Network


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

Hotspot


Dive into the research topics where Naoko Hara is active.

Publication


Featured researches published by Naoko Hara.


Journal of Immunology | 2012

Prevention of Virus-Induced Type 1 Diabetes with Antibiotic Therapy

Naoko Hara; Aimon K. Alkanani; Diana Ir; Charles E. Robertson; Brandie D. Wagner; Daniel N. Frank; Danny Zipris

Microbes were hypothesized to play a key role in the progression of type 1 diabetes (T1D). We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced T1D to test the hypothesis that the intestinal microbiota is involved in the mechanism leading to islet destruction. Treating LEW1.WR1 rats with KRV and a combination of trimethoprim and sulfamethoxazole (Sulfatrim) beginning on the day of infection protected the rats from insulitis and T1D. Pyrosequencing of bacterial 16S rRNA and quantitative RT-PCR indicated that KRV infection resulted in a transient increase in the abundance of Bifidobacterium spp. and Clostridium spp. in fecal samples from day 5- but not day 12-infected versus uninfected animals. Similar alterations in the gut microbiome were observed in the jejunum of infected animals on day 5. Treatment with Sulfatrim restored the level of intestinal Bifidobacterium spp. and Clostridium spp. We also observed that virus infection induced the expression of KRV transcripts and the rapid upregulation of innate immune responses in Peyer’s patches and pancreatic lymph nodes. However, antibiotic therapy reduced the virus-induced inflammation as reflected by the presence of lower amounts of proinflammatory molecules in both the Peyer’s patches and pancreatic lymph nodes. Finally, Sulfatrim treatment reduced the number of B cells in Peyer’s patches and downmodulated adaptive immune responses to KRV, but did not interfere with antiviral Ab responses or viral clearance from the spleen, pancreatic lymph nodes, and serum. The data suggest that gut microbiota may be involved in promoting virus-induced T1D in the LEW1.WR1 rat model.


Diabetes | 2015

Alterations in Intestinal Microbiota Correlate With Susceptibility to Type 1 Diabetes

Aimon K. Alkanani; Naoko Hara; Peter A. Gottlieb; Diana Ir; Charles E. Robertson; Brandie D. Wagner; Daniel N. Frank; Danny Zipris

We tested the hypothesis that alterations in the intestinal microbiota are linked with the progression of type 1 diabetes (T1D). Herein, we present results from a study performed in subjects with islet autoimmunity living in the U.S. High-throughput sequencing of bacterial 16S rRNA genes and adjustment for sex, age, autoantibody presence, and HLA indicated that the gut microbiomes of seropositive subjects differed from those of autoantibody-free first-degree relatives (FDRs) in the abundance of four taxa. Furthermore, subjects with autoantibodies, seronegative FDRs, and new-onset patients had different levels of the Firmicutes genera Lactobacillus and Staphylococcus compared with healthy control subjects with no family history of autoimmunity. Further analysis revealed trends toward increased and reduced abundances of the Bacteroidetes genera Bacteroides and Prevotella, respectively, in seropositive subjects with multiple versus one autoantibody. Canonical discriminant analysis suggested that the gut microbiomes of autoantibody-positive individuals and seronegative FDRs clustered together but separate from those of new-onset patients and unrelated healthy control subjects. Finally, no differences in biodiversity were evident in seropositive versus seronegative FDRs. These observations suggest that altered intestinal microbiota may be associated with disease susceptibility.


Clinical Immunology | 2013

The role of the intestinal microbiota in type 1 diabetes.

Naoko Hara; Aimon K. Alkanani; Diana Ir; Charles E. Robertson; Brandie D. Wagner; Daniel N. Frank; Danny Zipris

The digestive tract hosts trillions of bacteria that interact with the immune system and can influence the balance between pro-inflammatory and regulatory immune responses. Recent studies suggest that alterations in the composition of the intestinal microbiota may be linked with the development of type 1 diabetes (T1D). Data from the biobreeding diabetes prone (BBDP) and the LEW1.WR1 models of T1D support the hypothesis that intestinal bacteria may be involved in early disease mechanisms. The data indicate that cross-talk between the gut microbiota and the innate immune system may be involved in islet destruction. Whether a causal link between intestinal microbiota and T1D exists, the identity of the bacteria and the mechanism whereby they promote the disease remain to be examined. A better understanding of the interplay between microbes and innate immune pathways in early disease stages holds promise for the design of immune interventions and disease prevention in genetically susceptible individuals.


Diabetes | 2014

Induction of Diabetes in the RIP-B7.1 Mouse Model Is Critically Dependent on TLR3 and MyD88 Pathways and Is Associated With Alterations in the Intestinal Microbiome

Aimon K. Alkanani; Naoko Hara; Egil Lien; Diana Ir; Cassandra V. Kotter; Charles E. Robertson; Brandie D. Wagner; Daniel N. Frank; Danny Zipris

RIP-B7.1 transgenic mice express B7.1 costimulatory molecules in pancreatic islets and develop diabetes after treatment with polyinosinic:polycytidylic acid (poly I:C), a synthetic double-stranded RNA and agonist of Toll-like receptor (TLR) 3 and retinoic acid–inducible protein I. We used this model to investigate the role of TLR pathways and intestinal microbiota in disease progression. RIP-B7.1 mice homozygous for targeted disruption of TLR9, TLR3, and myeloid differentiation factor-88 (MyD88), and most of the wild-type RIP-B7.1 mice housed under normal conditions remained diabetes-free after poly I:C administration. However, the majority of TLR9-deficient mice and wild-type animals treated with poly I:C and an antibiotic developed disease. In sharp contrast, TLR3- and MyD88-deficient mice were protected from diabetes following the same treatment regimen. High-throughput DNA sequencing demonstrated that TLR9-deficient mice treated with antibiotics plus poly I:C had higher bacterial diversity compared with disease-resistant mice. Furthermore, principal component analysis suggested that TLR9-deficient mice had distinct gut microbiome compared with the diabetes-resistant mice. Finally, the administration of sulfatrim plus poly I:C to TLR9-deficient mice resulted in alterations in the abundance of gut bacterial communities at the phylum and genus levels. These data imply that the induction of diabetes in the RIP-B7.1 model is critically dependent on TLR3 and MyD88 pathways, and involves modulation of the intestinal microbiota.


Journal of Immunology | 2011

Apolipoprotein A-II Suppressed Concanavalin A-Induced Hepatitis via the Inhibition of CD4 T Cell Function

Junji Yamashita; Chiaki Iwamura; Tetsuya Sasaki; Kunitoshi Mitsumori; Kazutoshi Ohshima; Kaori Hada; Naoko Hara; Munehisa Takahashi; Yoshiaki Kaneshiro; Hitoshi Tanaka; Kenji Kaneko; Toshinori Nakayama

Con A-induced hepatitis has been used as a model of human autoimmune or viral hepatitis. During the process of identifying immunologically bioactive proteins in human plasma, we found that apolipoprotein A-II (ApoA-II), the second major apolipoprotein of high-density lipoprotein, inhibited the production of IFN-γ by Con A-stimulated mouse and human CD4 T cells. Con A-induced hepatitis was attenuated by the administration of ApoA-II. The beneficial effect of ApoA-II was associated with reduced leukocyte infiltration and decreased production of T cell-related cytokines and chemokines in the liver. ApoA-II inhibited the Con A-induced activation of ERK–MAPK and nuclear translocation of NFAT in CD4 T cells. Interestingly, exacerbated hepatitis was observed in ApoA-II–deficient mice, indicating that ApoA-II plays a suppressive role in Con A-induced hepatitis under physiological conditions. Moreover, the administration of ApoA-II after the onset of Con A-induced hepatitis was sufficient to suppress disease. Thus, the therapeutic effect of ApoA-II could be useful for patients with CD4 T cell-related autoimmune and viral hepatitis.


Innate Immunity | 2014

Modulation of virus-induced innate immunity and type 1 diabetes by IL-1 blockade.

Naoko Hara; Aimon K. Alkanani; Charles A. Dinarello; Danny Zipris

We used the LEW1.WR1 model of Kilham rat virus (KRV)-induced type 1 diabetes (T1D) to test the hypothesis that blocking IL-1 pathways early in the course of the disease can modulate virus-induced innate immunity and prevent disease progression. Administering KRV plus IL-1 receptor antagonist (Anakinra) for 14 d prevented insulitis and T1D. Anakinra reversed the KRV-induced systemic inflammation evidenced by the accumulation of T cells in the spleen and pancreatic lymph nodes on d 5 post-infection. Blocking IL-1 modulated the level of IRF-7 and IL-6 gene expression in the spleen and the p40 subunit of IL-12 and IL-23 in the serum. Anakinra did not interfere with the ability of LEW1.WR1 rats to clear the virus from the spleen, pancreatic lymph nodes or serum. Consistent with these data, normal levels of KRV-specific adaptive immune responses were detected in in the spleen and peripheral blood of the treated animals. Finally, blocking IL-1 pathways reversed the KRV-induced modulation of gut bacterial communities. The data may imply that IL-1 pathways are directly linked with early mechanisms whereby KRV infection leads to islet destruction, raising the hypothesis that blocking IL-1 pathways early in the course of the disease could be a useful therapeutic approach for disease prevention.


Virology | 2014

Kilham rat virus-induced type 1 diabetes involves beta cell infection and intra-islet JAK–STAT activation prior to insulitis

Aimon K. Alkanani; Naoko Hara; Roberto Gianani; Danny Zipris

We used the LEW1.WR1 rat model of Kilham Rat Virus (KRV)-induced type 1 diabetes (T1D) to test the hypothesis that disease mechanisms are linked with beta cell infection and intra-islet immune activation prior to insulitis. KRV induces genes involved in type I and type II interferon pathways in islet cell lines in vitro and in islets from day-5-infected animals in vivo via mechanisms that do not involve insulitis, beta cell apoptosis, or impaired insulin expression. Immunohistochemistry studies indicated that KRV protein is expressed in beta cells 5 days following infection. KRV induces the phosphorylation of Janus Kinase 1/2 (JAK1/2) and signal transducer and activator of transcription 1 (STAT-1) in islet cells via a mechanism that could involve TLR9 and NF-κB pathways. These data demonstrate for the first time that KRV-induced islet destruction is associated with beta cell infection and intra-islet innate immune upregulation early in the disease process.


Journal of Molecular Medicine | 2014

Histone deacetylase inhibitor suppresses virus-induced proinflammatory responses and type 1 diabetes.

Naoko Hara; Aimon K. Alkanani; Charles A. Dinarello; Danny Zipris


American Journal of Respiratory and Critical Care Medicine | 2014

Functional Prostacyclin Synthase Promoter Polymorphisms. Impact in Pulmonary Arterial Hypertension

Robert Stearman; Amber R. Cornelius; Xiao Lu; David S. Conklin; Mark J. Del Rosario; Anita M. Lowe; Mihret T. Elos; Lynsey M. Fettig; Randall Wong; Naoko Hara; Joy D. Cogan; John A. Phillips; Matthew R.G. Taylor; Brian B. Graham; Rubin M. Tuder; James E. Loyd; Mark W. Geraci


Archive | 2015

Alterations in Intestinal Microbiota Correlate with Susceptibility to

Aimon K. Alkanani; Naoko Hara; Peter A. Gottlieb; Diana Ir; Charles E. Robertson; Daniel N. Frank; Danny Zipris; Barbara Davis

Collaboration


Dive into the Naoko Hara's collaboration.

Top Co-Authors

Avatar

Aimon K. Alkanani

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Danny Zipris

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Charles E. Robertson

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Daniel N. Frank

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Diana Ir

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Brandie D. Wagner

Colorado School of Public Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles A. Dinarello

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Peter A. Gottlieb

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Amber R. Cornelius

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge