• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 3
  • 1
  • 1
  • Tagged with
  • 7
  • 7
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

THE CONTRIBUTIONS OF ACTIVIN B SIGNALING TO DIABETIC KIDNEY DISEASE / ACTIVIN B IN DIABETIC KIDNEY DISEASE

Khajehei, Mohammad January 2022 (has links)
DKD is the leading cause of kidney failure in Canada and its patients suffer the highest morbidity and mortality rates of any kidney failure patient group. Current interventions including strict glycemic control only delay DKD. Thus, there is a major need to identify new therapeutic targets. High glucose (HG) is identified as a major pathogenic factor, inducing the release of growth factors leading to kidney fibrosis. Although treatments have been developed to target these factors, their effectiveness is accompanied by adverse effects due to the lack of specificity. Recently, activins have been suggested to have a prominent role in promoting renal fibrosis and developing a specific anti-activin therapy can avoid potential side effects. Although there is evidence supporting an important role for activin A (ActA) in the induction of fibrosis in DKD, whether ActB also contributes is unknown. In this study, we aim to determine the potential contribution of ActB to promoting fibrosis. Our results show that ActA and ActB are upregulated in rodent and human DKD. We show that hyperglycemia leads to the secretion of ActA and ActB by mesangial cells (MC), whereas only ActB is secreted by renal fibroblasts (RF). Similar to HG, treatment with ActA or ActB leads to Smad2/3 activation and upregulation of extracellular matrix proteins, whereas specific inhibition of either ActA or ActB attenuates these effects. We show that ActA and ActB regulate HG-induced activation of MRTF-A/SRF in MC, leading to an activated phenotype characterized by increased α-SMA expression and ECM production. Lastly, we confirm the specificity and functionality of the activin propeptides in vitro, providing evidence for their effectiveness in vivo. This study will help further our knowledge of the role activins in DKD, potentially providing an alternative therapy. / Thesis / Master of Science (MSc) / As the leading cause of end stage renal disease, diabetic kidney disease (DKD) is described as the reduction in renal function due to chronic exposure to diabetes. This thesis is aimed to understand the pathways and mechanisms that contribute to the development and progression of DKD to help identify novel therapeutic options. This project identified activin B (ActB) as a contributor to the disease and gives evidence that blocking the actions of ActB can prevent profibrotic effects in cells, similar to the profibrotic effects seen in DKD. Furthermore, this thesis demonstrates preliminary evidence for the beneficial effects of anti-ActB therapy, providing a potential alternative therapeutic option for DKD patients.
2

The Role of Activin B in Skeletal Muscle Injury and Regeneration

Yaden, Melissa A. 12 1900 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Acute skeletal muscle injury leads to increases in activin B levels and when selectively neutralized with a monoclonal antibody, there is augmented skeletal muscle repair.
3

Activin B Promotes Hepatic Fibrogenesis

Wang, Yan 08 1900 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Liver fibrosis is a common consequence of various chronic liver diseases. Although transforming growth factor β 1 (TGFβ1) expression is known to be associated with liver fibrosis, the reduced clinical efficacy of TGFβ1 inhibition or the inefficiency to completely prevent liver fibrosis in mice with liver-specific knockout of TGF receptor II suggests that other factors can mediate liver fibrogenesis. As a TGFβ superfamily ligand, activin A signaling modulates liver injury by prohibiting hepatocyte proliferation, mediating hepatocyte apoptosis, promoting Kupffer cell activation, and inducing hepatic stellate cell (HSC) activation in vitro. However, the mechanism of action and in vivo functional significance of activin A in liver fibrosis models remain uncertain. Moreover, whether activin B, another ligand structurally related to activin A, is involved in liver fibrogenesis is not yet known. This study aimed to investigate the role of activin A and B in liver fibrosis initiation and progression. The levels of hepatic and circulating activin B and A were analyzed in patients with various chronic liver diseases, including end-stage liver diseases (ESLD), non-alcoholic steatohepatitis (NASH), and alcoholic liver disease (ALD). In addition, their levels were measured in mouse carbon tetrachloride (CCl4), bile duct ligation (BDL), and ALD liver injury models. Mouse primary hepatocytes, RAW264.7 cells, and LX-2 cells were used as in vitro models of hepatocytes, macrophages, and HSCs, respectively. The specificity and potency of anti-activin B monoclonal antibody (mAb) and anti-activin A mAb were evaluated using Smad2/3 luciferase assay. Activin A, activin B, or their combination were immunologically inactivated by the neutralizing mAbs in mice with progressive or established liver fibrosis induced by CCl4 or with developing cholestatic liver fibrosis induced by BDL surgery. In patients with ESLD, NASH, and ALD, increases in hepatic and circulating activin B, but not activin A, were associated with liver fibrosis, irrespective of etiology. In mice with CCl4-, BDL-, or alcohol-induced liver injury, activin B was persistently elevated in the liver and circulation, whereas activin A showed only transient increases. Activin B was expressed and secreted mainly by the hepatocytes and other cells, including cholangiocytes, activated HSCs, and immune cells. Exogenous administration of activin B promoted hepatocyte injury, activated macrophages to release cytokines, and induced a pro-fibrotic expression profile and septa formation in HSCs. Co-treatment of activin A and B interdependently activated the chemokine (C-X-C motif) ligand 1 (CXCL1)/inducible nitric oxide synthase (iNOS) pathway in macrophages and additively upregulated connective tissue growth factor expression in HSCs. Activin B and A had redundant, unique, and interactive effects on the transcripts related to HSC activation. The neutralization of activin B attenuated the development of liver fibrosis and improved liver function in mice with CCl4- or BDL-induced liver fibrosis and largely reversed the already established liver fibrosis in the CCl4 mouse model. These effects were improved by the administration of additional anti-activin A antibody. Combination of both antibodies also inhibited hepatic and circulating inflammatory cytokine production in the BDL mouse model. In conclusion, activin B is a potential circulating biomarker and potent promotor of liver fibrosis. Its levels in the liver and circulation increase significantly in both acute and chronic states of liver injury. Activin B might additively or interdependently cooperate with activin A, which directly acts on multiple liver cell populations during liver injury and fibrosis, as the combination of both proteins increases pro-inflammatory and pro-fibrotic responses in vitro. In addition, the neutralization of both activin A and activin B in vivo enhances the preventive and reversible effects of liver injury and fibrosis compared to that when activin B alone is neutralized. Our data reveal a novel target of liver fibrosis and the mechanism of activin B-mediated initiation of this process by damaging hepatocytes and activating macrophages and HSCs. Our findings show that activin B promotes hepatic fibrogenesis, and that targeting of activin B has anti-inflammatory and anti-fibrotic effects, which ameliorate liver injury by preventing or regressing liver fibrosis. Antagonizing either activin B alone or in combination with activin A prevents and regresses liver fibrosis in multiple animal studies, paving way for future clinical studies.
4

Transcriptional regulation of hepcidin by molecules mediating inflammatory responses / 炎症反応仲介分子によるヘプシジン転写の調節

Kanamori, Yohei 26 March 2018 (has links)
京都大学 / 0048 / 新制・課程博士 / 博士(農学) / 甲第21135号 / 農博第2261号 / 新制||農||1057(附属図書館) / 学位論文||H30||N5109(農学部図書室) / 京都大学大学院農学研究科応用生物科学専攻 / (主査)教授 松井 徹, 教授 久米 新一, 教授 廣岡 博之 / 学位規則第4条第1項該当 / Doctor of Agricultural Science / Kyoto University / DGAM
5

ACTIVIN B PROMOTES HEPATIC FIBROGENESIS

Yan Wang (7022162) 16 October 2019 (has links)
<p>Activin B, a TGFβ ligand, is associated with liver inflammatory response. We aimed to investigate whether it modulates liver fibrogenesis. <b> </b>Liver and serum activin B, along with its analog activin A, were analyzed in patients with liver fibrosis from different etiologies and in mouse acute liver injury and liver fibrosis models. Activin B, activin A, or both was immunologically neutralized in progressive or established carbon tetrachloride-induced mouse liver fibrosis. The direct effects of activin B and A on hepatocytes, macrophages, and hepatic stellate cells (HSCs) were evaluated <i>in vitro</i>. In human patients, increased activin B is associated with liver fibrosis irrespective of the etiologies. In mice, activin B exhibited persistent elevation in liver and circulation following the onset of liver injury, whereas activin A displayed transient increases. Neutralizing activin B largely prevented and remarkably regressed liver fibrosis, which was augmented by co-neutralizing activin A in mice. Mechanistically, activin B promoted hepatocyte injury, activated macrophages to release cytokines, and induced a pro-fibrotic expression profile and septa formation in HSCs, which were magnified by activin A. Furthermore, activin B and A interdependently activated the CXCL1/iNOS pathway in macrophages and additively upregulated CTGF transcript in HSCs <i>in vitro</i>. Consistently, the expression of these genes was prohibited by neutralizing either one of these two ligands in injured livers. Activin B potently drives the initiation and progression of liver fibrogenesis. It additively or interdependently cooperates with activin A, directly acts on multiple liver cell populations, and induces liver fibrogenesis.<b> </b>Antagonizing activin B or both activins B and A prevents and regresses liver fibrosis in mouse CCl<sub>4</sub> model, inspiring the development of a novel therapy of chronic liver diseases.</p>
6

Fer et immunité innée : vers une meilleure compréhension des mécanismes développés par l'hôte pour réduire le fer accessible aux pathogènes / Iron and innate immunity : toward a better understanding of the mechanisms developped by the host to reduce the iron availability for pathogens

Gineste, Aurélie 26 September 2016 (has links)
Le fer est un élément essentiel à de nombreux processus physiologiques fondamentaux. Lors d'une infection, une forte compétition entre l'hôte et l'agent pathogène a lieu ; alors que la bactérie a besoin d'acquérir le fer de l'hôte, pour son développement et sa virulence, l'hôte déploie de nombreux mécanismes pour protéger ses stocks de fer. Il sécrète notamment un peptide capable de moduler l'homéostasie du fer au niveau systémique, l'hepcidine, qui va causer une diminution de la quantité de fer sérique, une rétention intracellulaire du fer et donc une restriction du fer accessible aux pathogènes. Lors d'une inflammation, l'expression de l'hepcidine est décrite dans la littérature pour être principalement induite via l'activation de deux voies de signalisation : la voie STAT3 et la voie BMP/SMAD, l'altération de chacune de ces voies conduisant à un défaut d'induction d'hepcidine. Cependant, nos précédentes observations publiées ont infirmé le rôle de l'IL6, ligand de la voie STAT3, dans la régulation de l'hepcidine en réponse au LPS, et ont suggéré l'implication d'un peptide appartenant à la famille du TGFb, l'activine B, dans la régulation de l'hepcidine via l'activation de la voie BMP/SMAD in vivo. Dans cette étude, nous nous sommes intéressés au rôle de l'activine B dans la régulation de l'hepcidine in vivo, lors d'une infection. Grâce à l'utilisation de souris invalidées pour le gène codant l'activine B (Inhbb-/-), nous avons confirmé que l'activine B était un ligand endogène de la voie BMP/SMAD dans le foie, puisqu'elle induit la phosphorylation des effecteurs SMAD en réponse au LPS. Cependant, nous avons pu clairement démontrer que l'activine B, et donc l'augmentation de la phosphorylation des effecteurs SMAD, ne participaient pas à la régulation de l'hepcidine in vivo, en réponse à l'infection. Nous nous sommes alors intéressés à l'implication de l'IL6 dans la régulation de l'hepcidine. Alors que l'absence d'Il6 n'altère pas l'induction de l'hepcidine in vivo en réponse au LPS, cette cytokine semble jouer un rôle clé pour la réponse de l'hôte lors d'une infection par une bactérie. Dans ce contexte, la littérature décrit l'importance de l'IL6 pour une réponse immunitaire protectrice de l'hôte lors d'une infection. Lors d'une infection, nous proposons donc l'implication d'une nouvelle voie de signalisation dans l'expression de l'hepcidine. De plus, nous suggérons un rôle important de l'IL6, non pas dans la régulation transcriptionnelle de l'hepcidine, mais pour la protection de l'hôte lors d'une infection bactérienne. Enfin, nos résultats montrent qu'un niveau d'activation basal de la voie BMP/SMAD est requis pour une induction d'hepcidine lors d'inflammation, et que l'augmentation de la phosphorylation des effecteurs SMAD observée en réponse à l'inflammation ne participe pas à cette régulation. / Iron is essential for several fundamental metabolic processes. During infection, a strong competition between the host and the pathogen occurs; while the bacteria needs to acquire iron from the host, for its growth and virulence, hosts have developed several mechanisms to protect its iron stores. In particular, the host produces a peptide in order to modulate systemic iron homeostasis, hepcidin. Hepcidin decreases the amount of circulating iron, causes intracellular iron retention and thus a restriction of accessible iron to pathogens. During inflammation, hepcidin expression is described in the literature to be mainly mediated through activation of two signaling pathways: the STAT3 and the BMP/SMAD pathways. Impairment of one of these pathways leads to an impaired hepcidin induction. However, our previous published observations did not support the role of IL6, the major ligand of STAT3 pathway, in the regulation of hepcidin in response to LPS, but suggested the involvement of another protein, that belongs to the TGFb family, activin B, in the regulation of hepcidin via the activation of the BMP/SMAD pathway in vivo. In this study, we investigated the role of activin B in the regulation of hepcidin in vivo, during infection. By using knockout mice for the gene encoding activin B (Inhbb-/-), our results suggest that activin B is not involved in the regulation of hepcidin in vivo in response to infection. We then investigated the function of IL6 in the regulation of hepcidin. Although the absence of IL6 does not affect the induction of hepcidin in vivo in response to LPS, this cytokine appears to play a key role in the host response during bacterial infection. Indeed, the literature describes the importance of IL6 for a protective immune response of the host during infection. During infection, we hypothesize that another signaling pathway regulates hepcidin expression. In addition, we suggest an important role of IL6, not in the transcriptional regulation of hepcidin, but for the host protection during a bacterial infection. Finally, our results also show that basal level of BMP/SMAD pathway is required for an appropriate induction of hepcidin during inflammation.
7

The Role of Activin B in Skeletal Muscle Injury and Regeneration

Melissa A Yaden (11798105) 20 December 2021 (has links)
Activin B, a member of the transforming growth factor-β superfamily, is ubiquitously expressed in diverse tissues and is a regulator of reproduction, embryonic development, and adult tissue homeostasis. We aimed to determine whether activin B is involved in skeletal muscle injury and if selective inhibition of activin B would provide a regenerative benefit. The local introduction of activin B into normal skeletal muscle increased the expression of inflammatory and muscle atrophy genes TWEAK, TNFα, GDF3 and TRIM63, by 2-, 10-, 10-, and 4-fold, respectively. The data indicate a sensitive response of skeletal muscle to activin B. Six hours after cardiotoxin-induced skeletal muscle damage, circulating activin B protein expression in serum increased by 9-fold and InhβB gene expression increased by 30-fold in muscle. After cardiotoxin-induced skeletal muscle damage, activin B protein expression in muscle was significantly increased at 48- and 120-hours by 1.5 and 2-fold, respectively. Muscle histopathological features showed that activin B antibody–treated mice displayed a reduction in necrotic debris, with a concomitant reduction in intramyocellular space at 9-days after injury. Activin B treated C2C12 myoblasts also displayed a dose-dependent reduction in active myogenesis. Furthermore, the increased presence of activin B early in muscle injury impedes muscle repair and remodeling. In summary, acute muscle injury leads to increases in activin B levels and when selectively neutralized with a monoclonal antibody, there is augmented skeletal muscle repair.

Page generated in 0.0425 seconds