Network


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

Hotspot


Dive into the research topics where Anren Song is active.

Publication


Featured researches published by Anren Song.


Journal of Clinical Investigation | 2014

Elevated sphingosine-1-phosphate promotes sickling and sickle cell disease progression

Yujin Zhang; Vladimir Berka; Anren Song; Kaiqi Sun; Wei Wang; Weiru Zhang; Chen Ning; Chonghua Li; Qibo Zhang; Mikhail Bogdanov; Danny Alexander; Michael V. Milburn; Mostafa H. Ahmed; Han Lin; Modupe Idowu; Jun Zhang; Gregory J. Kato; Osheiza Abdulmalik; Wenzheng Zhang; William Dowhan; Rodney E. Kellems; Pumin Zhang; Jianping Jin; Martin K. Safo; Ah Lim Tsai; Harinder S. Juneja; Yang Xia

Sphingosine-1-phosphate (S1P) is a bioactive lipid that regulates multicellular functions through interactions with its receptors on cell surfaces. S1P is enriched and stored in erythrocytes; however, it is not clear whether alterations in S1P are involved in the prevalent and debilitating hemolytic disorder sickle cell disease (SCD). Here, using metabolomic screening, we found that S1P is highly elevated in the blood of mice and humans with SCD. In murine models of SCD, we demonstrated that elevated erythrocyte sphingosine kinase 1 (SPHK1) underlies sickling and disease progression by increasing S1P levels in the blood. Additionally, we observed elevated SPHK1 activity in erythrocytes and increased S1P in blood collected from patients with SCD and demonstrated a direct impact of elevated SPHK1-mediated production of S1P on sickling that was independent of S1P receptor activation in isolated erythrocytes. Together, our findings provide insights into erythrocyte pathophysiology, revealing that a SPHK1-mediated elevation of S1P contributes to sickling and promotes disease progression, and highlight potential therapeutic opportunities for SCD.


Circulation | 2016

Beneficial role of erythrocyte adenosine A2B receptor-mediated AMP-activated protein kinase activation in high-altitude hypoxia

Hong Liu; Yujin Zhang; Hongyu Wu; Angelo D’Alessandro; Gennady G. Yegutkin; Anren Song; Kaiqi Sun; Jessica Li; Ning-Yuan Cheng; Aji Huang; Yuan Edward Wen; Ting Ting Weng; Fayong Luo; Travis Nemkov; Hong Sun; Rodney E. Kellems; Harry Karmouty-Quintana; Kirk C. Hansen; Bihong Zhao; Andrew W. Subudhi; Sonja Jameson-Van Houten; Colleen G. Julian; Andrew T. Lovering; Holger K. Eltzschig; Michael R. Blackburn; Robert C. Roach; Yang Xia

Background: High altitude is a challenging condition caused by insufficient oxygen supply. Inability to adjust to hypoxia may lead to pulmonary edema, stroke, cardiovascular dysfunction, and even death. Thus, understanding the molecular basis of adaptation to high altitude may reveal novel therapeutics to counteract the detrimental consequences of hypoxia. Methods: Using high-throughput, unbiased metabolomic profiling, we report that the metabolic pathway responsible for production of erythrocyte 2,3-bisphosphoglycerate (2,3-BPG), a negative allosteric regulator of hemoglobin-O2 binding affinity, was significantly induced in 21 healthy humans within 2 hours of arrival at 5260 m and further increased after 16 days at 5260 m. Results: This finding led us to discover that plasma adenosine concentrations and soluble CD73 activity rapidly increased at high altitude and were associated with elevated erythrocyte 2,3-BPG levels and O2 releasing capacity. Mouse genetic studies demonstrated that elevated CD73 contributed to hypoxia-induced adenosine accumulation and that elevated adenosine-mediated erythrocyte A2B adenosine receptor activation was beneficial by inducing 2,3-BPG production and triggering O2 release to prevent multiple tissue hypoxia, inflammation, and pulmonary vascular leakage. Mechanistically, we demonstrated that erythrocyte AMP-activated protein kinase was activated in humans at high altitude and that AMP-activated protein kinase is a key protein functioning downstream of the A2B adenosine receptor, phosphorylating and activating BPG mutase and thus inducing 2,3-BPG production and O2 release from erythrocytes. Significantly, preclinical studies demonstrated that activation of AMP-activated protein kinase enhanced BPG mutase activation, 2,3-BPG production, and O2 release capacity in CD73-deficient mice, in erythrocyte-specific A2B adenosine receptor knockouts, and in wild-type mice and in turn reduced tissue hypoxia and inflammation. Conclusions: Together, human and mouse studies reveal novel mechanisms of hypoxia adaptation and potential therapeutic approaches for counteracting hypoxia-induced tissue damage.


Circulation | 2015

Elevated Placental Adenosine Signaling Contributes to the Pathogenesis of Preeclampsia

Takayuki Iriyama; Kaiqi Sun; Nicholas F. Parchim; Jessica Li; Cheng Zhao; Anren Song; Laura A. Hart; Sean C. Blackwell; Baha M. Sibai; Lee Nien L Chan; Teh Sheng Chan; M. John Hicks; Michael R. Blackburn; Rodney E. Kellems; Yang Xia

Background— Preeclampsia is a prevalent hypertensive disorder of pregnancy and a leading cause of maternal and neonatal morbidity and mortality worldwide. This pathogenic condition is speculated to be caused by placental abnormalities that contribute to the maternal syndrome. However, the specific factors and signaling pathways that lead to impaired placentas and maternal disease development remain elusive. Methods and Results— Using 2 independent animal models of preeclampsia (genetically engineered pregnant mice with elevated adenosine exclusively in placentas and a pathogenic autoantibody-induced preeclampsia mouse model), we demonstrated that chronically elevated placental adenosine was sufficient to induce hallmark features of preeclampsia, including hypertension, proteinuria, small fetuses, and impaired placental vasculature. Genetic and pharmacological approaches revealed that elevated placental adenosine coupled with excessive A2B adenosine receptor (ADORA2B) signaling contributed to the development of these features of preeclampsia. Mechanistically, we provided both human and mouse evidence that elevated placental CD73 is a key enzyme causing increased placental adenosine, thereby contributing to preeclampsia. Conclusions— We determined that elevated placental adenosine signaling is a previously unrecognized pathogenic factor for preeclampsia. Moreover, our findings revealed the molecular basis underlying the elevation of placental adenosine and the detrimental role of excess placental adenosine in the pathophysiology of preeclampsia, and thereby, we highlight novel therapeutic targets.


Nature Communications | 2016

Sphingosine-1-phosphate promotes erythrocyte glycolysis and oxygen release for adaptation to high-altitude hypoxia

Kaiqi Sun; Yujin Zhang; Angelo D'Alessandro; Travis Nemkov; Anren Song; Hongyu Wu; Hong Liu; Morayo G. Adebiyi; Aji Huang; Yuan E. Wen; Mikhail Bogdanov; Alejandro Vila; John O'Brien; Rodney E. Kellems; William Dowhan; Andrew W. Subudhi; Sonja Jameson-Van Houten; Colleen G. Julian; Andrew T. Lovering; Martin K. Safo; Kirk C. Hansen; Robert C. Roach; Yang Xia

Sphingosine-1-phosphate (S1P) is a bioactive signalling lipid highly enriched in mature erythrocytes, with unknown functions pertaining to erythrocyte physiology. Here by employing nonbiased high-throughput metabolomic profiling, we show that erythrocyte S1P levels rapidly increase in 21 healthy lowland volunteers at 5,260 m altitude on day 1 and continue increasing to 16 days with concurrently elevated erythrocyte sphingonisne kinase 1 (Sphk1) activity and haemoglobin (Hb) oxygen (O2) release capacity. Mouse genetic studies show that elevated erythrocyte Sphk1-induced S1P protects against tissue hypoxia by inducing O2 release. Mechanistically, we show that intracellular S1P promotes deoxygenated Hb anchoring to the membrane, enhances the release of membrane-bound glycolytic enzymes to the cytosol, induces glycolysis and thus the production of 2,3-bisphosphoglycerate (2,3-BPG), an erythrocyte-specific glycolytic intermediate, which facilitates O2 release. Altogether, we reveal S1P as an intracellular hypoxia-responsive biolipid promoting erythrocyte glycolysis, O2 delivery and thus new therapeutic opportunities to counteract tissue hypoxia.


Blood | 2015

Elevated Adenosine Signaling Via Adenosine A2B Receptor Induces Normal and Sickle Erythrocyte Sphingosine Kinase 1 Activity

Kaiqi Sun; Yujin Zhang; Mikhail Bogdanov; Hongyu Wu; Anren Song; Jessica Li; William Dowhan; Modupe Idowu; Harinder S. Juneja; Jose G. Molina; Michael R. Blackburn; Rodney E. Kellems; Yang Xia

Erythrocyte possesses high sphingosine kinase 1 (SphK1) activity and is the major cell type supplying plasma sphingosine-1-phosphate, a signaling lipid regulating multiple physiological and pathological functions. Recent studies revealed that erythrocyte SphK1 activity is upregulated in sickle cell disease (SCD) and contributes to sickling and disease progression. However, how erythrocyte SphK1 activity is regulated remains unknown. Here we report that adenosine induces SphK1 activity in human and mouse sickle and normal erythrocytes in vitro. Next, using 4 adenosine receptor-deficient mice and pharmacological approaches, we determined that the A2B adenosine receptor (ADORA2B) is essential for adenosine-induced SphK1 activity in human and mouse normal and sickle erythrocytes in vitro. Subsequently, we provide in vivo genetic evidence that adenosine deaminase (ADA) deficiency leads to excess plasma adenosine and elevated erythrocyte SphK1 activity. Lowering adenosine by ADA enzyme therapy or genetic deletion of ADORA2B significantly reduced excess adenosine-induced erythrocyte SphK1 activity in ADA-deficient mice. Finally, we revealed that protein kinase A-mediated extracellular signal-regulated kinase 1/2 activation functioning downstream of ADORA2B underlies adenosine-induced erythrocyte SphK1 activity. Overall, our findings reveal a novel signaling network regulating erythrocyte SphK1 and highlight innovative mechanisms regulating SphK1 activity in normal and SCD.


Nature Communications | 2017

Erythrocytes retain hypoxic adenosine response for faster acclimatization upon re-ascent

Anren Song; Yujin Zhang; Leng Han; Gennady G. Yegutkin; Hong Liu; Kaiqi Sun; Angelo D'Alessandro; Jessica Li; Harry Karmouty-Quintana; Takayuki Iriyama; Tingting Weng; Shushan Zhao; Wei Wang; Hongyu Wu; Travis Nemkov; Andrew W. Subudhi; Sonja Jameson-Van Houten; Colleen G. Julian; Andrew T. Lovering; Kirk C. Hansen; Hong Zhang; Mikhail Bogdanov; William Dowhan; Jianping Jin; Rodney E. Kellems; Holger K. Eltzschig; Michael R. Blackburn; Robert C. Roach; Yang Xia

Faster acclimatization to high altitude upon re-ascent is seen in humans; however, the molecular basis for this enhanced adaptive response is unknown. We report that in healthy lowlanders, plasma adenosine levels are rapidly induced by initial ascent to high altitude and achieved even higher levels upon re-ascent, a feature that is positively associated with quicker acclimatization. Erythrocyte equilibrative nucleoside transporter 1 (eENT1) levels are reduced in humans at high altitude and in mice under hypoxia. eENT1 deletion allows rapid accumulation of plasma adenosine to counteract hypoxic tissue damage in mice. Adenosine signalling via erythrocyte ADORA2B induces PKA phosphorylation, ubiquitination and proteasomal degradation of eENT1. Reduced eENT1 resulting from initial hypoxia is maintained upon re-ascent in humans or re-exposure to hypoxia in mice and accounts for erythrocyte hypoxic memory and faster acclimatization. Our findings suggest that targeting identified purinergic-signalling network would enhance the hypoxia adenosine response to counteract hypoxia-induced maladaptation.


Haematologica | 2017

Hypoxia modulates the purine salvage pathway and decreases red blood cell and supernatant levels of hypoxanthine during refrigerated storage

Travis Nemkov; Kaiqi Sun; Julie A. Reisz; Anren Song; Tatsuro Yoshida; Andrew Dunham; Matthew J. Wither; Richard O. Francis; Robert C. Roach; Monika Dzieciatkowska; Stephen C. Rogers; Allan Doctor; Anastasios G. Kriebardis; Marianna H. Antonelou; Issidora S. Papassideri; Carolyn T. Young; Tiffany Thomas; Kirk C. Hansen; Steven L. Spitalnik; Yang Xia; James C. Zimring; Eldad A. Hod; Angelo D’Alessandro

Hypoxanthine catabolism in vivo is potentially dangerous as it fuels production of urate and, most importantly, hydrogen peroxide. However, it is unclear whether accumulation of intracellular and supernatant hypoxanthine in stored red blood cell units is clinically relevant for transfused recipients. Leukoreduced red blood cells from glucose-6-phosphate dehydrogenase-normal or -deficient human volunteers were stored in AS-3 under normoxic, hyperoxic, or hypoxic conditions (with oxygen saturation ranging from <3% to >95%). Red blood cells from healthy human volunteers were also collected at sea level or after 1–7 days at high altitude (>5000 m). Finally, C57BL/6J mouse red blood cells were incubated in vitro with 13C1-aspartate or 13C5-adenosine under normoxic or hypoxic conditions, with or without deoxycoformycin, a purine deaminase inhibitor. Metabolomics analyses were performed on human and mouse red blood cells stored for up to 42 or 14 days, respectively, and correlated with 24 h post-transfusion red blood cell recovery. Hypoxanthine increased in stored red blood cell units as a function of oxygen levels. Stored red blood cells from human glucose-6-phosphate dehydrogenase-deficient donors had higher levels of deaminated purines. Hypoxia in vitro and in vivo decreased purine oxidation and enhanced purine salvage reactions in human and mouse red blood cells, which was partly explained by decreased adenosine monophosphate deaminase activity. In addition, hypoxanthine levels negatively correlated with post-transfusion red blood cell recovery in mice and – preliminarily albeit significantly - in humans. In conclusion, hypoxanthine is an in vitro metabolic marker of the red blood cell storage lesion that negatively correlates with post-transfusion recovery in vivo. Storage-dependent hypoxanthine accumulation is ameliorated by hypoxia-induced decreases in purine deamination reaction rates.


Scientific Reports | 2016

Hypoxia-mediated impaired erythrocyte Lands’ Cycle is pathogenic for sickle cell disease

Hongyu Wu; Mikhail Bogdanov; Yujin Zhang; Kaiqi Sun; Shushan Zhao; Anren Song; Renna Luo; Nicholas F. Parchim; Hong Liu; Aji Huang; Morayo G. Adebiyi; Jianping Jin; Danny Alexander; Michael V. Milburn; Modupe Idowu; Harinder S. Juneja; Rodney E. Kellems; William Dowhan; Yang Xia

Although Lands’ cycle was discovered in 1958, its function and cellular regulation in membrane homeostasis under physiological and pathological conditions remain largely unknown. Nonbiased high throughput metabolomic profiling revealed that Lands’ cycle was impaired leading to significantly elevated erythrocyte membrane lysophosphatidylcholine (LysoPC) content and circulating and erythrocyte arachidonic acid (AA) in mice with sickle cell disease (SCD), a prevalent hemolytic genetic disorder. Correcting imbalanced Lands’ cycle by knockdown of phospholipase 2 (cPLA2) or overexpression of lysophosphatidycholine acyltransferase 1 (LPCAT1), two key enzymes of Lands’ cycle in hematopoietic stem cells, reduced elevated erythrocyte membrane LysoPC content and circulating AA levels and attenuated sickling, inflammation and tissue damage in SCD chimeras. Human translational studies validated SCD mouse findings and further demonstrated that imbalanced Lands’ cycle induced LysoPC production directly promotes sickling in cultured mouse and human SCD erythrocytes. Mechanistically, we revealed that hypoxia-mediated ERK activation underlies imbalanced Lands’ cycle by preferentially inducing the activity of PLA2 but not LPCAT in human and mouse SCD erythrocytes. Overall, our studies have identified a pathological role of imbalanced Lands’ cycle in SCD erythrocytes, novel molecular basis regulating Lands’ cycle and therapeutic opportunities for the disease.


Scientific Reports | 2017

Structural and Functional Insight of Sphingosine 1-Phosphate-Mediated Pathogenic Metabolic Reprogramming in Sickle Cell Disease.

Kaiqi Sun; Angelo D'Alessandro; Mostafa H. Ahmed; Yujin Zhang; Anren Song; Tzu Ping Ko; Travis Nemkov; Julie A. Reisz; Hongyu Wu; Morayo G. Adebiyi; Zhangzhe Peng; Jing Gong; Hong Liu; Aji Huang; Yuan Edward Wen; Alexander Q. Wen; Vladimir Berka; Mikhail Bogdanov; Osheiza Abdulmalik; Leng Han; Ah Lim Tsai; Modupe Idowu; Harinder S. Juneja; Rodney E. Kellems; William Dowhan; Kirk C. Hansen; Martin K. Safo; Yang Xia

Elevated sphingosine 1-phosphate (S1P) is detrimental in Sickle Cell Disease (SCD), but the mechanistic basis remains obscure. Here, we report that increased erythrocyte S1P binds to deoxygenated sickle Hb (deoxyHbS), facilitates deoxyHbS anchoring to the membrane, induces release of membrane-bound glycolytic enzymes and in turn switches glucose flux towards glycolysis relative to the pentose phosphate pathway (PPP). Suppressed PPP causes compromised glutathione homeostasis and increased oxidative stress, while enhanced glycolysis induces production of 2,3-bisphosphoglycerate (2,3-BPG) and thus increases deoxyHbS polymerization, sickling, hemolysis and disease progression. Functional studies revealed that S1P and 2,3-BPG work synergistically to decrease both HbA and HbS oxygen binding affinity. The crystal structure at 1.9 Å resolution deciphered that S1P binds to the surface of 2,3-BPG-deoxyHbA and causes additional conformation changes to the T-state Hb. Phosphate moiety of the surface bound S1P engages in a highly positive region close to α1-heme while its aliphatic chain snakes along a shallow cavity making hydrophobic interactions in the “switch region”, as well as with α2-heme like a molecular “sticky tape” with the last 3–4 carbon atoms sticking out into bulk solvent. Altogether, our findings provide functional and structural bases underlying S1P-mediated pathogenic metabolic reprogramming in SCD and novel therapeutic avenues.


Hypertension | 2017

Elevated Adenosine Induces Placental DNA Hypomethylation Independent of A2B Receptor Signaling in Preeclampsia

Aji Huang; Hongyu Wu; Takayuki Iriyama; Yujin Zhang; Kaiqi Sun; Anren Song; Hong Liu; Zhangzhe Peng; Lili Tang; Minjung Lee; Yun Huang; Xin Ni; Rodney E. Kellems; Yang Xia

Preeclampsia is a prevalent pregnancy hypertensive disease with both maternal and fetal morbidity and mortality. Emerging evidence indicates that global placental DNA hypomethylation is observed in patients with preeclampsia and is linked to altered gene expression and disease development. However, the molecular basis underlying placental epigenetic changes in preeclampsia remains unclear. Using 2 independent experimental models of preeclampsia, adenosine deaminase–deficient mice and a pathogenic autoantibody-induced mouse model of preeclampsia, we demonstrate that elevated placental adenosine not only induces hallmark features of preeclampsia but also causes placental DNA hypomethylation. The use of genetic approaches to express an adenosine deaminase minigene specifically in placentas, or adenosine deaminase enzyme replacement therapy, restored placental adenosine to normal levels, attenuated preeclampsia features, and abolished placental DNA hypomethylation in adenosine deaminase–deficient mice. Genetic deletion of CD73 (an ectonucleotidase that converts AMP to adenosine) prevented the elevation of placental adenosine in the autoantibody-induced preeclampsia mouse model and ameliorated preeclampsia features and placental DNA hypomethylation. Immunohistochemical studies revealed that elevated placental adenosine–mediated DNA hypomethylation predominantly occurs in spongiotrophoblasts and labyrinthine trophoblasts and that this effect is independent of A2B adenosine receptor activation in both preeclampsia models. Extending our mouse findings to humans, we used cultured human trophoblasts to demonstrate that adenosine functions intracellularly and induces DNA hypomethylation without A2B adenosine receptor activation. Altogether, both mouse and human studies reveal novel mechanisms underlying placental DNA hypomethylation and potential therapeutic approaches for preeclampsia.

Collaboration


Dive into the Anren Song's collaboration.

Top Co-Authors

Avatar

Yang Xia

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Rodney E. Kellems

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Kaiqi Sun

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Hong Liu

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Yujin Zhang

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Michael R. Blackburn

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Harinder S. Juneja

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hongyu Wu

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Kirk C. Hansen

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge