Network


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

Hotspot


Dive into the research topics where Yava L. Jones-Hall is active.

Publication


Featured researches published by Yava L. Jones-Hall.


Cancer Research | 2013

Chemopreventive activity of plant flavonoid isorhamnetin in colorectal cancer is mediated by oncogenic Src and β-catenin

Shakir M. Saud; Matthew R. Young; Yava L. Jones-Hall; Lilia Ileva; Moses O. Evbuomwan; Jennifer Wise; Nancy H. Colburn; Young Seok Kim; Gerd Bobe

Analysis of the Polyp Prevention Trial showed an association between an isorhamnetin-rich diet and a reduced risk of advanced adenoma recurrence; however, the mechanism behind the chemoprotective effects of isorhamnetin remains unclear. Here, we show that isorhamnetin prevents colorectal tumorigenesis of FVB/N mice treated with the chemical carcinogen azoxymethane and subsequently exposed to colonic irritant dextran sodium sulfate (DSS). Dietary isorhamnetin decreased mortality, tumor number, and tumor burden by 62%, 35%, and 59%, respectively. MRI, histopathology, and immunohistochemical analysis revealed that dietary isorhamnetin resolved the DSS-induced inflammatory response faster than the control diet. Isorhamnetin inhibited AOM/DSS-induced oncogenic c-Src activation and β-catenin nuclear translocation, while promoting the expression of C-terminal Src kinase (CSK), a negative regulator of Src family of tyrosine kinases. Similarly, in HT-29 colon cancer cells, isorhamnetin inhibited oncogenic Src activity and β-catenin nuclear translocation by inducing expression of csk, as verified by RNA interference knockdown of csk. Our observations suggest the chemoprotective effects of isorhamnetin in colon cancer are linked to its anti-inflammatory activities and its inhibition of oncogenic Src activity and consequential loss of nuclear β-catenin, activities that are dependent on CSK expression.


Pathophysiology | 2014

Immunopathological characterization of selected mouse models of inflammatory bowel disease: Comparison to human disease

Yava L. Jones-Hall; Matthew B. Grisham

Inflammatory bowel diseases (IBD) are chronic, relapsing conditions of multifactorial etiology. The two primary diseases of IBD are Crohns disease (CD) and ulcerative colitis (UC). Both entities are hypothesized to occur in genetically susceptible individuals due to microbial alterations and environmental contributions. The exact etiopathogenesis, however, is not known for either disease. A variety of mouse models of CD and UC have been developed to investigate the pathogenesis of these diseases and evaluate treatment modalities. Broadly speaking, the mouse models can be divided into 4 categories: genetically engineered, immune manipulated, spontaneous and erosive/chemically induced. No one mouse model completely recapitulates the immunopathology of CD or UC, however each model possesses particular similarities to human IBD and offers advantageous for specific details of IBD pathogenesis. Here we discuss the more commonly used models in each category and critically evaluate how the immunopathology induced compares to CD or UC, as well as the advantages and disadvantages associated with each model.


PLOS ONE | 2015

Ablation of Tumor Necrosis Factor Is Associated with Decreased Inflammation and Alterations of the Microbiota in a Mouse Model of Inflammatory Bowel Disease

Yava L. Jones-Hall; Ariangela Kozik; Cindy H. Nakatsu

Inflammatory bowel disease (IBD) is associated with prolonged, excess secretions of Tumor Necrosis Factor (TNF). Many patients with IBD have successful management of IBD symptoms by blocking TNF secretion or signaling. However, some patients are non-responsive to this therapy, eventually become refractory to therapy, or Alterations in the microbiota that are associated with the lack of TNF could be a contributing cause of this therapeutic insufficiency seen in some patients. Here we use wildtype (WT) and mice lacking Tnf (Tnf -/-) in an acute TNBS colitis model to investigate the role of TNF in colitis and how its presence or absence affects the colonic microbiota. As expected, Tnf -/- had less severe inflammation than WT mice. Microbiome analysis revealed significant Tnf dependent-differences in alpha and beta diversity. There were also notable differences in many species that were also primarily Tnf dependent. Taken together, our data indicates that TNF contributes significantly to the inflammation and microbiotal alterations in that occur in IBD.


Inflammatory Bowel Diseases | 2015

Use of Humanized Mice to Study the Pathogenesis of Autoimmune and Inflammatory Diseases

Iurii Koboziev; Yava L. Jones-Hall; John F. Valentine; Cynthia Reinoso Webb; Kathryn L. Furr; Matthew B. Grisham

Abstract:Animal models of disease have been used extensively by the research community for the past several decades to better understand the pathogenesis of different diseases and assess the efficacy and toxicity of different therapeutic agents. Retrospective analyses of numerous preclinical intervention studies using mouse models of acute and chronic inflammatory diseases reveal a generalized failure to translate promising interventions or therapeutics into clinically effective treatments in patients. Although several possible reasons have been suggested to account for this generalized failure to translate therapeutic efficacy from the laboratory bench to the patients bedside, it is becoming increasingly apparent that the mouse immune system is substantially different from the human. Indeed, it is well known that >80 major differences exist between mouse and human immunology; all of which contribute to significant differences in immune system development, activation, and responses to challenges in innate and adaptive immunity. This inconvenient reality has prompted investigators to attempt to humanize the mouse immune system to address important human-specific questions that are impossible to study in patients. The successful long-term engraftment of human hematolymphoid cells in mice would provide investigators with a relatively inexpensive small animal model to study clinically relevant mechanisms and facilitate the evaluation of human-specific therapies in vivo. The discovery that targeted mutation of the IL-2 receptor common gamma chain in lymphopenic mice allows for the long-term engraftment of functional human immune cells has advanced greatly our ability to humanize the mouse immune system. The objective of this review is to present a brief overview of the recent advances that have been made in the development and use of humanized mice with special emphasis on autoimmune and chronic inflammatory diseases. In addition, we discuss the use of these unique mouse models to define the human-specific immunopathological mechanisms responsible for the induction and perpetuation of chronic gut inflammation.


Scientific Reports | 2015

Epidermal Growth Factor Receptor (EGFR) Pathway Genes and Interstitial Lung Disease: An Association Study

Chong Li; Rongrong Wei; Yava L. Jones-Hall; Ragini Vittal; Min Zhang; Wanqing Liu

The etiology and pathogenesis of idiopathic interstitial lung disease (ILD) remain incompletely understood. Genetic susceptibility to ILD has been demonstrated in previous studies. It is well known that EGFR inhibitors can induce ILD in human lung cancer patient with ethnic differences, which prompted us to hypothesize that genetic variation in EGFR pathway genes confer susceptibility to ILD. We aimed in this study to investigate whether functional polymorphisms of EGFR and its ligands genes (EGF and TGFA) were associated with ILD. Three EGFR [−216G/T (rs712830), −191A/C (rs712829), 497R > K(A/G) (rs2227983)], one EGF [61A/G, (rs4444903)] and one TGFA (rs3821262C/T) polymorphisms previously demonstrated to alter gene functions were genotyped in 229 sporadic idiopathic ILD patients and 693 normal healthy individuals. Allelic and genotypic association tests between these polymorphisms and ILD were performed. The EGF 61A/G polymorphism was significantly associated with elevated risk of ILD, with the frequency of G allele significantly increased in the ILD patient population (OR = 1.33, 95%CI = 1.07–1.66, P = 0.0099). None of the other polymorphisms were associated with risk of ILD. Our study suggested that the EGF 61A/G polymorphism may be associated with sporadic ILD. While a false positive finding cannot be excluded, independent studies are warranted to further validate this result.


Vascular Pharmacology | 2016

Deletion of P2Y2 receptor reveals a role for lymphotoxin-α in fatty streak formation

Shaomin Qian; April M. Hoggatt; Yava L. Jones-Hall; Carl F. Ware; Paul B. Herring; Cheikh I. Seye

Background Lymphotoxin alpha (LTα) is expressed in human atherosclerotic lesions and genetic variations in the LTα pathway have been linked to myocardial infarction. Activation of the P2Y2 nucleotide receptor (P2Y2R) regulates the production of LTα. in vitro. We aimed to uncover a potential pathway linking purinergic receptor to LTα-mediated inflammatory processes pivotal to the early stages of atherosclerosis in apolipoprotein E (ApoE−/−) deficient mice. Methods and results En face immunostaining revealed that P2Y2R and VCAM-1 are preferentially expressed in the atherosclerosis prone site of the mouse aortic sinus. Deletion of the P2Y2R gene suppresses VCAM-1 expression. Compared with ApoE−/−mice, ApoE−/−mice lacking the P2Y2R gene (ApoE−/−/P2Y2R−/−) did not develop fatty streak lesions when fed a standard chow diet for 15 weeks. Systemic and CD4+ T cell production of the pro-inflammatory cytokine lymphotoxin-alpha (LTα) were specifically inhibited in ApoE−/−/P2Y2R−/− mice. Anti-LTα preventive treatment was initiated in ApoE−/− mice with intraperitoneal administration of recombinant human tumor necrosis factor receptor 1 fusion protein (TNFR1-Fc) on 5 consecutive days before the disease onset. Remarkably, none of the TNFR1:Fc-treated ApoE−/− mice exhibited atherosclerotic lesions at any developmental stage. Significance ApoE−/− mice deficient in P2Y2R exhibit low endothelial cell VCAM-1 levels, decreased production of LTα and delayed onset of atherosclerosis. These data suggest that targeting this nucleotide receptor could be an effective therapeutic approach in atherosclerosis.


Frontiers in Genetics | 2013

Association between hTERT rs2736100 polymorphism and sensitivity to anti-cancer agents

Julie Kim; Yava L. Jones-Hall; Rongrong Wei; Jamie L. Myers; Yuan Qi; Gregory T. Knipp; Wanqing Liu

Background: The rs2736100 single nucleotide polymorphism (SNP) is located in the intron 2 of human telomerase reverse transcriptase (hTERT) gene. Recent genome-wide association studies (GWAS) have consistently supported the strong association between this SNP and risk for multiple cancers. Given the important role of the hTERT gene and this SNP in cancer biology, we hypothesize that rs2736100 may also confer susceptibility to anti-cancer drug sensitivity. In this study we aim to investigate the correlation between the rs2736100 genotype and the responsiveness to anti-cancer agents in the NCI-60 cancer cell panel. Methods and Materials: The hTERT rs2736100 was genotyped in the NCI-60 cancer cell lines. The relative telomere length (RTL) of each cell line was quantified using real-time PCR. The genotype was then correlated with publically available drug sensitivity data of two agents with telomerase-inhibition activity: Geldanamycin (HSP90 inhibitor) and RHPS4/BRACO19 (G-quadruplex stabilizer) as well as additional 110 commonly used agents with established mechanism of action. The association between rs2736100 and mutation status of TP53 gene was also tested. Results: The C allele of the SNP was significantly correlated with increased sensitivity to RHPS4/BRACO19 with an additive effect (r = −0.35, p = 0.009) but not with Geldanamycin. The same allele was also significantly associated with sensitivity to antimitotic agents compared to other agents (p = 0.003). The highest correlation was observed between the SNP and paclitaxel (r = −0.36, p = 0.005). The telomere length was neither associated with rs2736100 nor with sensitivity to anti-cancer agents. The C allele of rs2736100 was significantly associated with increased mutation rate in TP53 gene (p = 0.004). Conclusion: Our data suggested that the cancer risk allele of hTERT rs2736100 polymorphism may also affect the cancer cell response to both TERT inhibitor and anti-mitotic agents, which might be attributed to the elevated telomerase-independent activity of hTERT, as well as the increased risk for TP53 gene mutagenesis conferred by the polymorphism. Detailed mechanisms need to be further investigated.


Veterinary and Comparative Orthopaedics and Traumatology | 2018

In Vivo Evaluation of Biodegradability and Biocompatibility of Fe30Mn Alloy

Marine Traverson; Michael Heiden; Lia Stanciu; Eric A. Nauman; Yava L. Jones-Hall; Gert J. Breur

OBJECTIVES  This study aims to evaluate the biodegradability and biocompatibility of an alloy of iron and manganese (Fe30Mn) in a bone model in vivo. METHODS  Resorption of a Fe30Mn wire was compared with traditional permanent 316L stainless steel (SS) wire after bilateral transcondylar femoral implantation in 12 rats. Evaluation of biodegradation over 6 months was performed using radiography, post-mortem histology and microscopic implant surface analysis. RESULTS  Corrosion and resorption of the novel iron-manganese implant with formation of an iron oxide corrosion layer was noted on all post-mortem histological sections and macroscopic specimens (corrosion fraction of 0.84 and 0 for Fe30Mn and 316L SS, respectively). Increased bone ongrowth was observed at the wire-bone interface (bone ongrowth fraction of 0.61 and 0.34 for Fe30Mn and 316L SS, respectively). Occasionally, poorly stained newly formed bone and necrotic bone in contact with corrosion was seen. In bone marrow, Fe30Mn alloy was scored as a mild local irritant compared with 316L SS (biocompatibility score of 8.8 and 5.3, respectively). There was no evidence of systemic adverse reaction. CLINICAL SIGNIFICANCE  Resorbable iron-manganese alloys may offer a promising alternative to permanent metallic implants. Further in vivo studies to control implant resorption at a rate suitable for fracture healing and to confirm the biocompatibility and biosafety of the resorbable Fe30Mn metallic implant are necessary prior to use in clinical settings.


Gut microbes | 2016

The Intersection of TNF, IBD and the Microbiome.

Yava L. Jones-Hall; Cindy H. Nakatsu

ABSTRACT Inflammatory bowel disease (IBD), comprised of Crohns disease and ulcerative colitis, is a chronic inflammatory condition of multifactorial etiology and risk factors. Currently, one of the most effective treatments for IBD is the use of Tumor Necrosis Factor (TNF) functional inhibitor drugs, however, this treatment can cause adverse reactions and has a relatively large percentage of incomplete or non-responders. This lack of response may be related to differences in patients’ gut microbiomes prior to and after disease initiation or treatment. Recent observations in our lab using a rodent model of IBD support the theory that TNF drives acute colitis, but also that the microbiome differs in association with TNF production and colitis severity. Studies such as this and others provide new insights into host-microbiome interactions associated with colitis that can lead to new therapies to prevent or treat the disease.


Science Translational Medicine | 2018

Clostridioides difficile uses amino acids associated with gut microbial dysbiosis in a subset of patients with diarrhea

Eric J. Battaglioli; Vanessa L. Hale; Jun Chen; Patricio Jeraldo; Coral Ruiz-Mojica; Bradley A. Schmidt; Vayu Maini Rekdal; Lisa M. Till; Lutfi Huq; Samuel A. Smits; William J. Moor; Yava L. Jones-Hall; Thomas C. Smyrk; Sahil Khanna; Darrell S. Pardi; Madhusudan Grover; Robin Patel; Nicholas Chia; Heidi Nelson; Justin L. Sonnenburg; Gianrico Farrugia; Purna C. Kashyap

Increased amino acids in the dysbiotic gut influences susceptibility to Clostridioides difficile infection in mice and humans. To infect or not to infect? Our gut harbors a diverse microbial community that efficiently uses nutrients. Battaglioli et al. now report that a subset of patients with diarrhea show increased availability of gut amino acids due to deleterious changes in the gut microbiota (dysbiosis). These dysbiotic microbial communities when modeled in germ-free mice exhibited increased susceptibility to Clostridioides difficile, a pathogen that uses amino acids as a nutrient source. Prophylactic fecal microbiota transplant from healthy humans to mice with a dysbiotic gut microbiota restored microbial diversity and protected the mice from C. difficile infection. The gut microbiota plays a critical role in pathogen defense. Studies using antibiotic-treated mice reveal mechanisms that increase susceptibility to Clostridioides difficile infection (CDI), but risk factors associated with CDI in humans extend beyond antibiotic use. Here, we studied the dysbiotic gut microbiota of a subset of patients with diarrhea and modeled the gut microbiota of these patients by fecal transplantation into germ-free mice. When challenged with C. difficile, the germ-free mice transplanted with fecal samples from patients with dysbiotic microbial communities showed increased gut amino acid concentrations and greater susceptibility to CDI. A C. difficile mutant that was unable to use proline as an energy source was unable to robustly infect germ-free mice transplanted with a dysbiotic or healthy human gut microbiota. Prophylactic dietary intervention using a low-proline or low-protein diet in germ-free mice colonized by a dysbiotic human gut microbiota resulted in decreased expansion of wild-type C. difficile after challenge, suggesting that amino acid availability might be important for CDI. Furthermore, a prophylactic fecal microbiota transplant in mice with dysbiosis reduced proline availability and protected the mice from CDI. Last, we identified clinical risk factors that could potentially predict gut microbial dysbiosis and thus greater susceptibility to CDI in a retrospective cohort of patients with diarrhea. Identifying at-risk individuals and reducing their susceptibility to CDI through gut microbiota–targeted therapies could be a new approach to preventing C. difficile infection in susceptible patients.

Collaboration


Dive into the Yava L. Jones-Hall's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthew B. Grisham

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cynthia Reinoso Webb

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerd Bobe

Oregon State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge