Network


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

Hotspot


Dive into the research topics where Xiaobin Han is active.

Publication


Featured researches published by Xiaobin Han.


Endocrinology | 2013

Calcium Regulates FGF-23 Expression in Bone

Valentin David; Bing Dai; Aline Martin; Jinsong Huang; Xiaobin Han; L. Darryl Quarles

Calcium has recently been shown to regulate fibroblast growth factor 23 (FGF-23), a bone-derived phosphate and vitamin D-regulating hormone. To better understand the regulation of FGF-23 by calcium, phosphorus, 1,25 dihydroxyvitamin D3 [1,25(OH)2D], and PTH, we examined FGF-23 expression under basal conditions and in response to PTH, doxercalciferol, or high-calcium diet treatment in Gcm2(-/-) and Cyp27b1(-/-) mutant mice. Gcm2(-/-) mice exhibited low serum PTH and 1,25(OH)2D concentrations, hypocalcemia, and hyperphosphatemia, whereas Cyp27b1(-/-) mice had high PTH, undetectable 1,25(OH)2D, hypocalcemia, and hypophosphatemia. Serum FGF-23 levels were decreased in both mutant models. Doxercalciferol administration increased serum FGF-23 levels in both mutant models. PTH administration to Gcm2(-/-) mice also increased serum FGF-23 levels, in association with an increase in both 1,25(OH)2D and calcium concentrations. Multiple regression analysis of pooled data indicated that changes in FGF-23 were positively correlated with serum calcium and 1,25(OH)2D but not related to changes in serum phosphate concentrations. A high-calcium diet also increased serum FGF-23 concentrations in Cyp27b1(-/-) mice in the absence of 1,25(OH)2D and in Gcm2(-/-) mice with low PTH. The addition of calcium to the culture media also stimulated FGF-23 message expression in MC3T3-E1 osteoblasts. In addition, FGF-23 promoter activity in cultured osteoblasts was inhibited by the L-calcium-channel inhibitor nifedipine and stimulated by calcium ionophores. The effects of chronic low calcium to prevent 1,25(OH)2D and PTH stimulation of FGF-23 in these mutant mouse models suggest that suppression of FGF-23 plays an important physiological adaptive response to hypocalcemia.


PLOS ONE | 2014

Osteocyte-specific deletion of Fgfr1 suppresses FGF23.

Zhousheng Xiao; Jinsong Huang; Li Cao; Y. Liang; Xiaobin Han; Leigh Darryl Quarles

Increases in fibroblastic growth factor 23 (FGF23 or Fgf23) production by osteocytes result in hypophosphatemia and rickets in the Hyp mouse homologue of X-linked hypophosphatemia (XLH). Fibroblastic growth factor (FGF) signaling has been implicated in the pathogenesis of Hyp. Here, we conditionally deleted FGF receptor 1 (FGFR1 or Fgfr1) in osteocytes of Hyp mice to investigate the role of autocrine/paracrine FGFR signaling in regulating FGF23 production by osteocytes. Crossing dentin matrix protein 1 (Dmp1)-Cre;Fgfr1 null/+ mice with female Hyp;Fgfr1 flox/flox mice created Hyp and Fgfr1 (Fgfr1Dmp1-cKO)-null mice (Hyp;Fgfr1 Dmp1-cKO) with a 70% decrease in bone Fgfr1 transcripts. Fgfr1Dmp1-cKO-null mice exhibited a 50% reduction in FGF23 expression in bone and 3-fold reduction in serum FGF23 concentrations, as well as reductions in sclerostin (Sost), phosphate regulating endopeptidase on X chromosome (PHEX or Phex), matrix extracellular phosphoglycoprotein (Mepe), and Dmp1 transcripts, but had no demonstrable alterations in phosphate or vitamin D homeostasis or skeletal morphology. Hyp mice had hypophosphatemia, reductions in 1,25(OH)2D levels, rickets/osteomalacia and elevated FGF2 expression in bone. Compared to Hyp mice, compound Hyp;Fgfr1 Dmp1-cKO-null mice had significant improvement in rickets and osteomalacia in association with a decrease in serum FGF23 (3607 to 1099 pg/ml), an increase in serum phosphate (6.0 mg/dl to 9.3 mg/dl) and 1,25(OH)2D (121±23 to 192±34 pg/ml) levels, but only a 30% reduction in bone FGF23 mRNA expression. FGF23 promoter activity in osteoblasts was stimulated by FGFR1 activation and inhibited by overexpression of a dominant negative FGFR1(TK−), PLCγ and MAPK inhibitors. FGF2 also stimulated the translation of an FGF23 cDNA transfected into osteoblasts via a FGFR1 and PI3K/Akt-dependent mechanism. Thus, activation of autocrine/paracrine FGF pathways is involved in the pathogenesis of Hyp through FGFR1-dependent regulation of FGF23 by both transcriptional and post-transcriptional mechanisms. This may serve to link local bone metabolism with systemic phosphate and vitamin D homeostasis.


FEBS Letters | 2016

Counter-regulatory paracrine actions of FGF-23 and 1,25(OH)2D in macrophages

Xiaobin Han; Linqiang Li; Jiancheng Yang; Gwendalyn D. King; Zhousheng Xiao; Leigh Darryl Quarles

Mechanisms underlying the association between fibroblastic growth factor 23 (FGF‐23) and inflammation are uncertain. We found that FGF‐23 was markedly up‐regulated in LPS/INF‐γ‐induced proinflammatory M1 macrophages and Hyp mouse‐derived peritoneal macrophages, but not in IL‐4‐induced M2 anti‐inflammatory macrophages. NF‐КB and JAK/STAT1 pathways mediated the increased transcription of FGF‐23 in response to M1 polarization. FGF‐23 stimulated TNF‐α, but not IL‐6, expression in M0 macrophages and suppressed Arginase‐1 expression in M2 macrophages through FGFR‐mediated mechanisms. 1,25(OH)2D stimulated Arginase‐1 expression and inhibited FGF‐23 stimulation of TNF‐α. FGF‐23 has proinflammatory paracrine functions and counter‐regulatory actions to 1,25(OH)2D on innate immune responses.


Journal of Biological Chemistry | 2015

Membrane and Integrative Nuclear Fibroblastic Growth Factor Receptor (FGFR) Regulation of FGF-23

Xiaobin Han; Zhousheng Xiao; L. Darryl Quarles

Background: Mechanisms whereby local bone-derived factors regulate FGF-23 are unclear. Results: Low and high molecular weight FGF-2 stimulated FGF-23 promoter activity in osteoblasts through membrane FGFRmediated PLC and MAPK activation of NFAT and Ets1 and integrative nuclear FGFR1 signaling involving cAMP/CBP/CREB signaling pathways, respectively. Conclusion: Membrane FGFR and intranucelar FGFR/CBP pathways regulate FGF-23 transcription. Significance: Paracrine/autocrine FGFs control hormonal FGF-23.


PLOS ONE | 2016

Conditional Deletion of Fgfr1 in the Proximal and Distal Tubule Identifies Distinct Roles in Phosphate and Calcium Transport

Xiaobin Han; Jiancheng Yang; Linqiang Li; Jinsong Huang; Gwendalyn D. King; L. Darryl Quarles

A postnatal role of fibroblast growth factor receptor-1 (FGFR1) in the kidney is suggested by its binding to α-Klotho to form an obligate receptor for the hormone fibroblast growth factor-23 (FGF-23). FGFR1 is expressed in both the proximal and distal renal tubular segments, but its tubular specific functions are unclear. In this study, we crossed Fgfr1flox/flox mice with either gamma-glutamyltransferase-Cre (γGT-Cre) or kidney specific-Cre (Ksp-Cre) mice to selectively create proximal tubule (PT) and distal tubule (DT) Fgfr1 conditional knockout mice (designated Fgfr1PT-cKO and Fgfr1DT-cKO, respectively). Fgfr1PT-cKO mice exhibited an increase in sodium-dependent phosphate co-transporter expression, hyperphosphatemia, and refractoriness to the phosphaturic actions of FGF-23, consistent with a direct role of FGFR1 in mediating the proximal tubular phosphate responses to FGF-23. In contrast, Fgfr1DT-cKO mice unexpectedly developed hypercalciuria, secondary elevations of parathyroid hormone (PTH), hypophosphatemia and enhanced urinary phosphate excretion. Fgfr1PT-cKO mice also developed a curly tail/spina bifida-like skeletal phenotype, whereas Fgfr1DT-cKO mice developed renal tubular micro-calcifications and reductions in cortical bone thickness. Thus, FGFR1 has dual functions to directly regulate proximal and distal tubule phosphate and calcium reabsorption, indicating a physiological role of FGFR1 signaling in both phosphate and calcium homeostasis.


Journal of Biological Chemistry | 2015

Membrane and Integrative Nuclear FGFR Regulation of Hormonal FGF-23

Xiaobin Han; Zhousheng Xiao; L. Darryl Quarles

Background: Mechanisms whereby local bone-derived factors regulate FGF-23 are unclear. Results: Low and high molecular weight FGF-2 stimulated FGF-23 promoter activity in osteoblasts through membrane FGFR-mediated PLCγ and MAPK activation of NFAT and Ets1 and integrative nuclear FGFR1 signaling involving cAMP/CBP/CREB signaling pathways, respectively. Conclusion: Membrane FGFR and intranucelar FGFR/CBP pathways regulate FGF-23 transcription. Significance: Paracrine/autocrine FGFs control hormonal FGF-23. Fibroblastic growth factor receptor 1 (FGFR1) signaling pathways are implicated in the regulation of FGF-23 gene transcription, but the molecular pathways remain poorly defined. We used low molecular weight (LMW, 18 kDa) FGF-2 and high molecular weight (HMW) FGF-2 isoforms, which, respectively, activate cell surface FGF receptors and intranuclear FGFR1, to determine the roles of membrane FGFRs and integrative nuclear FGFR1 signaling (INFS) in the regulation of FGF-23 gene transcription in osteoblasts. We found that LMW-FGF-2 induced NFAT and Ets1 binding to conserved cis-elements in the proximal FGF-23 promoter and stimulated FGF-23 promoter activity through PLCγ/calcineurin/NFAT and MAPK pathways in SaOS-2 and MC3T3-E1 osteoblasts. In contrast, HMW-FGF-2 stimulated FGF-23 promoter activity in osteoblasts through a cAMP-dependent binding of FGFR1 and cAMP-response element-binding protein (CREB) to a conserved cAMP response element (CRE) contiguous with the NFAT binding site in the FGF-23 promoter. Mutagenesis of the NFAT and CRE binding sites, respectively, inhibited the effects of LMW-FGF-2 and HMW-FGF-23 to stimulate FGF-23 promoter activity. FGF-2 activation of both membrane FGFRs and INFS-dependent FGFR1 pathways may provide a means to integrate systemic and local regulation of FGF-23 transcription under diverse physiological and pathological conditions.


Bone research | 2016

The hypoxia-inducible factor-1α activates ectopic production of fibroblast growth factor 23 in tumor-induced osteomalacia.

Qian Zhang; Michele Doucet; Ryan E. Tomlinson; Xiaobin Han; L Darryl Quarles; Michael T. Collins; Thomas L. Clemens

Tumor-induced osteomalacia (TIO) is a rare paraneoplastic syndrome in which ectopic production of fibroblast growth factor 23 (FGF23) by non-malignant mesenchymal tumors causes phosphate wasting and bone fractures. Recent studies have implicated the hypoxia-inducible factor-1α (HIF-1α) in other phosphate wasting disorders caused by elevated FGF23, including X-linked hypophosphatemic rickets and autosomal dominant hypophosphatemia. Here we provide evidence that HIF-1α mediates aberrant FGF23 in TIO by transcriptionally activating its promoter. Immunohistochemical studies in phosphaturic mesenchymal tumors resected from patients with documented TIO showed that HIF-1α and FGF23 were co-localized in spindle-shaped cells adjacent to blood vessels. Cultured tumor tissue produced high levels of intact FGF23 and demonstrated increased expression of HIF-1α protein. Transfection of MC3T3-E1 and Saos-2 cells with a HIF-1α expression construct induced the activity of a FGF23 reporter construct. Prior treatment of tumor organ cultures with HIF-1α inhibitors decreased HIF-1α and FGF23 protein accumulation and inhibited HIF-1α-induced luciferase reporter activity in transfected cells. Chromatin immunoprecipitation assays confirmed binding to a HIF-1α consensus sequence within the proximal FGF23 promoter, which was eliminated by treatment with a HIF-1α inhibitor. These results show for the first time that HIF-1α is a direct transcriptional activator of FGF23 and suggest that upregulation of HIF-1α activity in TIO contributes to the aberrant FGF23 production in these patients.


Current Opinion in Nephrology and Hypertension | 2016

Multiple faces of fibroblast growth factor-23

Xiaobin Han; L. Darryl Quarles

Purpose of reviewThis review examines the role of fibroblast growth factor-23 (FGF-23) in mineral metabolism, innate immunity and adverse cardiovascular outcomes. Recent findingsFGF-23, produced by osteocytes in bone, activates FGFR/&agr;-Klotho (&agr;-Kl) complexes in the kidney. The resulting bone–kidney axis coordinates renal phosphate reabsorption with bone mineralization, and creates a counter-regulatory feedback loop to prevent vitamin D toxicity. FGF-23 acts to counter-regulate the effects of vitamin D on innate immunity and cardiovascular responses. FGF-23 is ectopically expressed along with &agr;-Kl in activated macrophages, creating a proinflammatory paracrine signaling pathway that counters the antiinflammatory actions of vitamin D. FGF-23 also inhibits angiotensin-converting enzyme 2 expression and increases sodium reabsorption in the kidney, leading to hypertension and left ventricular hypertrophy. Finally, FGF-23 is purported to cause adverse cardiac and impair neutrophil responses through activation of FGFRs in the absence of &agr;-Kl. Although secreted forms of &agr;-Kl have FGF-23 independent effects, the possibility of &agr;-Kl independent effects of FGF-23 is controversial and requires additional experimental validation. SummaryFGF-23 participates in a bone–kidney axis regulating mineral homeostasis, proinflammatory paracrine macrophage signaling pathways, and in a bone–cardio–renal axis regulating hemodynamics that counteract the effects of vitamin D.


Journal of The American Society of Nephrology | 2017

Cardiovascular Effects of Renal Distal Tubule Deletion of the FGF Receptor 1 Gene

Xiaobin Han; Jed Ross; Ganesh Kolumam; Min Pi; Junichiro Sonoda; Gwendalyn D. King; L. Darryl Quarles

The bone-derived hormone fibroblast growth factor-23 (FGF-23) activates complexes composed of FGF receptors (FGFRs), including FGFR1, and α-Klotho in the kidney distal tubule (DT), leading to increased sodium retention and hypertension. However, the role of FGFR1 in regulating renal processes linked to hypertension is unclear. Here, we investigated the effects of selective FGFR1 loss in the DT. Conditional knockout (cKO) of FGFR1 in the DT (FGFR1DT-cKO mice) resulted in left ventricular hypertrophy (LVH) and decreased kidney expression of α-Klotho in association with enhanced BP, decreased expression of angiotensin converting enzyme 2, and increased expression of the Na+-K+-2Cl- cotransporter. Notably, recombinant FGF-23 administration similarly decreased the kidney expression of α-Klotho and induced LVH in mice. Pharmacologic activation of FGFR1 with a monoclonal anti-FGFR1 antibody (R1MAb1) normalized BP and significantly attenuated LVH in the Hyp mouse model of excess FGF-23, but did not induce a response in FGFR1DT-cKO mice. The hearts of FGFR1DT-cKO mice showed increased expression of the transient receptor potential cation channel, subfamily C, member 6 (TRPC6), consistent with cardiac effects of soluble Klotho deficiency. Moreover, administration of recombinant soluble Klotho lowered BP in the Hyp mice. Thus, FGFR1 in the DT regulates systemic hemodynamic responses opposite to those predicted by the actions of FGF-23. These cardiovascular effects appear to be mediated by paracrine FGF control of kidney FGFR1 and subsequent regulation of soluble Klotho and TRPC6. FGFR1 in the kidney may provide a new molecular target for treating hypertension.


Scientific Reports | 2018

Cardiovascular Interactions between Fibroblast Growth Factor-23 and Angiotensin II

Min Pi; Xiaobin Han; Benjamin Armstrong; Xue Liu; Yuanjian Chen; Yao Sun; L. Darryl Quarles

Both the activation of the renin angiotensin aldosterone system (RAAS) and elevations of circulating Fibroblast Growth Factor-23 (FGF-23) have been implicated in the pathogenesis of left ventricular hypertrophy (LVH) in chronic kidney disease. To investigate potential cross-talk between RAAS and FGF-23, we administered angiotensin II (Ang II) to wild-type rodents and the Hyp mouse model of excess FGF-23. Ang II administration for four weeks to wild-type rodents resulted in significant increases in systolic blood pressure and LVH. Unexpectedly, FGF-23 circulating levels were increased by 1.5–1.7 fold in Ang II treated animals. In addition, Ang II treatment increased expression of FGF-23 message levels in bone, the predominant tissue for FGF-23 production, and induced expression of FGF-23 and its co-receptor α-Klotho in the heart, which normally does not express FGF-23 or α-Klotho in physiologically relevant levels. Hyp mice with elevated FGF-23 exhibited increased blood pressure and LVH at baseline. Ang II administration to Hyp mice resulted further increments in blood pressure and left ventricular hypertrophy, consistent with additive cardiovascular effects. These findings suggest that FGF-23 may participate in unexpected systemic and paracrine networks regulating hemodynamic and myocardial responses.

Collaboration


Dive into the Xiaobin Han's collaboration.

Top Co-Authors

Avatar

L. Darryl Quarles

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Zhousheng Xiao

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Gwendalyn D. King

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jinsong Huang

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Jiancheng Yang

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Leigh Darryl Quarles

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Linqiang Li

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Min Pi

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Aline Martin

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Bing Dai

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge