Network


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

Hotspot


Dive into the research topics where Huaxian Ma is active.

Publication


Featured researches published by Huaxian Ma.


Toxicology and Applied Pharmacology | 2009

Increased Nitration and Carbonylation of Proteins in MRL +/+ Mice Exposed to Trichloroethene: Potential Role of Protein Oxidation in Autoimmunity

Gangduo Wang; Jianling Wang; Huaxian Ma; M. Firoze Khan

Even though reactive oxygen and nitrogen species (RONS) are implicated as mediators of autoimmune diseases (ADs), little is known about contribution of protein oxidation (carbonylation and nitration) in the pathogenesis of such diseases. The focus of this study was, therefore, to establish a link between protein oxidation and induction and/or exacerbation of autoimmunity. To achieve this, female MRL +/+ mice were treated with trichloroethene (TCE), an environmental contaminant known to induce autoimmune response, for 6 or 12 weeks (10 mmol/kg, i.p., every 4(th) day). TCE treatment resulted in significantly increased formation of nitrotyrosine (NT) and induction of iNOS in the serum at both 6 and 12 weeks of treatment, but the response was greater at 12 weeks. Likewise, TCE treatment led to greater NT formation, and iNOS protein and mRNA expression in the livers and kidneys. Moreover, TCE treatment also caused significant increases ( approximately 3 fold) in serum protein carbonyls (a marker of protein oxidation) at both 6 and 12 weeks. Significantly increased protein carbonyls were also observed in the livers and kidneys (2.1 and 1.3 fold, respectively) at 6 weeks, and to a greater extent at 12 weeks (3.5 and 2.1 fold, respectively) following TCE treatment. The increases in TCE-induced protein oxidation (carbonylation and nitration) were associated with significant increases in Th1 specific cytokine (IL-2, IFN-gamma) release into splenocyte cultures. These results suggest an association between protein oxidation and induction/exacerbation of autoimmune response. The results present a potential mechanism by which oxidatively modified proteins could contribute to TCE-induced autoimmune response and necessitates further investigations for clearly establishing the role of protein oxidation in the pathogenesis of ADs.


Toxicology and Applied Pharmacology | 2013

N-Acetylcysteine protects against trichloroethene-mediated autoimmunity by attenuating oxidative stress.

Gangduo Wang; Jianling Wang; Huaxian Ma; G.A.S. Ansari; M. Firoze Khan

Exposure to trichloroethene (TCE), a ubiquitous environmental contaminant, is known to induce autoimmunity both in humans and animal models. However, mechanisms underlying TCE-mediated autoimmunity remain largely unknown. Previous studies from our laboratory in MRL+/+ mice suggest that oxidative stress may contribute to TCE-induced autoimmune response. The current study was undertaken to further assess the role of oxidative stress in TCE-induced autoimmunity by supplementing with an antioxidant N-acetylcysteine (NAC). Groups of female MRL+/+ mice were given TCE, NAC or TCE+NAC for 6 weeks (TCE, 10mmol/kg, i.p., every 4th day; NAC, 250mg/kg/day through drinking water). TCE exposure led to significant increases in serum levels of anti-nuclear, anti-dsDNA and anti-Sm antibodies. TCE exposure also led to significant induction of anti-malondiadelhyde (MDA)- and anti-hydroxynonenal (HNE)-protein adduct antibodies which were associated with increased ANA in the sera along with increased MDA-/HNE-protein adducts in the livers and kidneys, and increases in protein oxidation (carbonylation) in the sera, livers and kidneys, suggesting an overall increase in oxidative stress. Moreover, TCE exposure also resulted in increased release of IL-17 from splenocytes and increases in IL-17 mRNA expression. Remarkably, NAC supplementation attenuated not only the TCE-induced oxidative stress, IL-17 release and mRNA expression, but also the markers of autoimmunity, as evident from decreased levels of ANA, anti-dsDNA and anti-Sm antibodies in the sera. These results provide further support to a role of oxidative stress in TCE-induced autoimmune response. Attenuation of TCE-induced autoimmunity in mice by NAC provides an approach for preventive and/or therapeutic strategies.


Free Radical Biology and Medicine | 2010

Up-regulation of heme oxygenase-1 in rat spleen after aniline exposure.

Jianling Wang; Huaxian Ma; Paul J. Boor; V. M. Sadagopa Ramanujam; G.A.S. Ansari; M. Firoze Khan

The splenic toxicity of aniline is characterized by vascular congestion, hyperplasia, fibrosis, and the development of a variety of sarcomas in rats. However, the underlying mechanisms by which aniline elicits splenotoxic response are not well understood. Previously we have shown that aniline exposure causes oxidative damage to the spleen. To further explore the oxidative mechanism of aniline toxicity, we evaluated the potential contribution of heme oxygenase-1 (HO-1), which catalyzes heme degradation and releases free iron. Male SD rats were given 1 mmol/kg/day aniline in water by gavage for 1, 4, or 7 days, and respective controls received water only. Aniline exposure led to significant increases in HO-1 mRNA expression in the spleen (2-and 2.4-fold at days 4 and 7, respectively) with corresponding increases in protein expression, as confirmed by ELISA and Western blot analysis. Furthermore, immunohistochemical assessment of spleen showed stronger immunostaining for HO-1 in the spleens of rats treated for 7 days, confined mainly to the red pulp areas. No changes were observed in mRNA and protein levels of HO-1 after 1 day exposure. The increase in HO-1 expression was associated with increases in total iron (2.4-and 2.7-fold), free iron (1.9-and 3.5-fold), and ferritin levels (1.9-and 2.1-fold) at 4 and 7 days of aniline exposure. Our data suggest that HO-1 up-regulation in aniline-induced splenic toxicity could be a contributing pro-oxidant mechanism, mediated through iron release, and leading to oxidative damage.


Toxicology and Applied Pharmacology | 2011

Enhanced expression of cyclins and cyclin-dependent kinases in aniline-induced cell proliferation in rat spleen

Jianling Wang; Gangduo Wang; Huaxian Ma; M. Firoze Khan

Aniline exposure is associated with toxicity to the spleen leading to splenomegaly, hyperplasia, fibrosis and a variety of sarcomas of the spleen on chronic exposure. In earlier studies, we have shown that aniline exposure leads to iron overload, oxidative stress and activation of redox-sensitive transcription factors, which could regulate various genes leading to a tumorigenic response in the spleen. However, molecular mechanisms leading to aniline-induced cellular proliferation in the spleen remain largely unknown. This study was, therefore, undertaken on the regulation of G1 phase cell cycle proteins (cyclins), expression of cyclin-dependent kinases (CDKs), phosphorylation of retinoblastoma protein (pRB) and cell proliferation in the spleen, in an experimental condition preceding a tumorigenic response. Male SD rats were treated with aniline (0.5 mmol/kg/day via drinking water) for 30 days (controls received drinking water only), and splenocyte proliferation, protein expression of G1 phase cyclins, CDKs and pRB were measured. Aniline treatment resulted in significant increases in splenocyte proliferation, based on cell counts, cell proliferation markers including proliferating cell nuclear antigen (PCNA), nuclear Ki67 protein (Ki67) and minichromosome maintenance (MCM), MTT assay and flow cytometric analysis. Western blot analysis of splenocyte proteins from aniline-treated rats showed significantly increased expression of cyclins D1, D2, D3 and E, as compared to the controls. Similarly, real-time PCR analysis showed significantly increased mRNA expression for cyclins D1, D2, D3 and E in the spleens of aniline-treated rats. The overexpression of these cyclins was associated with increases in the expression of CDK4, CDK6, CDK2 as well as phosphorylation of pRB protein. Our data suggest that increased expression of cyclins, CDKs and phosphorylation of pRB protein could be critical in cell proliferation, and may contribute to aniline-induced tumorigenic response in the spleen.


Free Radical Biology and Medicine | 2010

Original ContributionUp-regulation of heme oxygenase-1 in rat spleen after aniline exposure

Jianling Wang; Huaxian Ma; Paul J. Boor; V. M. Sadagopa Ramanujam; G.A.S. Ansari; M. Firoze Khan

The splenic toxicity of aniline is characterized by vascular congestion, hyperplasia, fibrosis, and the development of a variety of sarcomas in rats. However, the underlying mechanisms by which aniline elicits splenotoxic response are not well understood. Previously we have shown that aniline exposure causes oxidative damage to the spleen. To further explore the oxidative mechanism of aniline toxicity, we evaluated the potential contribution of heme oxygenase-1 (HO-1), which catalyzes heme degradation and releases free iron. Male SD rats were given 1 mmol/kg/day aniline in water by gavage for 1, 4, or 7 days, and respective controls received water only. Aniline exposure led to significant increases in HO-1 mRNA expression in the spleen (2-and 2.4-fold at days 4 and 7, respectively) with corresponding increases in protein expression, as confirmed by ELISA and Western blot analysis. Furthermore, immunohistochemical assessment of spleen showed stronger immunostaining for HO-1 in the spleens of rats treated for 7 days, confined mainly to the red pulp areas. No changes were observed in mRNA and protein levels of HO-1 after 1 day exposure. The increase in HO-1 expression was associated with increases in total iron (2.4-and 2.7-fold), free iron (1.9-and 3.5-fold), and ferritin levels (1.9-and 2.1-fold) at 4 and 7 days of aniline exposure. Our data suggest that HO-1 up-regulation in aniline-induced splenic toxicity could be a contributing pro-oxidant mechanism, mediated through iron release, and leading to oxidative damage.


Toxicology and Applied Pharmacology | 2013

Induction of base excision repair enzymes NTH1 and APE1 in rat spleen following aniline exposure

Huaxian Ma; Jianling Wang; Sherif Z. Abdel-Rahman; Paul J. Boor; M. Firoze Khan

Mechanisms by which aniline exposure elicits splenotoxicity, especially a tumorigenic response, are not well-understood. Earlier, we have shown that aniline exposure leads to oxidative DNA damage and up-regulation of OGG1 and NEIL1/2 DNA glycosylases in rat spleen. However, the contribution of endonuclease III homolog 1 (NTH1) and apurinic/apyrimidinic endonuclease 1 (APE1) in the repair of aniline-induced oxidative DNA damage in the spleen is not known. This study was, therefore, focused on examining whether NTH1 and APE1 contribute to the repair of oxidative DNA lesions in the spleen, in an experimental condition preceding tumorigenesis. To achieve this, male SD rats were subchronically exposed to aniline (0.5 mmol/kg/day via drinking water for 30 days), while controls received drinking water only. By quantitating the cleavage products, the activities of NTH1 and APE1 were assayed using substrates containing thymine glycol (Tg) and tetrahydrofuran, respectively. Aniline treatment led to significant increases in NTH1- and APE1-mediated BER activity in the nuclear extracts of spleen of aniline-treated rats compared to the controls. NTH1 and APE1 mRNA expression in the spleen showed 2.9- and 3.2-fold increases, respectively, in aniline-treated rats compared to the controls. Likewise, Western blot analysis showed that protein expression of NTH1 and APE1 in the nuclear extracts of spleen from aniline-treated rats was 1.9- and 2.7-fold higher than the controls, respectively. Immunohistochemistry indicated that aniline treatment also led to stronger immunoreactivity for both NTH1 and APE1 in the spleens, confined to the red pulp areas. These results, thus, show that aniline exposure is associated with induction of NTH1 and APE1 in the spleen. The increased repair activity of NTH1 and APE1 could be an important mechanism for the removal of oxidative DNA lesions. These findings thus identify a novel mechanism through which NTH1 and APE1 may regulate the repair of oxidative DNA damage in aniline-induced splenic toxicity.


Toxicology and Applied Pharmacology | 2019

Contribution of poly(ADP-ribose)polymerase-1 activation and apoptosis in trichloroethene-mediated autoimmunity

Gangduo Wang; Huaxian Ma; Jianling Wang; M. Firoze Khan

ABSTRACT Trichloroethene (TCE), a common environmental toxicant and widely used industrial solvent, has been implicated in the development of various autoimmune diseases (ADs). Although oxidative stress has been involved in TCE‐mediated autoimmunity, the molecular mechanisms remain to be fully elucidated. These studies were, therefore, aimed to further explore the contribution of oxidative stress to TCE‐mediated autoimmune response by specifically assessing the role of oxidative DNA damage, its repair enzyme poly(ADP‐ribose)polymerase‐1 (PARP‐1) and apoptosis. To achieve this, groups of female MRL +/+ mice were treated with TCE, TCE plus N‐acetylcysteine (NAC) or NAC alone (TCE, 10mmol/kg, i.p., every 4th day; NAC, 250mg/kg/day in drinking water) for 6weeks. TCE treatment led to significantly higher levels of 8‐hydroxy‐2′‐deoxyguanosine (8‐OHdG) in the livers compared to controls, suggesting increased oxidative DNA damage. TCE‐induced DNA damage was associated with significant activation of PARP‐1 and increases in caspase‐3, cleaved caspase‐8 and ‐9, and alterations in Bcl‐2 and Bax in the livers. Moreover, the TCE‐mediated alterations corresponded with remarkable increases in the serum anti‐ssDNA antibodies. Interestingly, NAC supplementation not only attenuated elevated 8‐OHdG, PARP‐1, caspase‐3, cleaved caspase‐9, and Bax, but also the TCE‐mediated autoimmune response supported by significantly reduced serum anti‐ssDNA antibodies. These results suggest that TCE‐induced activation of PARP‐1 followed by increased apoptosis presents a novel mechanism in TCE‐associated autoimmune response and could potentially lead to development of targeted preventive and/or therapeutic strategies. HIGHLIGHTSTCE exposure resulted in increased DNA oxidation and activation of PARP‐1.TCE led to increased apoptosis evidenced by changes in caspases and Bcl‐2 family.TCE treatment resulted in significant increases in anti‐ssDNA antibodies in sera.NAC attenuated changes in 8‐OHdG, PARP‐1, caspases, and also the autoantibodiesApoptosis via activation of PARP‐1 contributed to TCE‐mediated autoimmunity


Toxicology and Applied Pharmacology | 2008

Oxidative DNA damage and its repair in rat spleen following subchronic exposure to aniline

Huaxian Ma; Jianling Wang; Sherif Z. Abdel-Rahman; Paul J. Boor; M. Firoze Khan


Toxicology and Applied Pharmacology | 2011

Induction of NEIL1 and NEIL2 DNA glycosylases in aniline-induced splenic toxicity.

Huaxian Ma; Jianling Wang; Sherif Z. Abdel-Rahman; Tapas K. Hazra; Paul J. Boor; M. Firoze Khan


Free Radical Biology and Medicine | 2010

Oxidative Stress and Trichloroethene-Mediated Autoimmunity

Gangduo Wang; Huaxian Ma; Xiuzhen Fan; Jianling Wang; G.A.S. Ansari; M. Firoze Khan

Collaboration


Dive into the Huaxian Ma's collaboration.

Top Co-Authors

Avatar

Jianling Wang

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

M. Firoze Khan

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Paul J. Boor

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Gangduo Wang

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

G.A.S. Ansari

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Sherif Z. Abdel-Rahman

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

V. M. Sadagopa Ramanujam

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

G. A. Shakeel Ansari

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Tapas K. Hazra

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Xiuzhen Fan

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge