371 results on '"Fn14"'
Search Results
2. Exercise, disease state and sex influence the beneficial effects of Fn14-depletion on survival and muscle pathology in the SOD1 G93A amyotrophic lateral sclerosis (ALS) mouse model
- Author
-
Gareth Hazell, Eve McCallion, Nina Ahlskog, Emma R. Sutton, Magnus Okoh, Emad I. H. Shaqoura, Joseph M. Hoolachan, Taylor Scaife, Sara Iqbal, Amarjit Bhomra, Anna J. Kordala, Frederique Scamps, Cedric Raoul, Matthew J. A. Wood, and Melissa Bowerman
- Subjects
Amyotrophic lateral sclerosis ,Skeletal muscle ,TWEAK ,Fn14 ,Exercise ,Sex ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background Amyotrophic lateral sclerosis (ALS) is a devastating and incurable neurodegenerative disease. Accumulating evidence strongly suggests that intrinsic muscle defects exist and contribute to disease progression, including imbalances in whole-body metabolic homeostasis. We have previously reported that tumour necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor inducible 14 (Fn14) are significantly upregulated in skeletal muscle of the SOD1 G93A ALS mouse model. While antagonising TWEAK did not impact survival, we did observe positive effects in skeletal muscle. Given that Fn14 has been proposed as the main effector of the TWEAK/Fn14 activity and that Fn14 can act independently from TWEAK in muscle, we suggest that manipulating Fn14 instead of TWEAK in the SOD1 G93A ALS mice could lead to differential and potentially improved benefits. Methods We thus investigated the contribution of Fn14 to disease phenotypes in the SOD1 G93A ALS mice. To do so, Fn14 knockout mice (Fn14 −/− ) were crossed onto the SOD1 G93A background to generate SOD1 G93A ;Fn14 −/− mice. Investigations were performed on both unexercised and exercised (rotarod and/or grid test) animals (wild type (WT), Fn14 −/−, SOD1 G93A and SOD1 G93A ;Fn14 −/− ). Results Here, we firstly confirm that the TWEAK/Fn14 pathway is dysregulated in skeletal muscle of SOD1 G93A mice. We then show that Fn14-depleted SOD1 G93A mice display increased lifespan, myofiber size, neuromuscular junction endplate area as well as altered expression of known molecular effectors of the TWEAK/Fn14 pathway, without an impact on motor function. Importantly, we also observe a complex interaction between exercise (rotarod and grid test), genotype, disease state and sex that influences the overall effects of Fn14 deletion on survival, expression of known molecular effectors of the TWEAK/Fn14 pathway, expression of myosin heavy chain isoforms and myofiber size. Conclusions Our study provides further insights on the different roles of the TWEAK/Fn14 pathway in pathological skeletal muscle and how they can be influenced by age, disease, sex and exercise. This is particularly relevant in the ALS field, where combinatorial therapies that include exercise regimens are currently being explored. As such, a better understanding and consideration of the interactions between treatments, muscle metabolism, sex and exercise will be of importance in future studies.
- Published
- 2024
- Full Text
- View/download PDF
3. Exercise, disease state and sex influence the beneficial effects of Fn14-depletion on survival and muscle pathology in the SOD1G93A amyotrophic lateral sclerosis (ALS) mouse model.
- Author
-
Hazell, Gareth, McCallion, Eve, Ahlskog, Nina, Sutton, Emma R., Okoh, Magnus, Shaqoura, Emad I. H., Hoolachan, Joseph M., Scaife, Taylor, Iqbal, Sara, Bhomra, Amarjit, Kordala, Anna J., Scamps, Frederique, Raoul, Cedric, Wood, Matthew J. A., and Bowerman, Melissa
- Subjects
SEX factors in disease ,AMYOTROPHIC lateral sclerosis ,FIBROBLAST growth factors ,SKELETAL muscle ,MUSCLE metabolism - Abstract
Background: Amyotrophic lateral sclerosis (ALS) is a devastating and incurable neurodegenerative disease. Accumulating evidence strongly suggests that intrinsic muscle defects exist and contribute to disease progression, including imbalances in whole-body metabolic homeostasis. We have previously reported that tumour necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor inducible 14 (Fn14) are significantly upregulated in skeletal muscle of the SOD1
G93A ALS mouse model. While antagonising TWEAK did not impact survival, we did observe positive effects in skeletal muscle. Given that Fn14 has been proposed as the main effector of the TWEAK/Fn14 activity and that Fn14 can act independently from TWEAK in muscle, we suggest that manipulating Fn14 instead of TWEAK in the SOD1G93A ALS mice could lead to differential and potentially improved benefits. Methods: We thus investigated the contribution of Fn14 to disease phenotypes in the SOD1G93A ALS mice. To do so, Fn14 knockout mice (Fn14−/− ) were crossed onto the SOD1G93A background to generate SOD1G93A ;Fn14−/− mice. Investigations were performed on both unexercised and exercised (rotarod and/or grid test) animals (wild type (WT), Fn14−/− , SOD1G93A and SOD1G93A ;Fn14−/− ). Results: Here, we firstly confirm that the TWEAK/Fn14 pathway is dysregulated in skeletal muscle of SOD1G93A mice. We then show that Fn14-depleted SOD1G93A mice display increased lifespan, myofiber size, neuromuscular junction endplate area as well as altered expression of known molecular effectors of the TWEAK/Fn14 pathway, without an impact on motor function. Importantly, we also observe a complex interaction between exercise (rotarod and grid test), genotype, disease state and sex that influences the overall effects of Fn14 deletion on survival, expression of known molecular effectors of the TWEAK/Fn14 pathway, expression of myosin heavy chain isoforms and myofiber size. Conclusions: Our study provides further insights on the different roles of the TWEAK/Fn14 pathway in pathological skeletal muscle and how they can be influenced by age, disease, sex and exercise. This is particularly relevant in the ALS field, where combinatorial therapies that include exercise regimens are currently being explored. As such, a better understanding and consideration of the interactions between treatments, muscle metabolism, sex and exercise will be of importance in future studies. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
4. Alpha-Ketoglutarate Regulates Tnfrsf12a/Fn14 Expression via Histone Modification and Prevents Cancer-Induced Cachexia.
- Author
-
Ruiz, Bryan, Lowman, Xazmin, Yang, Ying, Fan, Qi, Wang, Tianhong, Wu, Hongmei, Hanse, Eric, and Kong, Mei
- Subjects
FN14 ,TNFRSF12A ,alpha-ketoglutarate ,cachexia ,cancer ,colon cancer ,histone ,Animals ,Mice ,Cachexia ,Ketoglutaric Acids ,Histones ,Glutamine ,Histone Code ,Colonic Neoplasms ,Disease Models ,Animal ,Histone Methyltransferases ,Tumor Microenvironment - Abstract
Previous studies have shown that inhibition of TNF family member FN14 (gene: TNFRSF12A) in colon tumors decreases inflammatory cytokine expression and mitigates cancer-induced cachexia. However, the molecular mechanisms underlying the regulation of FN14 expression remain unclear. Tumor microenvironments are often devoid of nutrients and oxygen, yet how the cachexic response relates to the tumor microenvironment and, importantly, nutrient stress is unknown. Here, we looked at the connections between metabolic stress and FN14 expression. We found that TNFRSF12A expression was transcriptionally induced during glutamine deprivation in cancer cell lines. We also show that the downstream glutaminolysis metabolite, alpha-ketoglutarate (aKG), is sufficient to rescue glutamine-deprivation-promoted TNFRSF12A induction. As aKG is a co-factor for histone de-methylase, we looked at histone methylation and found that histone H3K4me3 at the Tnfrsf12a promoter is increased under glutamine-deprived conditions and rescued via DM-aKG supplementation. Finally, expression of Tnfrsf12a and cachexia-induced weight loss can be inhibited in vivo by DM-aKG in a mouse cancer cachexia model. These findings highlight a connection between metabolic stress and cancer cachexia development.
- Published
- 2023
5. Erratum: Antibody-based soluble and membrane-bound TWEAK mimicking agonists with FcγR-independent activity
- Author
-
Frontiers Production Office
- Subjects
agonistic antibodies ,cell death ,FcγR ,Fn14 ,NFκB ,TNF receptor superfamily ,Immunologic diseases. Allergy ,RC581-607 - Published
- 2023
- Full Text
- View/download PDF
6. TWEAK and Fn14 are overexpressed in immune-mediated necrotizing myopathy: implications for muscle damage and repair.
- Author
-
Yang, Mengge, Ge, Huizhen, Ji, Suqiong, Li, Yue, Xu, Li, Bi, Zhuajin, and Bu, Bitao
- Subjects
- *
MUSCLE diseases , *SKELETAL muscle , *CONVALESCENCE , *APOPTOSIS , *GUIDED tissue regeneration , *RESEARCH funding , *DESCRIPTIVE statistics , *MYOSITIS , *STATISTICAL models , *GAS gangrene - Abstract
Objectives TNF-like weak inducer of apoptosis (TWEAK) and its sole receptor fibroblast growth factor-inducible 14 (Fn14) are involved in various inflammatory conditions. This study was performed to investigate the potential role of TWEAK/Fn14 in immune-mediated necrotizing myopathy (IMNM). Methods Muscle biopsies from patients with IMNM (n = 37) and controls (n = 11) were collected. Human muscle cells were treated with TWEAK in vitro. Muscle biopsies and cultured muscle cells were analysed by immunostaining and quantitative PCR. Serum levels of TWEAK and Fn14 were detected by ELISA. Results TWEAK and Fn14 were overexpressed in IMNM muscle biopsies. The percentage of Fn14-positive myofibers correlated with disease severity, myonecrosis, regeneration and inflammation infiltrates. Fn14-positive myofibers tended to be surrounded or invaded by CD68 + macrophages. TWEAK treatment had a harmful effect on cultured muscle cells by inducing the production of multiple chemokines and pro-inflammatory cytokines. Serum Fn14 levels were increased in patients with IMNM and correlated with muscle weakness. Conclusions TWEAK/Fn14 signalling was activated in IMNM, most likely aggravating muscle damage via amplifying inflammatory response and macrophages chemotaxis. Fn14 seems to be a biomarker for assessing disease severity in IMNM. In addition, Fn14 may also contribute to muscle injury repair. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Protein therapy of skeletal muscle atrophy and mechanism by angiogenic factor AGGF1
- Author
-
Zuhan He, Qixue Song, Yubing Yu, Feng Liu, Jinyan Zhao, Waikeong Un, Xingwen Da, Chengqi Xu, Yufeng Yao, and Qing K. Wang
- Subjects
AGGF1 ,TWEAK ,Fn14 ,NF‐κB ,skeletal muscle atrophy ,cancer cachexia ,Diseases of the musculoskeletal system ,RC925-935 ,Human anatomy ,QM1-695 - Abstract
Abstract Background Skeletal muscle atrophy is a common condition without a pharmacologic therapy. AGGF1 encodes an angiogenic factor that regulates cell differentiation, proliferation, migration, apoptosis, autophagy and endoplasmic reticulum stress, promotes vasculogenesis and angiogenesis and successfully treats cardiovascular diseases. Here, we report the important role of AGGF1 in the pathogenesis of skeletal muscle atrophy and attenuation of muscle atrophy by AGGF1. Methods In vivo studies were carried out in impaired leg muscles from patients with lumbar disc herniation, two mouse models for skeletal muscle atrophy (denervation and cancer cachexia) and heterozygous Aggf1+/− mice. Mouse muscle atrophy phenotypes were characterized by body weight and myotube cross‐sectional areas (CSA) using H&E staining and immunostaining for dystrophin. Molecular mechanistic studies include co‐immunoprecipitation (Co‐IP), western blotting, quantitative real‐time PCR analysis and immunostaining analysis. Results Heterozygous Aggf1+/− mice showed exacerbated phenotypes of reduced muscle mass, myotube CSA, MyHC (myosin heavy chain) and α‐actin, increased inflammation (macrophage infiltration), apoptosis and fibrosis after denervation and cachexia. Intramuscular and intraperitoneal injection of recombinant AGGF1 protein attenuates atrophy phenotypes in mice with denervation (gastrocnemius weight 81.3 ± 5.7 mg vs. 67.3 ± 5.1 mg for AGGF1 vs. buffer; P
- Published
- 2023
- Full Text
- View/download PDF
8. Fn14 and TNFR2 as regulators of cytotoxic TNFR1 signaling
- Author
-
Daniela Siegmund, Olena Zaitseva, and Harald Wajant
- Subjects
apoptosis ,Fn14 ,necroptosis ,TNF ,TNFR1 ,TNFR2 ,Biology (General) ,QH301-705.5 - Abstract
Tumor necrosis factor (TNF) receptor 1 (TNFR1), TNFR2 and fibroblast growth factor-inducible 14 (Fn14) belong to the TNF receptor superfamily (TNFRSF). From a structural point of view, TNFR1 is a prototypic death domain (DD)-containing receptor. In contrast to other prominent death receptors, such as CD95/Fas and the two TRAIL death receptors DR4 and DR5, however, liganded TNFR1 does not instruct the formation of a plasma membrane-associated death inducing signaling complex converting procaspase-8 into highly active mature heterotetrameric caspase-8 molecules. Instead, liganded TNFR1 recruits the DD-containing cytoplasmic signaling proteins TRADD and RIPK1 and empowers these proteins to trigger cell death signaling by cytosolic complexes after their release from the TNFR1 signaling complex. The activity and quality (apoptosis versus necroptosis) of TNF-induced cell death signaling is controlled by caspase-8, the caspase-8 regulatory FLIP proteins, TRAF2, RIPK1 and the RIPK1-ubiquitinating E3 ligases cIAP1 and cIAP2. TNFR2 and Fn14 efficiently recruit TRAF2 along with the TRAF2 binding partners cIAP1 and cIAP2 and can thereby limit the availability of these molecules for other TRAF2/cIAP1/2-utilizing proteins including TNFR1. Accordingly, at the cellular level engagement of TNFR2 or Fn14 inhibits TNFR1-induced RIPK1-mediated effects reaching from activation of the classical NFκB pathway to induction of apoptosis and necroptosis. In this review, we summarize the effects of TNFR2- and Fn14-mediated depletion of TRAF2 and the cIAP1/2 on TNFR1 signaling at the molecular level and discuss the consequences this has in vivo.
- Published
- 2023
- Full Text
- View/download PDF
9. Antibody-based soluble and membrane-bound TWEAK mimicking agonists with FcγR-independent activity.
- Author
-
Zaitseva, Olena, Hoffmann, Annett, Löst, Margaretha, Anany, Mohamed A., Tengyu Zhang, Kucka, Kirstin, Wiegering, Armin, Otto, Christoph, and Wajant, Harald
- Subjects
ANTIBODY-dependent cell cytotoxicity ,FIBROBLAST growth factors ,CHIMERIC proteins ,TUMOR necrosis factors ,IMMUNE response - Abstract
Fibroblast growth factor (FGF)-inducible 14 (Fn14) activates the classical and alternative NFkB (nuclear factor 'kappa-light-chain-enhancer' of activated Bcells) signaling pathway but also enhances tumor necrosis factor (TNF)-induced cell death. Fn14 expression is upregulated in non-hematopoietic cells during tissue injury and is also often highly expressed in solid cancers. In view of the latter, there were and are considerable preclinical efforts to target Fn14 for tumor therapy, either by exploiting Fn14 as a target for antibodies with cytotoxic activity (e.g. antibody-dependent cellular cytotoxicity (ADCC)-inducing IgG variants, antibody drug conjugates) or by blocking antibodies with the aim to interfere with protumoral Fn14 activities. Noteworthy, there are yet no attempts to target Fn14 with agonistic Fc effector function silenced antibodies to unleash the proinflammatory and cell death-enhancing activities of this receptor for tumor therapy. This is certainly not at least due to the fact that anti-Fn14 antibodies only act as effective agonists when they are presented bound to Fcg receptors (FcgR). Thus, there are so far no antibodies that robustly and selectively engage Fn14 signaling without triggering unwanted FcgR-mediated activities. In this study, we investigated a panel of variants of the anti-Fn14 antibody 18D1 of different valencies and domain architectures with respect to their inherent FcgRindependent ability to trigger Fn14-associated signaling pathways. In contrast to conventional 18D1, the majority of 18D1 antibody variants with four or more Fn14 binding sites displayed a strong ability to trigger the alternative NFkB pathway and to enhance TNF-induced cell death and therefore resemble in their activity soluble (TNF)-like weak inducer of apoptosis (TWEAK), one form of the natural occurring ligand of Fn14. Noteworthy, activation of the classical NFkB pathway, which naturally is predominately triggered by membrane-bound TWEAK but not soluble TWEAK, was preferentially observed with a subset of constructs containing Fn14 binding sites at opposing sites of the IgG scaffold, e.g. IgG1-scFv fusion proteins. A superior ability of IgG1-scFv fusion proteins to trigger classical NFkB signaling was also observed with the anti-Fn14 antibody PDL192 suggesting that we identified generic structures for Fn14 antibody variants mimicking soluble and membrane-bound TWEAK. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Antibody-based soluble and membrane-bound TWEAK mimicking agonists with FcγR-independent activity
- Author
-
Olena Zaitseva, Annett Hoffmann, Margaretha Löst, Mohamed A. Anany, Tengyu Zhang, Kirstin Kucka, Armin Wiegering, Christoph Otto, and Harald Wajant
- Subjects
agonistic antibodies ,cell death ,FcγR ,Fn14 ,NFκB ,TNF receptor superfamily ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Fibroblast growth factor (FGF)-inducible 14 (Fn14) activates the classical and alternative NFκB (nuclear factor ‘kappa-light-chain-enhancer’ of activated B-cells) signaling pathway but also enhances tumor necrosis factor (TNF)-induced cell death. Fn14 expression is upregulated in non-hematopoietic cells during tissue injury and is also often highly expressed in solid cancers. In view of the latter, there were and are considerable preclinical efforts to target Fn14 for tumor therapy, either by exploiting Fn14 as a target for antibodies with cytotoxic activity (e.g. antibody-dependent cellular cytotoxicity (ADCC)-inducing IgG variants, antibody drug conjugates) or by blocking antibodies with the aim to interfere with protumoral Fn14 activities. Noteworthy, there are yet no attempts to target Fn14 with agonistic Fc effector function silenced antibodies to unleash the proinflammatory and cell death-enhancing activities of this receptor for tumor therapy. This is certainly not at least due to the fact that anti-Fn14 antibodies only act as effective agonists when they are presented bound to Fcγ receptors (FcγR). Thus, there are so far no antibodies that robustly and selectively engage Fn14 signaling without triggering unwanted FcγR-mediated activities. In this study, we investigated a panel of variants of the anti-Fn14 antibody 18D1 of different valencies and domain architectures with respect to their inherent FcγR-independent ability to trigger Fn14-associated signaling pathways. In contrast to conventional 18D1, the majority of 18D1 antibody variants with four or more Fn14 binding sites displayed a strong ability to trigger the alternative NFκB pathway and to enhance TNF-induced cell death and therefore resemble in their activity soluble (TNF)-like weak inducer of apoptosis (TWEAK), one form of the natural occurring ligand of Fn14. Noteworthy, activation of the classical NFκB pathway, which naturally is predominately triggered by membrane-bound TWEAK but not soluble TWEAK, was preferentially observed with a subset of constructs containing Fn14 binding sites at opposing sites of the IgG scaffold, e.g. IgG1-scFv fusion proteins. A superior ability of IgG1-scFv fusion proteins to trigger classical NFκB signaling was also observed with the anti-Fn14 antibody PDL192 suggesting that we identified generic structures for Fn14 antibody variants mimicking soluble and membrane-bound TWEAK.
- Published
- 2023
- Full Text
- View/download PDF
11. TWEAK/Fn14 signaling may function as a reactive compensatory mechanism against extracellular matrix accumulation in keloid fibroblasts
- Author
-
Yitian Zhang, Xiaoli Li, Wei Liu, Guanglei Hu, Hanjiang Gu, Xiao Cui, Dewu Zhang, Weihui Zeng, and Yumin Xia
- Subjects
α-SMA ,Extracellular matrix ,Fibroblast ,Fn14 ,Keloid ,TGF-β1 ,Cytology ,QH573-671 - Abstract
Overabundance of the extracellular matrix resulting from hyperproliferation of keloid fibroblasts (KFs) and dysregulation of apoptosis represents the main pathophysiology underlying keloids. TWEAK is a weak apoptosis inducer, and it plays a critical role in pathological tissue remodeling via its receptor, Fn14. However, the role of TWEAK/Fn14 signaling in the pathogenesis of keloids has not been investigated. In this study, we confirmed the overexpression levels of TWEAK and Fn14 in clinical keloid tissue specimens and primary KFs. The extracellular matrix (ECM)-related genes were also evaluated between primary KFs and their normal counterparts to determine the factors leading to the formation or development of keloids. Unexpectedly, exogenous TWEAK significantly reduced the levels of collagen I and collagen III, as well as alpha-smooth muscle actin (α-SMA). Additionally, TWEAK promoted MMPs expression and apoptosis activity of KFs. Furthermore, we verified that the inhibitory effect of TWEAK on KFs is through down-regulation of Polo-like kinase 5, which modulates cell differentiation and apoptosis. The TWEAK-Fn14 axis seems to be a secondary, although less effective, compensatory mechanism to increase the catabolic functions of fibroblasts in an attempt to further decrease the accumulation of collagen. Data Availability: All data generated or analyzed during this study are included in this published article (and its Supporting Information files).
- Published
- 2023
- Full Text
- View/download PDF
12. Dysregulation of Tweak and Fn14 in skeletal muscle of spinal muscular atrophy mice
- Author
-
Katharina E. Meijboom, Emma R. Sutton, Eve McCallion, Emily McFall, Daniel Anthony, Benjamin Edwards, Sabrina Kubinski, Ines Tapken, Ines Bünermann, Gareth Hazell, Nina Ahlskog, Peter Claus, Kay E. Davies, Rashmi Kothary, Matthew J. A. Wood, and Melissa Bowerman
- Subjects
Spinal muscular atrophy ,Survival motor neuron ,Smn ,Tweak ,Fn14 ,Glucose metabolism ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background Spinal muscular atrophy (SMA) is a childhood neuromuscular disorder caused by depletion of the survival motor neuron (SMN) protein. SMA is characterized by the selective death of spinal cord motor neurons, leading to progressive muscle wasting. Loss of skeletal muscle in SMA is a combination of denervation-induced muscle atrophy and intrinsic muscle pathologies. Elucidation of the pathways involved is essential to identify the key molecules that contribute to and sustain muscle pathology. The tumor necrosis factor-like weak inducer of apoptosis (TWEAK)/TNF receptor superfamily member fibroblast growth factor-inducible 14 (Fn14) pathway has been shown to play a critical role in the regulation of denervation-induced muscle atrophy as well as muscle proliferation, differentiation, and metabolism in adults. However, it is not clear whether this pathway would be important in highly dynamic and developing muscle. Methods We thus investigated the potential role of the TWEAK/Fn14 pathway in SMA muscle pathology, using the severe Taiwanese Smn −/−; SMN2 and the less severe Smn 2B/− SMA mice, which undergo a progressive neuromuscular decline in the first three post-natal weeks. We also used experimental models of denervation and muscle injury in pre-weaned wild-type (WT) animals and siRNA-mediated knockdown in C2C12 muscle cells to conduct additional mechanistic investigations. Results Here, we report significantly dysregulated expression of Tweak, Fn14, and previously proposed downstream effectors during disease progression in skeletal muscle of the two SMA mouse models. In addition, siRNA-mediated Smn knockdown in C2C12 myoblasts suggests a genetic interaction between Smn and the TWEAK/Fn14 pathway. Further analyses of SMA, Tweak −/− , and Fn14 −/− mice revealed dysregulated myopathy, myogenesis, and glucose metabolism pathways as a common skeletal muscle feature, providing further evidence in support of a relationship between the TWEAK/Fn14 pathway and Smn. Finally, administration of the TWEAK/Fn14 agonist Fc-TWEAK improved disease phenotypes in the two SMA mouse models. Conclusions Our study provides mechanistic insights into potential molecular players that contribute to muscle pathology in SMA and into likely differential responses of the TWEAK/Fn14 pathway in developing muscle.
- Published
- 2022
- Full Text
- View/download PDF
13. Fn14 exacerbates acute lung injury by activating the NLRP3 inflammasome in mice
- Author
-
Xin-Xin Guan, Hui-Hui Yang, Wen-Jing Zhong, Jia-Xi Duan, Chen-Yu Zhang, Hui-Ling Jiang, Yang Xiang, Yong Zhou, and Cha-Xiang Guan
- Subjects
Acute lung injury ,Fn14 ,NLRP3 inflammasome ,TWEAK ,Macrophage ,Therapeutics. Pharmacology ,RM1-950 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Uncontrolled inflammation is an important factor in the occurrence and development of acute lung injury (ALI). Fibroblast growth factor-inducible 14 (Fn14), a plasma membrane-anchored receptor, takes part in the pathological process of a variety of acute and chronic inflammatory diseases. However, the role of Fn14 in ALI has not yet been elucidated. This study aimed to investigate whether the activation of Fn14 exacerbated lipopolysaccharide (LPS)-induced ALI in mice. Methods In vivo, ALI was induced by intratracheal LPS-challenge combined with/without Fn14 receptor blocker aurintricarboxylic acid (ATA) treatment in C57BL/6J mice. Following LPS administration, the survival rate, lung tissue injury, inflammatory cell infiltration, inflammatory factor secretion, oxidative stress, and NLRP3 inflammasome activation were assessed. In vitro, primary murine macrophages were used to evaluate the underlying mechanism by which Fn14 activated the NLRP3 inflammasome. Lentivirus was used to silence Fn14 to observe its effect on the activation of NLRP3 inflammasome in macrophages. Results In this study, we found that Fn14 expression was significantly increased in the lungs of LPS-induced ALI mice. The inhibition of Fn14 with ATA downregulated the protein expression of Fn14 in the lungs and improved the survival rate of mice receiving a lethal dose of LPS. ATA also attenuated lung tissue damage by decreasing the infiltration of macrophages and neutrophils, reducing inflammation, and suppressing oxidative stress. Importantly, we found that ATA strongly inhibited the activation of NLRP3 inflammasome in the lungs of ALI mice. Furthermore, in vitro, TWEAK, a natural ligand of Fn14, amplified the activation of NLRP3 inflammasome in the primary murine macrophage. By contrast, inhibition of Fn14 with shRNA decreased the expression of Fn14, NLRP3, Caspase-1 p10, and Caspase-1 p20, and the production of IL-1β and IL-18. Furthermore, the activation of Fn14 promoted the production of reactive oxygen species and inhibited the activation of Nrf2-HO-1 in activated macrophages. Conclusions Our study first reports that the activation of Fn14 aggravates ALI by amplifying the activation of NLRP3 inflammasome. Therefore, blocking Fn14 may be a potential way to treat ALI.
- Published
- 2022
- Full Text
- View/download PDF
14. تأثیر تمرین تناوبی با شدت بالا همراه با مصرف پودر اسپیرولینا بر تغییرات بیان ژن مسیر التهاب و آتروفی در عضلهی نعلی رتهای مسن چاق مبتلا به دیابت
- Author
-
مرضیه السادات آذرنیوه, رویا عسکری, and امیرحسین حقیقی
- Subjects
تمرین تناوبی با شدت بالا ,اسپیرولینا ,آتروفی ,tweak ,fn14 ,atrogin-1 موش ,Medicine ,Medicine (General) ,R5-920 - Abstract
مقدمه: فعالیتهای ورزشی، روشی پیشنهادی جهت کاهش التهاب و آتروفی و کنترل بیماری دیابت میباشد. اسپیرولینا نیز دارای اثرات ضد التهابی است. هدف از انجام پژوهش حاضر، بررسی تأثیر هشت هفته تمرینات تناوبی با شدت بالا (High-intensity interval training یا HIIT) همراه با مصرف اسپیرولینا، بر تغییرات بیان ژنهای مسیر التهاب و آتروفی در عضلهی نعلی رتهای مسن چاق مبتلابه دیابت بود.روشها: در این مطالعهی تجربی، 40 سر رت نر مسن نژاد ویستار 20 ماهه با میانگین وزنی 325-280 گرم، پس از خریداری، هشت هفته تحت رژیم پرچرب چاق و پس از آن، با تزریق درون صفاقی 40 میلیگرم بر کیلوگرم استرپتوزوسین (Streptozotocin یا STZ) دیابتی شدند. سپس در پنج گروه مساوی (HIIT، HIIT + مکمل، مکمل، شم و شاهد) به مدت هشت هفته و 5 جلسه در هفته، به اجرای HIIT روی تردمیل (با شدت 90 درصد حداکثر اکسیژن مصرفی) در گروههای تمرینی و مصرف اسپیرولینا در روزهای تمرین برای گروههای HIIT + مکمل و مکمل (50 میلیگرم به ازای هر کیلوگرم وزن بدن) پرداختند. بیان ژنها به روش Real-time polymerase chain reaction (Real time-PCR) اندازهگیری شد. دادهها با استفاده از آزمون One-way ANOVA و آزمون تعقیبی Tukey مورد تجزیه و تحلیل قرار گرفت.یافتهها: القای دیابت در رتهای چاق و مسن، موجب افزایش معنیداری در بیان ژنهای Tweak، Fn14 و Atrogin-1 بین گروههای مورد بررسی نسبت به گروه شاهد گردید، اما تمرین و مصرف مکمل این متغیرها را در گروههای تمرینی و مکمل نسبت به سایر گروهها به طور معنیداری کاهش داد. پس از تمرین، تودهی بدنی در گروههای HIIT، مکمل و HIIT + مکمل نسبت به گروههای شم و شاهد به طور معنیداری کاهش یافت و تودهی عضلهی نعلی نسبت به گروه شاهد تنها در گروههای HIIT و HIIT + مکمل، به طور معنیداری افزایش داشت.نتیجهگیری: با توجه به نتایج تحقیق حاضر، ترکیب HIIT + مکمل به عنوان یک روش مؤثر جهت کاهش آتروفی و افزایش سلامت عضلانی در سالمندان مبتلا به دیابت پیشنهاد میشود.
- Published
- 2022
- Full Text
- View/download PDF
15. Targeting fibroblast growth factor (FGF)-inducible 14 (Fn14) for tumor therapy.
- Author
-
Zaitseva, Olena, Hoffmann, Annett, Otto, Christoph, and Wajant, Harald
- Subjects
ANTIBODY-dependent cell cytotoxicity ,TUMOR necrosis factors ,MITOGEN-activated protein kinases ,CELL death ,FIBROBLAST growth factors ,TUMOR microenvironment - Abstract
Fibroblast growth factor-inducible 14 (Fn14) is a member of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF) and is activated by its ligand TNF-like weak inducer of apoptosis (TWEAK). The latter occurs as a homotrimeric molecule in a soluble and a membrane-bound form. Soluble TWEAK (sTWEAK) activates the weakly inflammatory alternative NF-KB pathway and sensitizes for TNF-induced cell death while membrane TWEAK (memTWEAK) triggers additionally robust activation of the classical NF-KB pathway and various MAP kinase cascades. Fn14 expression is limited in adult organisms but becomes strongly induced in non-hematopoietic cells by a variety of growth factors, cytokines and physical stressors (e.g., hypoxia, irradiation). Since all these Fn14-inducing factors are frequently also present in the tumor microenvironment, Fn14 is regularly found to be expressed by non-hematopoietic cells of the tumor microenvironment and most solid tumor cells. In general, there are three possibilities how the tumor-Fn14 linkage could be taken into consideration for tumor therapy. First, by exploitation of the cancer associated expression of Fn14 to direct cytotoxic activities (antibody-dependent cell-mediated cytotoxicity (ADCC), cytotoxic payloads, CART-cells) to the tumor, second by blockade of potential protumoral activities of the TWEAK/Fn14 system, and third, by stimulation of Fn14 which not only triggers proinflammtory activities but also sensitizes cells for apoptotic and necroptotic cell death. Based on a brief description of the biology of the TWEAK/Fn14 system and Fn14 signaling, we discuss the features of the most relevant Fn14-targeting biologicals and review the preclinical data obtained with these reagents. In particular, we address problems and limitations which became evident in the preclinical studies with Fn14-targeting biologicals and debate possibilities how they could be overcome. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
16. Targeting fibroblast growth factor (FGF)-inducible 14 (Fn14) for tumor therapy
- Author
-
Olena Zaitseva, Annett Hoffmann, Christoph Otto, and Harald Wajant
- Subjects
agonistic antibodies ,cell death ,Fn14 ,NFκB ,TNF ,TWEAK ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Fibroblast growth factor-inducible 14 (Fn14) is a member of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF) and is activated by its ligand TNF-like weak inducer of apoptosis (TWEAK). The latter occurs as a homotrimeric molecule in a soluble and a membrane-bound form. Soluble TWEAK (sTWEAK) activates the weakly inflammatory alternative NF-κB pathway and sensitizes for TNF-induced cell death while membrane TWEAK (memTWEAK) triggers additionally robust activation of the classical NF-κB pathway and various MAP kinase cascades. Fn14 expression is limited in adult organisms but becomes strongly induced in non-hematopoietic cells by a variety of growth factors, cytokines and physical stressors (e.g., hypoxia, irradiation). Since all these Fn14-inducing factors are frequently also present in the tumor microenvironment, Fn14 is regularly found to be expressed by non-hematopoietic cells of the tumor microenvironment and most solid tumor cells. In general, there are three possibilities how the tumor-Fn14 linkage could be taken into consideration for tumor therapy. First, by exploitation of the cancer associated expression of Fn14 to direct cytotoxic activities (antibody-dependent cell-mediated cytotoxicity (ADCC), cytotoxic payloads, CAR T-cells) to the tumor, second by blockade of potential protumoral activities of the TWEAK/Fn14 system, and third, by stimulation of Fn14 which not only triggers proinflammtory activities but also sensitizes cells for apoptotic and necroptotic cell death. Based on a brief description of the biology of the TWEAK/Fn14 system and Fn14 signaling, we discuss the features of the most relevant Fn14-targeting biologicals and review the preclinical data obtained with these reagents. In particular, we address problems and limitations which became evident in the preclinical studies with Fn14-targeting biologicals and debate possibilities how they could be overcome.
- Published
- 2022
- Full Text
- View/download PDF
17. Exocyst inactivation in urothelial cells disrupts autophagy and activates non-canonical NF-κB signaling
- Author
-
Michael A. Ortega, Ross K. Villiger, Malia Harrison-Chau, Suzanna Lieu, Kadee-Kalia Tamashiro, Amanda J. Lee, Brent A. Fujimoto, Geetika Y. Patwardhan, Joshua Kepler, and Ben Fogelgren
- Subjects
autophagy ,exocyst ,fn14 ,nf-κb signaling pathway ,urothelium ,Medicine ,Pathology ,RB1-214 - Published
- 2022
- Full Text
- View/download PDF
18. Dysregulation of Tweak and Fn14 in skeletal muscle of spinal muscular atrophy mice.
- Author
-
Meijboom, Katharina E., Sutton, Emma R., McCallion, Eve, McFall, Emily, Anthony, Daniel, Edwards, Benjamin, Kubinski, Sabrina, Tapken, Ines, Bünermann, Ines, Hazell, Gareth, Ahlskog, Nina, Claus, Peter, Davies, Kay E., Kothary, Rashmi, Wood, Matthew J. A., and Bowerman, Melissa
- Subjects
SPINAL muscular atrophy ,NECROSIS ,SKELETAL muscle ,METABOLIC regulation ,MUSCULAR atrophy ,NEUROMUSCULAR diseases ,SPINAL cord - Abstract
Background: Spinal muscular atrophy (SMA) is a childhood neuromuscular disorder caused by depletion of the survival motor neuron (SMN) protein. SMA is characterized by the selective death of spinal cord motor neurons, leading to progressive muscle wasting. Loss of skeletal muscle in SMA is a combination of denervation-induced muscle atrophy and intrinsic muscle pathologies. Elucidation of the pathways involved is essential to identify the key molecules that contribute to and sustain muscle pathology. The tumor necrosis factor-like weak inducer of apoptosis (TWEAK)/TNF receptor superfamily member fibroblast growth factor-inducible 14 (Fn14) pathway has been shown to play a critical role in the regulation of denervation-induced muscle atrophy as well as muscle proliferation, differentiation, and metabolism in adults. However, it is not clear whether this pathway would be important in highly dynamic and developing muscle. Methods: We thus investigated the potential role of the TWEAK/Fn14 pathway in SMA muscle pathology, using the severe Taiwanese Smn
−/− ; SMN2 and the less severe Smn2B/− SMA mice, which undergo a progressive neuromuscular decline in the first three post-natal weeks. We also used experimental models of denervation and muscle injury in pre-weaned wild-type (WT) animals and siRNA-mediated knockdown in C2C12 muscle cells to conduct additional mechanistic investigations. Results: Here, we report significantly dysregulated expression of Tweak, Fn14, and previously proposed downstream effectors during disease progression in skeletal muscle of the two SMA mouse models. In addition, siRNA-mediated Smn knockdown in C2C12 myoblasts suggests a genetic interaction between Smn and the TWEAK/Fn14 pathway. Further analyses of SMA, Tweak−/− , and Fn14−/− mice revealed dysregulated myopathy, myogenesis, and glucose metabolism pathways as a common skeletal muscle feature, providing further evidence in support of a relationship between the TWEAK/Fn14 pathway and Smn. Finally, administration of the TWEAK/Fn14 agonist Fc-TWEAK improved disease phenotypes in the two SMA mouse models. Conclusions: Our study provides mechanistic insights into potential molecular players that contribute to muscle pathology in SMA and into likely differential responses of the TWEAK/Fn14 pathway in developing muscle. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
19. The TWEAK/Fn14/CD163 axis—implications for metabolic disease.
- Author
-
Ratajczak, Wiktoria, Atkinson, Sarah D, and Kelly, Catriona
- Abstract
TWEAK (tumor necrosis factor-like weak inducer of apoptosis) is a member of the TNF superfamily that controls a multitude of cellular events including proliferation, migration, differentiation, apoptosis, angiogenesis, and inflammation. TWEAK control of these events is via an expanding list of intracellular signalling pathways which include NF-κB, ERK/MAPK, Notch, EGFR and AP-1. Two receptors have been identified for TWEAK – Fn14, which targets the membrane bound form of TWEAK, and CD163, which scavenges the soluble form of TWEAK. TWEAK appears to elicit specific events based on the receptor to which it binds, tissue type in which it is expressed, specific extrinsic conditions, and the presence of other cytokines. TWEAK signalling is protective in healthy tissues, but in chronic inflammatory states become detrimental to the tissue. Consistent data show a role for the TWEAK/FN14/CD163 axis in metabolic disease, chronic autoimmune diseases, and acute ischaemic stroke. Low circulating concentrations of soluble TWEAK are predictive of poor cardiovascular outcomes in those with and without diabetes. This review details the current understanding of the TWEAK/Fn14/CD163 axis as one of the chief regulators of immune signalling and its cell-specific role in metabolic disease development and progression. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. Proposed Hypothesis of TWEAK/Fn14 Receptor Modulation in Autism Spectrum Disorder.
- Author
-
Khan H, Rihal V, Kaur A, and Singh TG
- Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with a complex, multiple etiology that is marked by impaired social interaction, communication, and repetitive behaviour. There is presently no pharmaceutical treatment for the core symptoms of ASD, even though the prevalence of ASD is increasing worldwide. Treatment of autism spectrum disorder involves the interaction of numerous signalling pathways, such as the Wnt/beta-catenin pathway, probiotics and kynurenine pathway, PPAR pathway, PI3K-AKT-mTOR pathway, Hedgehog signaling pathway, etc. The scientific literature has revealed TWEAK/Fn14 to not be explored in the autism spectrum disorder. In vitro and in vivo, TWEAK can control a wide range of cellular responses. Recent research has revealed that TWEAK and Fn14 are expressed in the Central Nervous System (CNS) and upregulated in perivascular endothelial cells, astrocytes, neurons, and microglia in response to various stimuli, including cerebral ischemia. This upregulation is followed by cell death and an increase in Blood-brain Barrier (BBB) permeability. The study has revealed that Aurintricarboxylic Acid (ATA) acts as an agent that suppresses TWEAK/Fn14 signaling. Similarly, from the discussion, it has been emphasized that the proposed molecular TWEAK/Fn14 signalling pathway can be considered as a therapeutic approach in the management of autism spectrum disorder., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2024
- Full Text
- View/download PDF
21. Emerging role of TWEAK‐Fn14 axis in lupus, a disease related to autoimmunity and fibrosis.
- Author
-
Xu, Yuan, Wei, Hui‐Ting, Jing, Hao‐Yue, Tan, Xiao‐Yang, Zhou, Xiao‐Tao, and Ma, Yue‐Rong
- Subjects
- *
SYSTEMIC lupus erythematosus , *TUMOR necrosis factors , *AUTOIMMUNITY , *RENAL fibrosis , *FIBROSIS , *AUTOIMMUNE diseases - Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder. Lupus nephritis (LN) is one of the severe clinical implications in SLE, and this was relates to fibrosis in the kidney. As an important marker in the tumor necrosis factor (TNF) superfamily, TNF‐like weak inducer of apoptosis (TWEAK) has been given much attention with respect to its role in regulating pro‐inflammatory immune response. Fibroblast growth factor‐inducible 14 (Fn14), the sole receptor for TWEAK, has been found expressed in different immune and non‐immune cells. TWEAK binds to Fn14, and then regulates inflammatory components production via downstream signaling pathways. To date, dysregulated expression of TWEAK, Fn14 has been reported in SLE, LN patients, and in vivo, in vitro studies have discussed the significant role of TWEAK‐Fn14 axis in SLE, LN pathogenesis, partly through mediating the fibrosis process. In this review, we will discuss the association of TWEAK‐Fn14 axis in lupus. Understanding the relationship will better realize the potential for making TWEAK‐Fn14 as a marker for the diseases, and will help to give many clues for targeting them in treatment of lupus in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. Fn14 overcomes cisplatin resistance of high-grade serous ovarian cancer by promoting Mdm2-mediated p53-R248Q ubiquitination and degradation
- Author
-
An-Yue Wu, Li-Ying Gu, Wei Cang, Meng-Xing Cheng, Wen-Jing Wang, Wen Di, Lei Huang, and Li-Hua Qiu
- Subjects
HGSOC ,Cisplatin resistance ,Fn14 ,p53-R248Q ,Hsp90 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background High-grade serous ovarian cancer (HGSOC) is the most lethal of all gynecological malignancies. Patients often suffer from chemoresistance. Several studies have reported that Fn14 could regulate migration, invasion, and angiogenesis in cancer cells. However, its functional role in chemoresistance of HGSOC is still unknown. Methods The expression of Fn14 in tissue specimens was detected by IHC. CCK-8 assay was performed to determine changes in cell viability. Apoptosis was measured by flow cytometry. The potential molecular mechanism of Fn14-regulated cisplatin resistance in HGSOC was investigated using qRT-PCR, western blotting, and Co-IP assays. The role of Fn14 in HGSOC was also investigated in a xenograft mouse model. Results In this study, we found that Fn14 was significantly downregulated in patients with cisplatin resistance. Patients with low Fn14 expression were associated with shorter progression-free survival and overall survival. Overexpression of Fn14 suppressed cisplatin resistance in OVCAR-3 cells, whereas knockdown of Fn14 did not affect cisplatin resistance in SKOV-3 cells. Interestingly, Fn14 could exert anti-chemoresistance effect only in OVCAR-3 cells harboring a p53-R248Q mutation, but not in SKOV-3 cells with a p53-null mutation. We isolated and identified primary cells from two patients with the p53-R248Q mutation from HGSOC patients and the anti-chemoresistance effect of Fn14 was observed in both primary cells. Mechanistic studies demonstrated that overexpression of Fn14 could reduce the formation of a Mdm2-p53-R248Q-Hsp90 complex by downregulating Hsp90 expression, indicating that degradation of p53-R248Q was accelerated via Mdm2-mediated ubiquitin-proteasomal pathway. Conclusion Our findings demonstrate for the first time that Fn14 overcomes cisplatin resistance through modulation of the degradation of p53-R248Q and restoration of Fn14 expression might be a novel strategy for the treatment of HGSOC.
- Published
- 2019
- Full Text
- View/download PDF
23. Fn14
- Author
-
Martin-Sanchez, Diego, Fontecha-Barriuso, Miguel, Sanchez-Niño, Maria D., Izquierdo, Maria C., Ucero, Alvaro C., Ortiz, Alberto, Sanz, Ana B., and Choi, Sangdun, editor
- Published
- 2018
- Full Text
- View/download PDF
24. Inhibition of fibroblast growth factor-inducible 14 attenuates experimental tubulointerstitial fibrosis and profibrotic factor expression of proximal tubular epithelial cells.
- Author
-
Luo, Mai, Liu, Mengmeng, Liu, Wei, Cui, Xiao, Zhai, Siyue, Gu, Hanjiang, Wang, Huixia, Wu, Kunyi, Zhang, Wen, Li, Ke, and Xia, Yumin
- Subjects
- *
EPITHELIAL cells , *RENAL fibrosis , *FIBROSIS , *EXTRACELLULAR matrix , *URETERIC obstruction - Abstract
Background and aim: As a proinflammatory cytokine, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) participates in the progression of renal fibrosis by binding to its receptor, fibroblast growth factor-inducible 14 (Fn14). However, the effect of Fn14 inhibition on tubular epithelial cell-mediated tubulointerstitial fibrosis remains unclear. This study aimed to elucidate the role of TWEAK/Fn14 interaction in the development of experimental tubulointerstitial fibrosis as well as the protective effect of Fn14 knockdown on proximal tubular epithelial cells. Methods: A murine model of unilateral ureteral obstruction was constructed in both wild-type and Fn14-deficient BALB/c mice, followed by observation of the tubulointerstitial pathologies. Results: Fn14 deficiency ameliorated the pathological changes, including inflammatory cell infiltration and cell proliferation, accompanied by reduced production of profibrotic factors and extracellular matrix deposition. In vitro experiments showed that TWEAK dose-dependently enhanced the expression of collagen I, fibronectin, and α-smooth muscle actin in proximal tubular epithelial cells. Interestingly, TWEAK also upregulated the expression levels of Notch1/Jagged1. Fn14 knockdown and Notch1/Jagged1 inhibition also mitigated the effect of TWEAK on these cells. Conclusions: In conclusion, TWEAK/Fn14 signals contributed to tubulointerstitial fibrosis by acting on proximal tubular epithelial cells. Fn14 inhibition might be a therapeutic strategy for protecting against renal interstitial fibrosis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. TWEAK/Fn14 axis is an important player in fibrosis.
- Author
-
Zhang, Yitian, Zeng, Weihui, and Xia, Yumin
- Subjects
- *
TUMOR necrosis factors , *FIBROBLAST growth factors , *FIBROSIS , *GROWTH factors , *EXTRACELLULAR matrix , *SOFT tissue injuries , *MYOFIBROBLASTS - Abstract
Fibrosis is a common pathological condition associated with abnormal repair after tissue injury. However, the etiology and molecular mechanisms of fibrosis are still not well‐understood. Tumor necrosis factor (TNF)‐like weak inducer of apoptosis (TWEAK) belongs to the TNF superfamily and acts by binding to its receptor, fibroblast growth factor‐inducible 14 (Fn14), thereby activating a variety of intracellular signal transduction pathways in various types of cells. Besides promoting the expression of growth factors, activation of TWEAK/Fn14 signaling after tissue injury can promote the expression of pro‐inflammatory cytokines, which trigger the immune response, thereby exacerbating the injury. Severe or repetitive injury leads to a dysregulated tissue repair process, in which the TWEAK/Fn14 axis promotes the activation and proliferation of myofibroblasts, induces the secretion of the extracellular matrix, and regulates profibrotic mediators to further perpetuate and sustain the fibrotic process. In this review, we summarize the available experimental evidence on the underlying molecular mechanisms by which the TWEAK/Fn14 pathway mediates the development and progression of fibrosis. In addition, we discuss the therapeutic potential of the TWEAK/Fn14 pathway in fibrosis‐associated diseases based on evidence derived from multiple models and cells from injured tissue and fibrotic tissue. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
26. Furin is involved in uterine activation for labor.
- Author
-
Zhang, Youyi, Xu, Chen, Tang, Zhengshan, Guo, Dewei, Yao, Ruojin, Zhao, Huina, Chen, Zixi, and Ni, Xin
- Abstract
The uterus undergoes distinct molecular and functional changes during pregnancy and parturition. These processes are associated with the dramatic changes in various proteins. Given that the maturation and activation of many proteins require proteolytic processing by proprotein convertases (PCs), we sought to explore the role of PCs in uterine activation for labor. First, we found that furin was the most dramatically increased PC member in myometrial tissues from the pregnant women after onset of labor at term. Using the model of cultured human myometrial smooth muscle cells (HMSMCs), we showed that furin inhibitor CMK, D6R treatment and furin siRNA transfection suppressed contractility. Inhibition of furin activity or interfering furin expression decreased connexin 43 (CX43), prostaglandin (PG) endoperoxide synthase‐2 (COX‐2) and PGF2α receptor (FP) expression and NF‐κB activation. In mouse model, administration of furin inhibitors prolonged gestational length. However, D6R treatment did not affect RU38486‐ and lipopolysaccharides (LPS)‐induced preterm birth. Furthermore, D6R and furin siRNA treatment reduced the release of soluble form of tumor necrosis factor (TNF)‐related weak inducer of apoptosis (TWEAK), while furin overexpression led to an increase in soluble TWEAK release in cultured HMSMCs. D6R treatment decreased TWEAK level in blood of pregnant mice. TWEAK treatment promoted contractility and NF‐κB activation, while TWEAK receptor fibroblast growth factor‐inducible 14 (FN14) antagonist treatment inhibited contractility and NF‐κB activation in HMSMCs. In pregnant mice, administration of FN14 antagonist prolonged gestational length. Our data suggest that furin can act as a stimulator for uterine activation for labor at term. TWEAK is one of the potential substrates which mediate furin regulation of parturition initiation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
27. TWEAK–Fn14 as a common pathway in the heart and the kidneys in cardiorenal syndrome.
- Author
-
Poveda, Jonay, Vázquez‐Sánchez, Sara, Sanz, Ana B, Ortiz, Alberto, Ruilope, Luis M, and Ruiz‐Hurtado, Gema
- Subjects
HEART failure ,RENAL fibrosis ,KIDNEYS ,SYNDROMES ,CHRONIC kidney failure ,CARDIOVASCULAR diseases - Abstract
There is a complex relationship between cardiac and renal disease, often referred to as the cardiorenal syndrome. Heart failure adversely affects kidney function, and both acute and chronic kidney disease are associated with structural and functional changes to the myocardium. The pathological mechanisms and contributing interactions that surround this relationship remain poorly understood, limiting the opportunities for therapeutic intervention. The cytokine tumor necrosis factor‐like weak inducer of apoptosis (TWEAK) and its receptor, fibroblast growth factor‐inducible 14 (Fn14), are abundantly expressed in injured kidneys and heart. The TWEAK–Fn14 axis promotes responses that drive tissue injury such as inflammation, proliferation, fibrosis, and apoptosis, while restraining the expression of tissue protective factors such as the anti‐aging factor Klotho and the master regulator of mitochondrial biogenesis peroxisome proliferator‐activated receptor‐γ coactivator‐1α (PGC‐1α). High levels of TWEAK induce cardiac remodeling, and promote inflammation, tubular and podocyte injury and death, fibroblast proliferation, and, ultimately, renal fibrosis. Accordingly, targeting the TWEAK–Fn14 axis is protective in experimental kidney and heart disease. TWEAK has also emerged as a biomarker of kidney damage and cardiovascular outcomes and has been successfully targeted in clinical trials. In this review, we update our current knowledge of the roles of the TWEAK–Fn14 axis in cardiovascular and kidney disease and its potential contribution to the cardiorenal syndrome. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
28. TWEAK/Fn14 signalling promotes cholangiocarcinoma niche formation and progression.
- Author
-
Dwyer, Benjamin J., Jarman, Edward J., Gogoi-Tiwari, Jully, Ferreira-Gonzalez, Sofia, Boulter, Luke, Guest, Rachel V., Kendall, Timothy J., Kurian, Dominic, Kilpatrick, Alastair M., Robson, Andrew J., O'Duibhir, Eoghan, Man, Tak Yung, Campana, Lara, Starkey Lewis, Philip J., Wigmore, Stephen J., Olynyk, John K., Ramm, Grant A., Tirnitz-Parker, Janina E.E., and Forbes, Stuart J.
- Subjects
- *
HUMAN carcinogenesis , *MONOCYTE chemotactic factor , *CHOLANGIOCARCINOMA , *LIVER cancer , *CELL aggregation , *PROGNOSIS - Abstract
Cholangiocarcinoma (CCA) is a cancer of the hepatic bile ducts that is rarely resectable and is associated with poor prognosis. Tumour necrosis factor-like weak inducer of apoptosis (TWEAK) is known to signal via its receptor fibroblast growth factor-inducible 14 (Fn14) and induce cholangiocyte and myofibroblast proliferation in liver injury. We aimed to characterise its role in CCA. The expression of the TWEAK ligand and Fn14 receptor was assessed immunohistochemically and by bulk RNA and single cell transcriptomics of human liver tissue. Spatiotemporal dynamics of pathway regulation were comprehensively analysed in rat and mouse models of thioacetamide (TAA)-mediated CCA. Flow cytometry, qPCR and proteomic analyses of CCA cell lines and conditioned medium experiments with primary macrophages were performed to evaluate the downstream functions of TWEAK/Fn14. In vivo pathway manipulation was assessed via TWEAK overexpression in NICD/AKT-induced CCA or genetic Fn14 knockout during TAA-mediated carcinogenesis. Our data reveal TWEAK and Fn14 overexpression in multiple human CCA cohorts, and Fn14 upregulation in early TAA-induced carcinogenesis. TWEAK regulated the secretion of factors from CC-SW-1 and SNU-1079 CCA cells, inducing polarisation of proinflammatory CD206+ macrophages. Pharmacological blocking of the TWEAK downstream target chemokine monocyte chemoattractant protein 1 (MCP-1 or CCL2) significantly reduced CCA xenograft growth, while TWEAK overexpression drove cancer-associated fibroblast proliferation and collagen deposition in the tumour niche. Genetic Fn14 ablation significantly reduced inflammatory, fibrogenic and ductular responses during carcinogenic TAA-mediated injury. These novel data provide evidence for the action of TWEAK/Fn14 on macrophage recruitment and phenotype, and cancer-associated fibroblast proliferation in CCA. Targeting TWEAK/Fn14 and its downstream signals may provide a means to inhibit CCA niche development and tumour growth. Cholangiocarcinoma is an aggressive, chemotherapy-resistant liver cancer. Interactions between tumour cells and cells that form a supportive environment for the tumour to grow are a source of this aggressiveness and resistance to chemotherapy. Herein, we describe interactions between tumour cells and their supportive environment via a chemical messenger, TWEAK and its receptor Fn14. TWEAK/Fn14 alters the recruitment and type of immune cells in tumours, increases the growth of cancer-associated fibroblasts in the tumour environment, and is a potential target to reduce tumour formation. [Display omitted] • Fn14 is upregulated on tumour cells and CAFs, and TWEAK is expressed by TAMs in human CCA. • TWEAK/Fn14 are progressively upregulated during rodent tumour development. • TWEAK/Fn14 signalling induces TAM accumulation via TWEAK-inducible MCP-1 chemotaxis which can be blocked in vivo. • TWEAK-inducible factors from tumour cells pattern macrophages to a TAM-like phenotype. • TWEAK overexpression in experimental tumour formation drives CAF proliferation, collagen deposition and increases macrophages in mice. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
29. Fn14-targeted BiTE and CAR-T cells demonstrate potent preclinical activity against glioblastoma
- Author
-
Gaowei Li, Zongliang Zhang, Linjun Cai, Xin Tang, Jianhan Huang, Lingyu Yu, Guoqing Wang, Kunhong Zhong, Yi Cao, Chang Liu, Yuelong Wang, Aiping Tong, and Liangxue Zhou
- Subjects
gbm ,fn14 ,bite ,car-t ,immunotherapy ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
T cell-engaging therapies involving bispecific T cell engager (BiTE) and chimeric antigen receptor T (CAR-T) cells have achieved great success in the treatment of hematological tumors. However, the paucity of ideal cell surface molecules that can be targeted on glioblastoma (GBM) partially reduces the immunotherapeutic efficacy. Recently, high expression of Fn14 has been reported in several solid tumors, so the strategy of exploiting this specific antigen for GBM immunotherapy is worth studying. Consequently, we constructed Fn14× CD3 BiTE and Fn14-specific CAR-T cells and investigated their cytotoxic activity against GBM in vitro and in vivo. First, expression of Fn14 was confirmed in glioma tissues and GBM cells. Then, we designed Fn14-specific BiTE and CAR-T cells and tested their cytotoxicity in GBM cell cultures and mouse models of GBM. Fn14 was highly expressed in GBM tissues and cell lines, while it was undetectable in normal brain samples. Fn14× CD3 BiTE, Fn14 CAR-T cells and Fn14 CAR-T/IL-15 cells were antigen-specific and highly cytotoxic, showing good antitumor activity in vitro and causing significant regression of established solid tumors in xenograft models. However, the xenografts treated with Fn14 CAR-T cells regrew, whereas xenografts treated with Fn14 CAR-T/IL-15 cells did not. IL-15 engineering augmented the antitumor activity of Fn14 CAR-T cells and resulted in significant antitumor effects similar to those of Fn14× CD3 BiTE. Our results suggest that Fn14 is an appropriate target for GBM. Anti-Fn14 BiTE and Fn14-specific CAR-T/IL-15 cells may be exciting immunotherapeutic options for malignant brain cancer.
- Published
- 2021
- Full Text
- View/download PDF
30. TWEAK Promotes the Proliferation of Squamous Cell Carcinoma Cells Through Activating cIAP1 Signals
- Author
-
Lili Liang, Chuantao Cheng, Guanglei Hu, Xuening Wang, Jing Liu, Zhu Yan, Weihui Zeng, and Yumin Xia
- Subjects
cIAP1 ,Fn14 ,proliferation ,squamous cell carcinoma ,TWEAK ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Recent studies showed that tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) induces the proliferation of squamous cell carcinoma (SCC) cells. However, the precise mechanism underlying such effect of TWEAK remains unclear. This study was designed to elucidate the role of cellular inhibitor of apoptosis 1 (cIAP1) in TWEAK-induced proliferation of SCC cells. Human SCC cells (SCC-13, A431, and SCC-9) were cultured in vitro, receiving the stimulation of TWEAK or TNF-related apoptosis-inducing ligand (TRAIL). We found that TWEAK induced cytoplasmic cIAP1 importation and RIP1 ubiquitination in cells, followed by the activation of canonical nuclear factor kappa B signals. MV1, a cIAP1 inhibitor, abrogated TWEAK-induced proliferation of these cells. Moreover, the interaction between TWEAK and its receptor, fibroblast growth factor-inducible 14 (Fn14), enhanced the expression of TRAIL receptor types 3 and 4 (TRAIL-R3/4). Furthermore, the transfection of TRAIL-R3/4 siRNA abrogated the promotion effect of TWEAK on SCC-13 cell proliferation and cIAP1 expression. Therefore, TWEAK/Fn14 interaction promotes the proliferation of SCC cells through activating cIAP1 signals. Targeting the downstream cIAP1 signals might attenuate the effect of TWEAK on SCC cells.
- Published
- 2020
- Full Text
- View/download PDF
31. Developmental synaptic regulator, TWEAK/Fn14 signaling, is a determinant of synaptic function in models of stroke and neurodegeneration.
- Author
-
Nagy, Dávid, Ennis, Katelin A., Wei, Ru, Su, Susan C., Hinckley, Christopher A., Rong-Fang Gu, Benbo Gao, Massol, Ramiro H., Ehrenfels, Chris, Jandreski, Luke, Thomas, Ankur M., Nelson, Ashley, Gyoneva, Stefka, Hajós, Mihály, and Burkly, Linda C.
- Subjects
- *
CENTRAL nervous system , *NEURAL circuitry , *NEURAL transmission , *NEUROPLASTICITY , *NEUROLOGICAL disorders - Abstract
Identifying molecular mediators of neural circuit development and/or function that contribute to circuit dysfunction when aberrantly reengaged in neurological disorders is of high importance. The role of the TWEAK/Fn14 pathway, which was recently reported to be a microglial/neuronal axis mediating synaptic refinement in experience-dependent visual development, has not been explored in synaptic function within the mature central nervous system. By combining electrophysiological and phosphoproteomic approaches, we show that TWEAK acutely dampens basal synaptic transmission and plasticity through neuronal Fn14 and impacts the phosphorylation state of pre- and postsynaptic proteins in adult mouse hippocampal slices. Importantly, this is relevant in two models featuring synaptic deficits. Blocking TWEAK/Fn14 signaling augments synaptic function in hippocampal slices from amyloid-beta-overexpressing mice. After stroke, genetic or pharmacological inhibition of TWEAK/Fn14 signaling augments basal synaptic transmission and normalizes plasticity. Our data support a glial/neuronal axis that critically modifies synaptic physiology and pathophysiology in different contexts in the mature brain and may be a therapeutic target for improving neurophysiological outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
32. Fibroblast growth factor inducible 14 signaling facilitates anti‐dsDNA IgG penetration into mesangial cells.
- Author
-
Li, Ruilian, Jia, Fangyan, Ren, Kaixuan, Luo, Mai, Min, Xiaoyun, Xiao, Shengxiang, and Xia, Yumin
- Subjects
- *
SUPPRESSORS of cytokine signaling , *SYSTEMIC lupus erythematosus , *FIBROBLAST growth factors , *CELLS - Abstract
Anti‐double‐stranded DNA (dsDNA) antibodies induce renal damage in patients with systemic lupus erythematosus by triggering fibrotic processes in kidney cells. However, the precise mechanism underlying penetration of anti‐dsDNA immunoglubolin G (IgG) into cells remains unclear. This study was designed to investigate the effect of tumor necrosis factor‐like weak inducer of apoptosis (TWEAK)/fibroblast growth factor inducible 14 (Fn14) signaling on anti‐dsDNA IgG penetration into cells. Mesangial cells were cultured in vitro, and stimulated with TWEAK and anti‐dsDNA IgG. The results revealed that TWEAK dose‐dependently enhanced cellular internalization of anti‐dsDNA IgG and the expression of high‐mobility group box 1 (HMGB1). In addition, TWEAK and anti‐dsDNA IgG synthetically downregulate suppressor of cytokine signaling 1, and induce the expression of various fibrotic factors. Furthermore, inhibition of HMGB1 attenuates the enhancement effect of TWEAK on anti‐dsDNA IgG internalization. The TWEAK upregulation of HMGB1 involves the nuclear factor‐κB and phosphatidylinositide 3‐kinase/protein kinase B pathways. Therefore, TWEAK/Fn14 signaling contributes to the penetration of anti‐dsDNA IgG and relevant fibrotic processes in mesangial cells. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
33. TWEAK/Fn14 axis in respiratory diseases.
- Author
-
Wang, Min, Xie, Zhijuan, Xu, Jin, and Feng, Zhuyu
- Subjects
- *
RESPIRATORY diseases , *OBSTRUCTIVE lung diseases , *IDIOPATHIC pulmonary fibrosis , *NON-small-cell lung carcinoma , *SLEEP apnea syndromes - Abstract
• TWEAK, acting on Fn14, takes part in diverse pathological processes. • TWEAK has key functions in non-respiratory disease and respiratory disease. • Various signaling pathways take part in the respiratory disease. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a well known multifunctional cytokine extensively distributed in cell types and tissues. Accumulating evidence has shown that TWEAK binding to the receptor factor-inducible 14 (Fn14) participates in diverse pathologic processes including cell proliferation and death, angiogenesis, carcinogenesis and inflammation. Interestingly, alterations of intracellular signaling cascades are correlated to the development of respiratory disease. Recently, a several lines of evidence suggests that TWEAK in lung tissues are closely associated with these signaling pathways. In this review, we explore if TWEAK could provide a novel therapeutic strategy for managing respiratory disease in general and pulmonary arterial hypertension (PAH), obstructive sleep apnea syndrome (OSAS), asthma, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC), specifically. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
34. TWEAK Promotes the Proliferation of Squamous Cell Carcinoma Cells Through Activating cIAP1 Signals.
- Author
-
Liang, Lili, Cheng, Chuantao, Hu, Guanglei, Wang, Xuening, Liu, Jing, Yan, Zhu, Zeng, Weihui, and Xia, Yumin
- Subjects
SQUAMOUS cell carcinoma ,CELL proliferation ,NF-kappa B ,TUMOR necrosis factors - Abstract
Recent studies showed that tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) induces the proliferation of squamous cell carcinoma (SCC) cells. However, the precise mechanism underlying such effect of TWEAK remains unclear. This study was designed to elucidate the role of cellular inhibitor of apoptosis 1 (cIAP1) in TWEAK-induced proliferation of SCC cells. Human SCC cells (SCC-13, A431, and SCC-9) were cultured in vitro , receiving the stimulation of TWEAK or TNF-related apoptosis-inducing ligand (TRAIL). We found that TWEAK induced cytoplasmic cIAP1 importation and RIP1 ubiquitination in cells, followed by the activation of canonical nuclear factor kappa B signals. MV1, a cIAP1 inhibitor, abrogated TWEAK-induced proliferation of these cells. Moreover, the interaction between TWEAK and its receptor, fibroblast growth factor-inducible 14 (Fn14), enhanced the expression of TRAIL receptor types 3 and 4 (TRAIL-R3/4). Furthermore, the transfection of TRAIL-R3/4 siRNA abrogated the promotion effect of TWEAK on SCC-13 cell proliferation and cIAP1 expression. Therefore, TWEAK/Fn14 interaction promotes the proliferation of SCC cells through activating cIAP1 signals. Targeting the downstream cIAP1 signals might attenuate the effect of TWEAK on SCC cells. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
35. Tumor Necrosis Factor Receptor Mediates Fibroblast Growth Factor-Inducible 14 Signaling
- Author
-
Xuening Wang, Shengxiang Xiao, and Yumin Xia
- Subjects
TWEAK ,Fn14 ,TRAF ,TNFR ,Apoptosis ,Proliferation ,Keratinocyte ,Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Abstract
Tumor necrosis factor (TNF)-related weak inducer of apoptosis (TWEAK) engages its sole receptor, fibroblast growth factor–inducible 14 (Fn14), which participates in various inflammatory and immunologic processes. TWEAK/Fn14 interaction induces different cell fates depending on the local microenvironment, which correlates with certain expression profiles of TNF receptors (TNFR). The predominant expression of TNFR1 or TNFR2 facilitates cell death or proliferation, respectively, on TWEAK/Fn14 activation. TNFR-associated factors (TRAF) interact with Fn14, cellular inhibitor of apoptosis protein (cIAP)-1, and TNFR, consequently transducing signals from TWEAK to downstream cytokines and cell cycle mediators. An Fn14-TRAF2-TNFR axis has been suggested in the function of TWEAK/Fn14 signaling, which may serve as a target in the development of novel therapeutic strategies for many diseases that have Fn14-overexpressing cells in affected tissues. The aims of this review are: 1) to present the main results on TWEAK/Fn14 regulation of cell fates, 2) to analyze the mechanism of the Fn14-TRAF2-TNFR axis, and 3) to summarize the potential strategies in the pharmacologic targeting of this axis.
- Published
- 2017
- Full Text
- View/download PDF
36. Integrative Keratinocyte Responses to TWEAK with IL-13 and IL-22 Reveal Pathogenic Transcriptomes Associated with Atopic Dermatitis.
- Author
-
Gupta RK, Fung K, Figueroa DS, Ay F, and Croft M
- Subjects
- Humans, Cytokine TWEAK genetics, Cytokine TWEAK metabolism, Dermatitis, Atopic genetics, Dermatitis, Atopic immunology, Interleukin-13 genetics, Interleukin-13 metabolism, Interleukin-22, Keratinocytes metabolism, Transcriptome
- Published
- 2024
- Full Text
- View/download PDF
37. Anti-Fn14 Antibody-Conjugated Nanoparticles Display Membrane TWEAK-Like Agonism
- Author
-
Ahmed Aido, Olena Zaitseva, Harald Wajant, Matej Buzgo, and Aiva Simaite
- Subjects
Fn14 ,nanoparticles ,surface modification ,drug-delivery ,anti-TNFRSF receptor (TNFR) antibodies ,Pharmacy and materia medica ,RS1-441 - Abstract
Conventional bivalent IgG antibodies targeting a subgroup of receptors of the TNF superfamily (TNFSF) including fibroblast growth factor-inducible 14 (anti-Fn14) typically display no or only very limited agonistic activity on their own and can only trigger receptor signaling by crosslinking or when bound to Fcγ receptors (FcγR). Both result in proximity of multiple antibody-bound TNFRSF receptor (TNFR) molecules, which enables engagement of TNFR-associated signaling pathways. Here, we have linked anti-Fn14 antibodies to gold nanoparticles to mimic the “activating” effect of plasma membrane-presented FcγR-anchored anti-Fn14 antibodies. We functionalized gold nanoparticles with poly-ethylene glycol (PEG) linkers and then coupled antibodies to the PEG surface of the nanoparticles. We found that Fn14 binding of the anti-Fn14 antibodies PDL192 and 5B6 is preserved upon attachment to the nanoparticles. More importantly, the gold nanoparticle-presented anti-Fn14 antibody molecules displayed strong agonistic activity. Our results suggest that conjugation of monoclonal anti-TNFR antibodies to gold nanoparticles can be exploited to uncover their latent agonism, e.g., for immunotherapeutic applications.
- Published
- 2021
- Full Text
- View/download PDF
38. Fn14 Participates in Neuropathic Pain Through NF-κB Pathway in Primary Sensory Neurons.
- Author
-
Huang, Li-Na, Zou, Yun, Wu, Shao-Gen, Zhang, Hong-Hong, Mao, Qing-Xiang, Li, Jin-Bao, and Tao, Yuan-Xiang
- Abstract
Fibroblast growth factor-inducible-14 (Fn14), a receptor for tumor necrosis-like weak inducer of apoptosis, is expressed in the neurons of dorsal root ganglion (DRG). Its mRNA is increased in the injured DRG following peripheral nerve injury. Whether this increase contributes to neuropathic pain is unknown. We reported here that peripheral nerve injury caused by spinal nerve ligation (SNL) increased the expression of Fn14 at both protein and mRNA levels in the injured DRG. Blocking this increase attenuated the development of SNL-induced mechanical, thermal, and cold pain hypersensitivities. Conversely, mimicking this increase produced the increases in the levels of phosphorylated extracellular signal-regulated kinase ½ and glial fibrillary acidic protein in ipsilateral dorsal horn and the enhanced responses to mechanical, thermal, and cold stimuli in the absence of SNL. Mechanistically, the increased Fn14 activated the NF-κB pathway through promoting the translocation of p65 into the nucleus of the injured DRG neurons. Our findings suggest that Fn14 may be a potential target for the therapeutic treatment of peripheral neuropathic pain. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
39. Tweak/Fn14 system is involved in rhabdomyolysis-induced acute kidney injury.
- Author
-
Guerrero-Hue, Melania, Vallejo-Mudarra, Mercedes, García-Caballero, Cristina, Córdoba-David, Gina Marcela, Palomino-Antolín, Alejandra, Herencia, Carmen, Vendrell-Casana, Beatriz, Rubio-Navarro, Alfonso, Egido, Jesús, Blanco-Colio, Luis Miguel, and Moreno, Juan Antonio
- Subjects
- *
ACUTE kidney failure , *KIDNEY diseases , *RENAL fibrosis , *CHRONIC kidney failure , *GENE expression , *CELL death - Abstract
Rhabdomyolysis is a severe clinical syndrome associated to acute kidney injury (AKI) and chronic kidney disease (CKD). TWEAK/Fn14 signaling axis regulates renal inflammation and tubular cell death. However, the functional role of TWEAK/Fn14 in rhabdomyolysis remains unknown. Rhabdomyolysis was induced in wild-type, TWEAK- and Fn14-deficient mice or mice treated with TWEAK blocking antibody. Renal injury, inflammation, fibrosis and cell death were assessed. Additionally, we performed in vivo and in vitro studies to explore the possible signalling pathways involved in Fn14 regulation. Fn14 renal expression was increased in mice with rhabdomyolysis, correlating with decline of renal function. Mechanistically, myoglobin (Mb) induced Fn14 expression via ERK and p38 pathway, whereas Nrf2 activation diminished Mb-mediated Fn14 upregulation in cultured renal cells. TWEAK or Fn14 genetic depletion ameliorated rhabdomyolysis-associated loss of renal function, histological damage, tubular cell death, inflammation, and expression of both tubular and endothelial injury markers. Deficiency of TWEAK or Fn14 also decreased long-term renal inflammation and fibrosis in mice with rhabdomyolysis. Finally, pharmacological treatment with a blocking TWEAK antibody diminished the expression of acute renal injury markers and cell death and lessened residual kidney fibrosis and chronic inflammation in rhabdomyolysis. TWEAK/Fn14 axis participates in the pathogenesis of rhabdomyolysis-AKI and subsequent AKI-CKD transition. Blockade of this signaling pathway may represent a promising therapeutic strategy for reducing rhabdomyolysis-mediated renal injury. Spanish Ministry of Science and Innovation, ISCIII and Junta de Andalucía. [Display omitted] • Rhabdomyolysis-derived myoglobin upregulates Fn14 expression in the kidney. • Genetic deletion of TWEAK or Fn14 reduces acute renal damage during rhabdomyolysis. • TWEAK/Fn14 axis participates in rhabdomyolysis-mediated fibrosis and inflammation. • Anti-TWEAK therapy reduces renal injury at early and late phases of rhabdomyolysis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Novel molecular factors in cardiac hypertrophic response
- Author
-
Säkkinen, H. (Hanna), Rysä, J. (Jaana), and Ruskoaho, H. (Heikki)
- Subjects
dyksiini ,sydänlihassolujen liikakasvu ,cardiac hypertrophy ,TWEAK ,LMCD1 ,Fn14 ,heart failure ,Reg3γ ,sydämen vajaatoiminta - Abstract
Cardiac hypertrophy is an adaptive mechanism for maintaining adequate cardiac output during increased workload. In prolonged situations, this process becomes maladaptive and leads to cardiac dysfunction and even heart failure (HF). The molecular mechanisms responsible for left ventricular (LV) remodeling have been extensively studied, and this research has been instrumental in developing pharmacological treatments decreasing mortality in HF patients. However, there is no cure for HF if heart transplantation is not an option. Transcriptomics studies have provided a tool for screening activated genes in diseased tissue. The goal of this work was to characterize the role of four selected genes from microarray screenings in cardiac hypertrophic response. Tumour necrosis factor -like weak inducer of apoptosis (TWEAK), fibroblast growth factor inducible 14 (Fn14), regenerating islet-derived 3 gamma (Reg3γ), and dyxin, known as LIM and cysteine-rich domains 1 (LMCD1) have been previously shown to be upregulated in different experimental models of cardiac stress. The expression and regulation of TWEAK, Fn14 and Reg3γ were examined in different models of cardiac stress in vivo and in vitro. In addition, the effect of intramyocardial adenoviral gene transfer-inducing overexpression of dyxin on cardiac structure and function was examined during angiotensin II (Ang II)-induced hypertension. The study showed that Fn14 and Reg3γ were rapidly upregulated due to pressure overload and post-infarction remodeling in vivo along with some known inducers of cardiac myocyte hypertrophy in vitro. Immunohistochemical analysis showed that Reg3γ was mainly localized in myofibroblasts in the inflammatory area, while Fn14 showed progressive immunoreactivity in fibroblasts. TWEAK was localized in cardiac myocytes and endothelial cells, but it did not respond to any of the hypertrophic stimuli. Overexpression of cardiac dyxin resulted in thickening of LV structure during Ang II-induced hypertension together with attenuation of the activation of certain cardiac hypertrophy-associated genes. This study provides new information on the gene expression of novel factors in pathological cardiac hypertrophy as well as new insights for pharmacological therapies in HF. Tiivistelmä Suurentuneessa kuormituksessa työskentelevä sydän joutuu sopeutumaan uuteen tilanteeseen kasvattamalla sydänlihassolujen kokoa, jotta sydämen minuuttitilavuus säilyisi ennallaan. Pitkittyessään prosessi muuttuu patologiseksi ja johtaa sydämen vajaatoiminnan kehittymiseen. Tällöin sydänlihassolujen geenien ilmentyminen muuttuu. Muutoksia on tutkittu laajasti, ja näiden tutkimusten pohjalta on kehitetty lääkehoitoja, jotka parantavat sydämen vajaatoimintaa sairastavien potilaiden ennustetta. Parantavaa lääkehoitoa ei sairauteen kuitenkaan ole tarjolla. Vain harvojen potilaiden kohdalla sydämen siirto on mahdollinen parannuskeino. Koko genomin kattavat transkriptomiikkatutkimukset ovat antaneet viitteitä useiden geenien osuudesta sydämen hypertrofian kehittymisessä. Tässä väitöskirjatyössä tutkittiin neljän geenin, TWEAK:in, Fn14:n, Reg3γ:n ja dyksiinin, merkitystä sydämessä hypertrofian kehittymisen näkökulmasta. TWEAK-, Fn14- ja Reg3γ-geenien ilmentymistä ja säätelyä tutkittiin sydämen kuormitusmalleissa koe-eläimillä sekä sydänlihassoluviljelmissä. Lisäksi koe-eläimillä tutkittiin adenovirusvälitteisen dyksiini-geenin yli-ilmentymisen vaikutuksia sydämessä painekuormituksen aikana. Tutkimus osoitti, että Fn14 ja Reg3γ aktivoituivat sekä sydämen painekuormituksessa että infarktin jälkeisen uudelleen muovautumisen aikana niin koe-eläimillä kuin sydänlihassolujen liikakasvua aiheuttavien tekijöiden vaikutuksesta sydänlihassoluviljelmissä. Immunohistologisessa tutkimuksessa Reg3γ paikantui pääasiassa tulehdusalueen myofibroblasteihin ja Fn14 fibroblasteihin, ja Fn14:n värjäytyminen tehostui fibroosin edetessä. Sydänlihassoluihin ja verisuonten endoteelisoluihin paikantuva TWEAK ei juurikaan aktivoitunut kyseisissä koemalleissa. Dyksiinin yli-ilmentyminen painekuormituksen aikana lisäsi sydämen vasemman kammion seinämäpaksuutta ja kumosi eräiden sydämen liikakasvuun liittyvien geenien painekuormituksen aiheuttaman aktivoitumisen. Väitöskirjatutkimus tarjoaa uutta tietoa eräiden uusien geenien ilmentymisestä sydämen hypertrofian kehittymisen aikana ja antaa uusia näkökulmia sydämen vajaatoiminnan lääkehoitojen kehitykselle.
- Published
- 2023
41. Topical TWEAK Accelerates Healing of Experimental Burn Wounds in Mice
- Author
-
Jing Liu, Lingling Peng, Yale Liu, Kunyi Wu, Sijia Wang, Xuening Wang, Qilu Liu, Yumin Xia, and Weihui Zeng
- Subjects
TWEAK ,Fn14 ,burn wound ,animal model ,differentiation ,dermal fibroblast ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The interaction of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor inducible 14 (Fn14) participates in inflammatory responses, fibrosis, and tissue remodeling, which are central in the repair processes of wounds. Fn14 is expressed in main skin cells including dermal fibroblasts. This study was designed to explore the therapeutic effect of TWEAK on experimental burn wounds and the relevant mechanism underlying such function. Third-degree burns were introduced in two BALB/c mouse strains. Recombinant TWEAK was administrated topically, followed by the evaluation of wound areas and histologic changes. Accordingly, the downstream cytokines, inflammatory cell infiltration, and extracellular matrix synthesis were examined in lesional tissue. Moreover, the differentiation markers were analyzed in cultured human dermal fibroblasts upon TWEAK stimulation. The results showed that topical TWEAK accelerated the healing of burn wounds in wild-type mice but not in Fn14-deficient mice. TWEAK strengthened inflammatory cell infiltration, and exaggerated the production of growth factor and extracellular matrix components in wound areas of wild-type mice. Moreover, TWEAK/Fn14 activation elevated the expression of myofibroblastic differentiation markers, including alpha-smooth muscle actin and palladin, in cultured dermal fibroblasts. Therefore, topical TWEAK exhibits therapeutic effect on experimental burn wounds through favoring regional inflammation, cytokine production, and extracellular matrix synthesis. TWEAK/Fn14 activation induces the myofibroblastic differentiation of dermal fibroblasts, partially contributing to the healing of burn wounds.
- Published
- 2018
- Full Text
- View/download PDF
42. TWEAK Receptor Deficiency Has Opposite Effects on Female and Male Mice Subjected to Neonatal Hypoxia–Ischemia
- Author
-
Anton Kichev, Ana A. Baburamani, Regina Vontell, Pierre Gressens, Linda Burkly, Claire Thornton, and Henrik Hagberg
- Subjects
TWEAK ,Fn14 ,perinatal brain injury ,hypoxia–ischemia ,brain ,TweakR ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a multifunctional cytokine member of the TNF family. TWEAK binds to its only known receptor, Fn14, enabling it to activate downstream signaling processes in response to tissue injury. The aim of this study was to investigate the role of TWEAK signaling in neonatal hypoxia–ischemia (HI). We found that after neonatal HI, both TWEAK and Fn14 expression were increased to a greater extent in male compared with female mice. To assess the role of TWEAK signaling after HI, the size of the injury was measured in neonatal mice genetically deficient in Fn14 and compared with their wild-type and heterozygote littermates. A significant sex difference in the Fn14 knockout (KO) animals was observed. Fn14 gene KO was beneficial in females; conversely, reducing Fn14 expression exacerbated the brain injury in male mice. Our findings indicate that the TWEAK/Fn14 pathway is critical for development of hypoxic–ischemic brain injury in immature animals. However, as the responses are different in males and females, clinical implementation depends on development of sex-specific therapies.
- Published
- 2018
- Full Text
- View/download PDF
43. Mechanical stress modulates bone remodeling signals
- Author
-
Matsui, Hiroyuki, Fukuno, Naoto, Suzuki, Osamu, Takeda, Kohsuke, Ichijo, Hidenori, Kobayashi, Takayasu, Tamura, Shinri, Sasaki, Keiichi, Sasano, Takashi, editor, and Suzuki, Osamu, editor
- Published
- 2010
- Full Text
- View/download PDF
44. Muscle health and performance in monozygotic twins with 30 years of discordant exercise habits.
- Author
-
Bathgate, Katherine E., Tobias, Irene S., Brown, Lee E., Coburn, Jared W., Arevalo, Jose A., Galpin, Andrew J., Bagley, James R., Jo, Edward, Talmadge, Robert J., and Segal, Nancy L.
- Subjects
- *
MUSCLE physiology , *TWINS , *EXERCISE physiology , *HERITABILITY , *MYOSIN , *HEALTH , *GENETICS - Abstract
Introduction: Physical health and function depend upon both genetic inheritance and environmental factors (e.g., exercise training).Purpose: To enhance the understanding of heritability/adaptability, we explored the skeletal muscle health and physiological performance of monozygotic (MZ) twins with > 30 years of chronic endurance training vs. no specific/consistent exercise.Methods: One pair of male MZ twins (age = 52 years; Trained Twin, TT; Untrained Twin, UT) underwent analyses of: (1) anthropometric characteristics and blood profiles, (2) markers of cardiovascular and pulmonary health, and (3) skeletal muscle size, strength, and power and molecular markers of muscle health.Results: This case study represents the most comprehensive physiological comparison of MZ twins with this length and magnitude of differing exercise history. TT exhibited: (1) lower body mass, body fat%, resting heart rate, blood pressure, cholesterol, triglycerides, and plasma glucose, (2) greater relative cycling power, anaerobic endurance, and aerobic capacity (VO2max), but lower muscle size/strength and poorer muscle quality, (3) more MHC I (slow-twitch) and fewer MHC IIa (fast-twitch) fibers, (4) greater AMPK protein expression, and (5) greater PAX7, IGF1Ec, IGF1Ea, and FN14 mRNA expression than UT.Conclusions: Several measured differences are the largest reported between MZ twins (TT expressed 55% more MHC I fibers, 12.4 ml/kg/min greater VO2max, and 8.6% lower body fat% vs. UT). These data collectively (a) support utilizing chronic endurance training to improve body composition and cardiovascular health and (b) suggest the cardiovascular and skeletal muscle systems exhibit greater plasticity than previously thought, further highlighting the importance of studying MZ twins with large (long-term) differences in exposomes. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
45. TWEAK/Fn14 mediates atrial‐derived HL‐1 myocytes hypertrophy via JAK2/STAT3 signalling pathway.
- Author
-
Hao, Li, Ren, Manyi, Rong, Bing, Xie, Fei, Lin, Ming‐jie, Zhao, Ya‐chao, Yue, Xin, Han, Wen‐qiang, and Zhong, Jing‐quan
- Subjects
MUSCULAR hypertrophy ,ATRIAL fibrillation ,STAT proteins ,CELLULAR signal transduction ,CARDIAC hypertrophy ,PATIENTS - Abstract
Abstract: Atrial myocyte hypertrophy is one of the most important substrates in the development of atrial fibrillation (AF). The TWEAK/Fn14 axis is a positive regulator of cardiac hypertrophy in cardiomyopathy. This study therefore investigated the effects of Fn14 on atrial hypertrophy and underlying cellular mechanisms using HL‐1 atrial myocytes. In patients with AF, Fn14 protein levels were higher in atrial myocytes from atrial appendages, and expression of TWEAK was increased in peripheral blood mononuclear cells, while TWEAK serum levels were decreased. In vitro, Fn14 expression was up‐regulated in response to TWEAK treatment in HL‐1 atrial myocytes. TWEAK increased the expression of ANP and Troponin T, and Fn14 knockdown counteracted the effect. Inhibition of JAK2, STAT3 by specific siRNA attenuated TWEAK‐induced HL‐1 atrial myocytes hypertrophy. In conclusion, TWEAK/Fn14 axis mediates HL‐1 atrial myocytes hypertrophy partly through activation of the JAK2/STAT3 pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
46. TRAF3IP2 mediates TWEAK/TWEAKR-induced pro-fibrotic responses in cultured cardiac fibroblasts and the heart.
- Author
-
Das, Nitin A., Carpenter, Andrea J., Yoshida, Tadashi, Kumar, Senthil A., Gautam, Sandeep, Mostany, Ricardo, Izadpanah, Reza, Kumar, Ashok, Mummidi, Srinivas, Siebenlist, Ulrich, and Chandrasekar, Bysani
- Subjects
- *
ADAPTOR proteins , *TUMOR necrosis factor receptors , *FIBROBLASTS , *CYTOKINE receptors , *VENTRICULAR remodeling , *THERAPEUTICS - Abstract
Persistent inflammation promotes development and progression of heart failure (HF). TWEAK (TNF-Related WEAK Inducer Of Apoptosis), a NF-κB- and/or AP-1-responsive proinflammatory cytokine that signals via TWEAK receptor (TWEAKR), is expressed at high levels in human and preclinical models of HF. Since the adapter molecule TRAF3IP2 (TRAF3 Interacting Protein 2) is an upstream regulator of various proinflammatory pathways, including those activated by NF-κB and AP-1, we hypothesized that targeting TRAF3IP2 inhibits TWEAK-induced proinflammatory and pro-fibrotic responses in vitro and in vivo. Consistent with the hypothesis, forced expression of TRAF3IP2 upregulated TWEAK and its receptor expression in cultured adult mouse cardiac fibroblasts (CF). Further, exogenous TWEAK upregulated TRAF3IP2 expression in a time- and dose-dependent manner, suggesting a positive-feedback regulation of TRAF3IP2 and TWEAK. TWEAK also promoted TRAF3IP2 nuclear translocation. Confirming its critical role in TWEAK signaling, silencing TRAF3IP2 inhibited TWEAK autoregulation, TWEAKR upregulation, p38 MAPK, NF-κB and AP-1 activation, inflammatory cytokine expression, MMP and TIMP1 activation, collagen expression and secretion, and importantly, proliferation and migration. Recapitulating these in vitro results, continuous infusion of TWEAK for 7 days increased systolic blood pressure (SBP), upregulated TRAF3IP2 expression, activated p38 MAPK, NF-κB and AP-1, induced the expression of multiple proinflammatory and pro-fibrotic mediators, and interstitial fibrosis in hearts of wild type mice. These proinflammatory and pro-fibrotic changes occurred in conjunction with myocardial hypertrophy and contractile dysfunction. Importantly, genetic ablation of TRAF3IP2 inhibited these TWEAK-induced adverse cardiac changes independent of increases in SBP, indicating that TRAF3IP2 plays a causal role, and thus a therapeutic target, in chronic inflammatory and fibro-proliferative diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
47. Overexpression of Fn14 in gliomas: tumor progression and poor prognosis.
- Author
-
Dan Tan, Feng-Mei Pang, DanLi, Longbo Zhang, JunWu, Zhao-Qian Liu, Xi Li, Han Yan, Tan, Dan, Pang, Feng-Mei, Li, Dan, Zhang, Longbo, Wu, Jun, Liu, Zhao-Qian, Li, Xi, and Yan, Han
- Abstract
Aim: To confirm whether the expression level of Fn14 could affect progression or prognosis of glioma patients.Methods: Glioma cohorts in The Cancer Genome Atlas, Gene Expression Omnibus and Chinese Glioma Genome Atlas databases were comprehensively analyzed.Results: Low-grade patients had lower expression level of Fn14, while patients with higher expression of Fn14 tended to harbor shorter overall survival and disease-free survival. The expression level of Fn14 was downregulated by IDH1/IDH2 mutations while its gene body methylation was upregulated. After adjusting age, the expression level of Fn14 was still significantly associated with overall survival and disease-free survival in low-grade gliomas. In a cell line data analysis, Fn14 expression was positively correlated with temozolomide dosage.Conclusion: Fn14 was an independent predictive biomarker for the progression and prognosis in low-grade gliomas. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
48. Differential expression of the TWEAK receptor Fn14 in IDH1 wild-type and mutant gliomas.
- Author
-
Hersh, David S., Peng, Sen, Dancy, Jimena G., Galisteo, Rebeca, Eschbacher, Jennifer M., Castellani, Rudy J., Heath, Jonathan E., Legesse, Teklu, Kim, Anthony J., Woodworth, Graeme F., Tran, Nhan L., and Winkles, Jeffrey A.
- Abstract
The TNF receptor superfamily member Fn14 is overexpressed by many solid tumor types, including glioblastoma (GBM), the most common and lethal form of adult brain cancer. GBM is notable for a highly infiltrative growth pattern and several groups have reported that high Fn14 expression levels can increase tumor cell invasiveness. We reported previously that the mesenchymal and proneural GBM transcriptomic subtypes expressed the highest and lowest levels of Fn14 mRNA, respectively. Given the recent histopathological re-classification of human gliomas by the World Health Organization based on isocitrate dehydrogenase 1 (IDH1) gene mutation status, we extended this work by comparing Fn14 gene expression in IDH1 wild-type (WT) and mutant (R132H) gliomas and in cell lines engineered to overexpress the IDH1 R132H enzyme. We found that both low-grade and high-grade (i.e., GBM) IDH1 R132H gliomas exhibit low Fn14 mRNA and protein levels compared to IDH1 WT gliomas. Forced overexpression of the IDH1 R132H protein in glioma cells reduced Fn14 expression, while treatment of IDH1 R132H-overexpressing cells with the IDH1 R132H inhibitor AGI-5198 or the DNA demethylating agent 5-aza-2′-deoxycytidine increased Fn14 expression. These results support a role for Fn14 in the more aggressive and invasive phenotype associated with IDH1 WT tumors and indicate that the low levels of Fn14 gene expression noted in IDH1 R132H mutant gliomas may be due to epigenetic regulation via changes in DNA methylation. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
49. TWEAK and RIPK1 mediate a second wave of cell death during AKI.
- Author
-
Martin-Sanchez, Diego, Fontecha-Barriuso, Miguel, Carrasco, Susana, Sanchez-Niño, Maria Dolores, von Mässenhausen, Anne, Linkermann, Andreas, Cannata-Ortiz, Pablo, Ruiz-Ortega, Marta, Egido, Jesus, Ortiz, Alberto, and Sanz, Ana Belen
- Subjects
- *
ACUTE kidney failure , *CELL death , *NECROSIS , *PROTEIN kinases , *LABORATORY mice - Abstract
Acute kidney injury (AKI) is characterized by necrotic tubular cell death and inflammation. The TWEAK/Fn14 axis is a mediator of renal injury. Diverse pathways of regulated necrosis have recently been reported to contribute to AKI, but there are ongoing discussions on the timing or molecular regulators involved. We have now explored the cell death pathways induced by TWEAK/Fn14 activation and their relevance during AKI. In cultured tubular cells, the inflammatory cytokine TWEAK induces apoptosis in a proinflammatory environment. The default inhibitor of necroptosis [necrostatin-1 (Nec-1)] was protective, while caspase inhibition switched cell death to necroptosis. Additionally, folic acid-induced AKI in mice resulted in increased expression of Fn14 and necroptosis mediators, such as receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage domainlike protein (MLKL). Targeting necroptosis with Nec-1 or by genetic RIPK3 deficiency and genetic Fn14 ablation failed to be protective at early time points (48 h). However, a persistently high cell death rate and kidney dysfunction (72-96 h) were dependent on an intact TWEAK/Fn14 axis driving necroptosis. This was prevented by Nec-1, or MLKL, or RIPK3 deficiency and by Nec-1 stable (Nec-1s) administered before or after induction of AKI. These data suggest that initial kidney damage and cell death are amplified through recruitment of inflammation-dependent necroptosis, opening a therapeutic window to treat AKI once it is established. This may be relevant for clinical AKI, since using current diagnostic criteria, severe injury had already led to loss of renal function at diagnosis. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
50. TWEAK Receptor Deficiency Has Opposite Effects on Female and Male Mice Subjected to Neonatal Hypoxia–Ischemia.
- Author
-
Kichev, Anton, Baburamani, Ana A., Vontell, Regina, Gressens, Pierre, Burkly, Linda, Thornton, Claire, and Hagberg, Henrik
- Subjects
TUMOR necrosis factors ,APOPTOSIS ,BRAIN injuries - Abstract
Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a multifunctional cytokine member of the TNF family. TWEAK binds to its only known receptor, Fn14, enabling it to activate downstream signaling processes in response to tissue injury. The aim of this study was to investigate the role of TWEAK signaling in neonatal hypoxia–ischemia (HI). We found that after neonatal HI, both TWEAK and Fn14 expression were increased to a greater extent in male compared with female mice. To assess the role of TWEAK signaling after HI, the size of the injury was measured in neonatal mice genetically deficient in Fn14 and compared with their wild-type and heterozygote littermates. A significant sex difference in the Fn14 knockout (KO) animals was observed. Fn14 gene KO was beneficial in females; conversely, reducing Fn14 expression exacerbated the brain injury in male mice. Our findings indicate that the TWEAK/Fn14 pathway is critical for development of hypoxic-ischemic brain injury in immature animals. However, as the responses are different in males and females, clinical implementation depends on development of sex-specific therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.