sensors and deployed them in the unicellular eukaryote Saccharomyces cerevisiae. We find that LH is relatively abundant in the cytosol, but exceedingly low in the mitochondria and nucleus. Further, we find that LH can be mobilized by signaling molecules like nitric oxide. We also find that the glycolytic enzyme glyceraldehyde phosphate dehydrogenase constitutes a major heme buffer and is responsible for regulating the activity of a heme-dependent transcription factor. Altogether, our work will have profound implications for understanding the mechanisms of heme utilization. Heme is an essential cofactor and signaling molecule. Heme acquisition by proteins and heme signaling are ultimately reliant on the ability to mobilize labile heme (LH). However, the properties of LH pools, including concentration, oxidation state, distribution, speciation, and dynamics, are poorly understood. Herein, we elucidate the nature and dynamics of LH using genetically encoded ratiometric fluorescent heme sensors in the unicellular eukaryote Saccharomyces cerevisiae. We find that the subcellular distribution of LH is heterogeneous; the cytosol maintains LH at ∼20–40 nM, whereas the mitochondria and nucleus maintain it at concentrations below 2.5 nM. Further, we find that the signaling molecule nitric oxide can initiate the rapid mobilization of heme in the cytosol and nucleus from certain thiol-containing factors. We also find that the glycolytic enzyme glyceraldehyde phosphate dehydrogenase constitutes a major cellular heme buffer, and is responsible for maintaining the activity of the heme-dependent nuclear transcription factor heme activator protein (Hap1p). Altogether, we demonstrate that the heme sensors can be used to reveal fundamental aspects of heme trafficking and dynamics and can be used across multiple organisms, including Escherichia coli, yeast, and human cell lines.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Regulation of intracellular heme trafficking revealed by subcellular reporters

Xiaojing Yuan; Nicole Rietzschel; Hanna Kwon; Ana Beatriz Walter Nuno; David A. Hanna; John D. Phillips; Emma Lloyd Raven; Amit R. Reddi; Iqbal Hamza

Significance The intracellular and extracellular trafficking of heme, a hydrophobic and potentially cytotoxic cofactor in proteins such as hemoglobin, remains an underexplored area. While cellular heme can be derived exogenously or from de novo synthesis, it is unclear if there is differential trafficking of heme from these two sources. To critically examine this possibility, we developed peroxidase-based enzymatic reporters for heme and deployed them in subcellular compartments in mammalian cells and in several tissues in the Caenorhabditis elegans animal model. Our results show that extracellular and endogenous heme is trafficked to virtually all intracellular compartments via distinct cellular routes and that interorgan heme transport is essential for systemic regulation of heme homeostasis in C. elegans. Heme is an essential prosthetic group in proteins that reside in virtually every subcellular compartment performing diverse biological functions. Irrespective of whether heme is synthesized in the mitochondria or imported from the environment, this hydrophobic and potentially toxic metalloporphyrin has to be trafficked across membrane barriers, a concept heretofore poorly understood. Here we show, using subcellular-targeted, genetically encoded hemoprotein peroxidase reporters, that both extracellular and endogenous heme contribute to cellular labile heme and that extracellular heme can be transported and used in toto by hemoproteins in all six subcellular compartments examined. The reporters are robust, show large signal-to-background ratio, and provide sufficient range to detect changes in intracellular labile heme. Restoration of reporter activity by heme is organelle-specific, with the Golgi and endoplasmic reticulum being important sites for both exogenous and endogenous heme trafficking. Expression of peroxidase reporters in Caenorhabditis elegans shows that environmental heme influences labile heme in a tissue-dependent manner; reporter activity in the intestine shows a linear increase compared with muscle or hypodermis, with the lowest heme threshold in neurons. Our results demonstrate that the trafficking pathways for exogenous and endogenous heme are distinct, with intrinsic preference for specific subcellular compartments. We anticipate our results will serve as a heuristic paradigm for more sophisticated studies on heme trafficking in cellular and whole-animal models.


PLOS Neglected Tropical Diseases | 2015

The Heme Transport Capacity of LHR1 Determines the Extent of Virulence in Leishmania amazonensis

Rebecca L. Renberg; Xiaojing Yuan; Tamika K. Samuel; Danilo C. Miguel; Iqbal Hamza; Norma W. Andrews; Andrew R. Flannery

Leishmania spp. are trypanosomatid parasites that replicate intracellularly in macrophages, causing serious human morbidity and mortality throughout the world. Trypanosomatid protozoa cannot synthesize heme, so must acquire this essential cofactor from their environment. Earlier studies identified LHR1 as a Leishmania amazonensis transmembrane protein that mediates heme uptake. Null mutants of LHR1 are not viable and single knockout strains have reduced virulence, but very little is known about the properties of LHR1 directly associated with heme transport. Here, we use functional assays in Saccharomyces cerevisiae to show that specific tyrosine residues within the first three predicted transmembrane domains of LHR1 are required for efficient heme uptake. These tyrosines are unique to LHR1, consistent with the low similarity between LHR1 and its corresponding homologs in C. elegans and human. Substitution of these tyrosines in LHR1 resulted in varying degrees of heme transport inhibition, phenotypes that closely mirrored the impaired ability of L. amazonensis to replicate as intracellular amastigotes in macrophages and generate cutaneous lesions in mice. Taken together, our results imply that the mechanism for heme transport by LHR1 is distinctive and may have adapted to secure heme, a limiting cofactor, inside the host. Since LHR1 is significantly divergent from the human heme transporter HRG1, our findings lay the groundwork for selective targeting of LHR1 by small molecule antagonists.


FEBS Journal | 2017

Characterization of Plasma Labile Heme in Hemolytic Conditions

Zélia Gouveia; Ana Rita Carlos; Xiaojing Yuan; Frederico Aires-da-Silva; Roland Stocker; Ghassan J. Maghzal; Sónia S. Leal; Cláudio M. Gomes; Smilja Todorovic; Olga Iranzo; Susana Ramos; Ana Catarina Santos; Iqbal Hamza; João Gonçalves; Miguel P. Soares

Extracellular hemoglobin, a byproduct of hemolysis, can release its prosthetic heme groups upon oxidation. This produces metabolically active heme that is exchangeable between acceptor proteins, macromolecules and low molecular weight ligands, termed here labile heme. As it accumulates in plasma labile heme acts in a pro‐oxidant manner and regulates cellular metabolism while exerting pro‐inflammatory and cytotoxic effects that foster the pathogenesis of hemolytic diseases. Here, we developed and characterized a panel of heme‐specific single domain antibodies (sdAbs) that together with a cellular‐based heme reporter assay, allow for quantification and characterization of labile heme in plasma during hemolytic conditions. Using these approaches, we demonstrate that when generated during hemolytic conditions labile heme is bound to plasma molecules with an affinity higher than 10−7 m and that 2–8% (~ 2–5 μm) of the total amount of heme detected in plasma can be internalized by bystander cells, termed here bioavailable heme. Acute, but not chronic, hemolysis is associated with transient reduction of plasma heme‐binding capacity, that is, the ability of plasma molecules to bind labile heme with an affinity higher than 10−7 m. The heme‐specific sdAbs neutralize the pro‐oxidant activity of soluble heme in vitro, suggesting that these maybe used to counter the pathologic effects of labile heme during hemolytic conditions. Finally, we show that heme‐specific sdAbs can be used to visualize cellular heme. In conclusion, we describe a panel of heme‐specific sdAbs that when used with other approaches provide novel insights to the pathophysiology of heme.


The FASEB Journal | 2016

Heme acquisition in the parasitic filarial nematode Brugia malayi

Ashley N. Luck; Xiaojing Yuan; Denis Voronin; Barton E. Slatko; Iqbal Hamza; Jeremy M. Foster

Nematodes lack a heme biosynthetic pathway and must acquire heme from exogenous sources. Given the indispensable role of heme, this auxotrophy may be exploited to develop drugs that interfere with heme uptake in parasites. Although multiple heme‐responsive genes (HRGs) have been characterized within the free‐living nematode Caenorhabditis elegans, we have undertaken the first study of heme transport in Brugia malayi, a causative agent of lymphatic filariasis. Through functional assays in yeast, as well as heme analog, RNAi, and transcriptomic experiments, we have shown that the heme transporter B. malayi HRG‐1 (BmHRG‐1) is indeed functional in B. malayi. In addition, BmHRG‐1 localizes both to the endocytic compartments and cell membrane when expressed in yeast cells. Transcriptomic sequencing revealed that BmHRG‐1, BmHRG‐2, and BmMRP‐5 (all orthologs of HRGs in C. elegans) are down‐regulated in heme‐treated B. malayi, as compared tonon–heme‐treated control worms. Likely because of short gene lengths, multiple exons, other HRGs in B. malayi (BmHRG‐3–6) remain unidentified. Although the precise mechanisms of heme homeostasis in a nematode with the ability to acquire heme remains unknown, this study clearly demonstrates that the filarial nematode B. malayi is capable of transporting exogenous heme.—Luck, A. N., Yuan, X., Voronin, D., Slatko, B. E., Hamza, I., Foster, J. M. Heme acquisition in the parasitic filarial nematode Brugia malayi. FASEB J. 30, 3501–3514 (2016). www.fasebj.org


Analytical Chemistry | 2018

Label-Free Imaging of Heme Dynamics in Living Organisms by Transient Absorption Microscopy

Andy Chen; Xiaojing Yuan; Junjie Li; Puting Dong; Iqbal Hamza; Ji-Xin Cheng

Heme, a hydrophobic and cytotoxic macrocycle, is an essential cofactor in a large number of proteins and is important for cell signaling. This must mean that heme is mobilized from its place of synthesis or entry into the cell to other parts of the cell where hemoproteins reside. However, the cellular dynamics of heme movement is not well understood, in large part due to the inability to image heme noninvasively in live biological systems. Here, using high-resolution transient absorption microscopy, we showed that heme storage and distribution is dynamic in Caenorhabditis elegans. Intracellular heme exists in concentrated granular puncta which localizes to lysosomal-related organelles. These granules are dynamic, and their breaking down into smaller granules provides a mechanism by which heme stores can be mobilized. Collectively, these direct and noninvasive dynamic imaging techniques provide new insights into heme storage and transport and open a new avenue for label-free investigation of heme function and regulation in living systems.


Communications Biology | 2018

Structural basis for promotion of duodenal iron absorption by enteric ferric reductase with ascorbate

Menega Ganasen; Hiromi Togashi; Hanae Takeda; Honami Asakura; Takehiko Tosha; Keitaro Yamashita; Kunio Hirata; Yuko Nariai; Takeshi Urano; Xiaojing Yuan; Iqbal Hamza; A. Grant Mauk; Yoshitsugu Shiro; Hiroshi Sugimoto; Hitomi Sawai

Dietary iron absorption is regulated by duodenal cytochrome b (Dcytb), an integral membrane protein that catalyzes reduction of nonheme Fe3+ by electron transfer from ascorbate across the membrane. This step is essential to enable iron uptake by the divalent metal transporter. Here we report the crystallographic structures of human Dcytb and its complex with ascorbate and Zn2+. Each monomer of the homodimeric protein possesses cytoplasmic and apical heme groups, as well as cytoplasmic and apical ascorbate-binding sites located adjacent to each heme. Zn2+ coordinates to two hydroxyl groups of the apical ascorbate and to a histidine residue. Biochemical analysis indicates that Fe3+ competes with Zn2+ for this binding site. These results provide a structural basis for the mechanism by which Fe3+ uptake is promoted by reducing agents and should facilitate structure-based development of improved agents for absorption of orally administered iron.Ganasen et al. report the crystallographic structures of human duodenal cytochrome b and its complex with ascorbate and Zn2+. This study provides mechanistic insights into how reducing agents promote the uptake of orally administered iron and may facilitate the development of such interventions.

Collaboration


Dive into the Xiaojing Yuan's collaboration.

Top Co-Authors

Avatar

Caroline C. Philpott

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amit R. Reddi

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

David A. Hanna

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hanna Kwon

University of Leicester

View shared research outputs
Top Co-Authors

Avatar

Ana Beatriz Walter Nuno

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ariane Lapierre

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge