scholarly journals Activin signaling mediates muscle-to-adipose communication in a mitochondria dysfunction-associated obesity model

2017 ◽  
Vol 114 (32) ◽  
pp. 8596-8601 ◽  
Author(s):  
Wei Song ◽  
Edward Owusu-Ansah ◽  
Yanhui Hu ◽  
Daojun Cheng ◽  
Xiaochun Ni ◽  
...  

Mitochondrial dysfunction has been associated with obesity and metabolic disorders. However, whether mitochondrial perturbation in a single tissue influences mitochondrial function and metabolic status of another distal tissue remains largely unknown. We analyzed the nonautonomous role of muscular mitochondrial dysfunction in Drosophila. Surprisingly, impaired muscle mitochondrial function via complex I perturbation results in simultaneous mitochondrial dysfunction in the fat body (the fly adipose tissue) and subsequent triglyceride accumulation, the major characteristic of obesity. RNA-sequencing (RNA-seq) analysis, in the context of muscle mitochondrial dysfunction, revealed that target genes of the TGF-β signaling pathway were induced in the fat body. Strikingly, expression of the TGF-β family ligand, Activin-β (Actβ), was dramatically increased in the muscles by NF-κB/Relish (Rel) signaling in response to mitochondrial perturbation, and decreasing Actβ expression in mitochondrial-perturbed muscles rescued both the fat body mitochondrial dysfunction and obesity phenotypes. Thus, perturbation of muscle mitochondrial activity regulates mitochondrial function in the fat body nonautonomously via modulation of Activin signaling.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 302-302
Author(s):  
Jean-Baptiste Micol ◽  
Nicolas Duployez ◽  
Alessandro Pastore ◽  
Robert Williams ◽  
Eunhee Kim ◽  
...  

Abstract Mutations in Addition of Sex Combs Like 1 (ASXL1) are common in patients with myeloid leukemias. More recently, mutations in ASXL2, a paralog of ASXL1 with ~40% shared amino acid homology, have been discovered to occur specifically in patients with acute myeloid leukemia (AML) patients bearing the RUNX1-ETO (AML1-ETO; RUNX1-RUNX1T1) translocation and are amongst the most common mutations in RUNX1-ETO AML (mutated in 20-25% of patients). Although ASXL1 is critical for Polycomb Repressive Complex 2 function in myeloid hematopoietic cells and loss of Asxl1 recapitulates key aspects of myelodysplastic syndrome (MDS), the function of ASXL2 in normal or malignant hematopoiesis is unknown. We therefore set out to perform a functional comparison of ASXL1and ASXL2on hematopoiesis and transcription and determine the functional basis for frequent mutations in RUNX1-ETO AML. In vitro analyses of ASXL2 insertion/deletion mutations revealed that these mutations resulted in substantial reduction of ASXL2 protein expression, stability, and half-life. We therefore generated Asxl2 conditional knockout (cKO) mice to delineate the effect of ASXL2 loss on hematopoiesis. Competitive (Fig. 1A) and noncompetitive transplantation revealed that Asxl2 or compound Asxl1/2 loss resulted in cell-autonomous, rapid defects of hematopoietic stem cell function, self-renewal, and number with peripheral blood leukopenia and thrombocytopenia but without any obvious MDS features- phenotypes distinct from Asxl1 cKO mice. Mice with heterozygous deletion of Asxl2 demonstrated an intermediate phenotype between control and homozygous cKO mice indicating a gene dosage effect of Asxl2 loss. RNA sequencing (RNA-seq) of hematopoietic stem/progenitor cells from Asxl2- and Asxl1-deficient mice revealed twenty-fold greater differentially expressed genes in Asxl2 cKO mice relative to Asxl1 cKO mice. Interestingly, genes differentially expressed with Asxl2 loss significantly overlapped with direct transcriptional targets of RUNX1-ETO, findings not seen in Asxl1 cKO mice (Fig. 1B). Asxl2 target genes appeared to also be targets of RUNX1, a key gene repressed by RUNX1-ETO to promote leukemogenesis. Consistent with this, genome-wide analysis of Asxl2 binding sites through anti-Asxl2 ChIP-seq revealed that Asxl2 binding sites substantially overlap with those of Runx1. Overall, the above data suggest that Asxl2 may be a critical mediator of RUNX1-ETO mediated leukemogenesis by affecting the expression of RUNX1 and/or RUNX1-ETO target genes. RNA-seq of primary RUNX1-ETO AML patient samples revealed that ASXL2-mutant RUNX1-ETO patients form a distinct transcriptional subset of RUNX1-ETO AML (Fig. 1C) suggesting a specific role of ASXL2 in leukemogenesis. To functionally interrogate the role of ASXL2 loss in RUNX1-ETO mediated leukemogenesis we first utilized an in vitro model with RNAi-mediated depletion of ASXL1 or ASXL2 in the SKNO1 cell line (the only ASXL-wildtype human RUNX1-ETO cell line). RNA-seq revealed distinct target genes dysregulated by ASXL1 versus ASXL2 loss in these cells without any significant overlap. Anti-ASXL2, RUNX1, and RUNX1-ETO ChIPSeq in SKNO1 cells revealed significant co-occupancy of ASXL2 with RUNX1 and RUNX1-ETO binding sites. Moreover, analysis of histone modification ChIPSeq revealed an enrichment in intergenic and enhancer H3K4me1 abundance following ASXL2 loss in SKNO1 cells. Next, to understand the in vivo effects of Asxl2 loss in the context of RUNX1-ETO, we performed retroviral bone marrow (BM) transplantation assays using RUNX1-ETO9a in Asxl2 cKO mice. In contrast to the failure of hematopoietic stem cell function with Asxl2 deletion alone, mice reconstituted with BM cells expressing RUNX1-ETO9a in Asxl2-deficient background had a shortened leukemia-free survival compared to Asxl2 -wildtype control. Overall, these data reveal that ASXL2 is required for hematopoiesis and has differing biological and transcriptional functions from ASXL1. Moreover, this work identifies ASXL2 as a novel mediator of RUNX1-ETOtranscriptional function and provides a new model of penetrant RUNX1-ETO AML based on genetic events found in a substantial proportion of t(8;21) AML patients. Further interrogation of the enhancer alterations generated by ASXL2 loss in RUNX1-ETO AML may highlight new therapeutic approaches for this subset of AML. Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.


2015 ◽  
Vol 2015 ◽  
pp. 1-8 ◽  
Author(s):  
C. Simoncini ◽  
D. Orsucci ◽  
E. Caldarazzo Ienco ◽  
G. Siciliano ◽  
U. Bonuccelli ◽  
...  

Alzheimer’s disease (AD) is the most common form of dementia in the elderly. This neurodegenerative disorder is clinically characterized by impairment of cognitive functions and changes in behaviour and personality. The pathogenesis of AD is still unclear. Recent evidence supports some role of mitochondria dysfunction and oxidative stress in the development of the neurodegenerative process. In this review, we discuss the role of mitochondrial dysfunction in AD, focusing on the mechanisms that lead to mitochondrial impairment, oxidative stress, and neurodegeneration, a “vicious circle” that ends in dementia.


2017 ◽  
Vol 29 (2) ◽  
pp. 449-461 ◽  
Author(s):  
Yan Guo ◽  
Jiajia Ni ◽  
Shuang Chen ◽  
Mi Bai ◽  
Jiajuan Lin ◽  
...  

Mitochondrial dysfunction has important roles in the pathogenesis of AKI, yet therapeutic approaches to improve mitochondrial function remain limited. In this study, we investigated the pathogenic role of microRNA-709 (miR-709) in mediating mitochondrial impairment and tubular cell death in AKI. In a cisplatin-induced AKI mouse model and in biopsy samples of human AKI kidney tissue, miR-709 was significantly upregulated in the proximal tubular cells (PTCs). The expression of miR-709 in the renal PTCs of patients with AKI correlated with the severity of kidney injury. In cultured mouse PTCs, overexpression of miR-709 markedly induced mitochondrial dysfunction and cell apoptosis, and inhibition of miR-709 ameliorated cisplatin-induced mitochondrial dysfunction and cell injury. Further analyses showed that mitochondrial transcriptional factor A (TFAM) is a target gene of miR-709, and genetic restoration of TFAM attenuated mitochondrial dysfunction and cell injury induced by cisplatin or miR-709 overexpression in vitro. Moreover, antagonizing miR-709 with an miR-709 antagomir dramatically attenuated cisplatin-induced kidney injury and mitochondrial dysfunction in mice. Collectively, our results suggest that miR-709 has an important role in mediating cisplatin-induced AKI via negative regulation of TFAM and subsequent mitochondrial dysfunction. These findings reveal a pathogenic role of miR-709 in acute tubular injury and suggest a novel target for the treatment of AKI.


2020 ◽  
Author(s):  
Rwik Sen ◽  
Ezra Lencer ◽  
Elizabeth A. Geiger ◽  
Kenneth L. Jones ◽  
Tamim H. Shaikh ◽  
...  

AbstractCongenital Heart Defects (CHDs) are the most common form of birth defects, observed in 4-10/1000 live births. CHDs result in a wide range of structural and functional abnormalities of the heart which significantly affect quality of life and mortality. CHDs are often seen in patients with mutations in epigenetic regulators of gene expression, like the genes implicated in Kabuki syndrome – KMT2D and KDM6A, which play important roles in normal heart development and function. Here, we examined the role of two epigenetic histone modifying enzymes, KMT2D and KDM6A, in the expression of genes associated with early heart and neural crest cell (NCC) development. Using CRISPR/Cas9 mediated mutagenesis of kmt2d, kdm6a and kdm6al in zebrafish, we show cardiac and NCC gene expression is reduced, which correspond to affected cardiac morphology and reduced heart rates. To translate our results to a human pathophysiological context and compare transcriptomic targets of KMT2D and KDM6A across species, we performed RNA sequencing (seq) of lymphoblastoid cells from Kabuki Syndrome patients carrying mutations in KMT2D and KDM6A. We compared the human RNA-seq datasets with RNA-seq datasets obtained from mouse and zebrafish. Our comparative interspecies analysis revealed common targets of KMT2D and KDM6A, which are shared between species, and these target genes are reduced in expression in the zebrafish mutants. Taken together, our results show that KMT2D and KDM6A regulate common and unique genes across humans, mice, and zebrafish for early cardiac and overall development that can contribute to the understanding of epigenetic dysregulation in CHDs.


Author(s):  
Hannah E. Wilson ◽  
David A. Stanton ◽  
Stephanie Rellick ◽  
Werner Geldenhuys ◽  
Emidio E. Pistilli

The peroxisome-proliferator activated receptors (PPARs) have been previously implicated in the pathophysiology of skeletal muscle dysfunction in women with breast cancer (BC) and in animal models of BC. This study investigated alterations induced in skeletal muscle by BC-derived factors in an in vitro conditioned media (CM) system and tested the hypothesis that BC cells secrete a factor that represses PPAR-gamma (PPARG) expression and its transcriptional activity, leading to downregulation of PPARG target genes involved in mitochondrial function and other metabolic pathways. We found that BC-derived factors repress PPAR-mediated transcriptional activity without altering protein expression of PPARG. Further, we show that BC-derived factors induce significant alterations in skeletal muscle mitochondrial function and lipid accumulation, which are rescued with exogenous expression of PPARG. The PPARG agonist drug rosiglitazone was able to rescue BC-induced lipid accumulation, but did not rescue effects of BC-derived factors on PPAR-mediated transcription or mitochondrial function. These data suggest that BC-derived factors alter lipid accumulation and mitochondrial function via different mechanisms that are both related to PPARG signaling, with mitochondrial dysfunction likely being altered via repression of PPAR-mediated transcription, and lipid accumulation being altered via transcription-independent functions of PPARG.


2021 ◽  
Vol 8 ◽  
Author(s):  
Daniel L. Galvan ◽  
Koki Mise ◽  
Farhad R. Danesh

The role and nature of mitochondrial dysfunction in diabetic kidney disease (DKD) has been extensively studied. Yet, the molecular drivers of mitochondrial remodeling in DKD are poorly understood. Diabetic kidney cells exhibit a cascade of mitochondrial dysfunction ranging from changes in mitochondrial morphology to significant alterations in mitochondrial biogenesis, biosynthetic, bioenergetics and production of reactive oxygen species (ROS). How these changes individually or in aggregate contribute to progression of DKD remain to be fully elucidated. Nevertheless, because of the remarkable progress in our basic understanding of the role of mitochondrial biology and its dysfunction in DKD, there is great excitement on future targeted therapies based on improving mitochondrial function in DKD. This review will highlight the latest advances in understanding the nature of mitochondria dysfunction and its role in progression of DKD, and the development of mitochondrial targets that could be potentially used to prevent its progression.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4408-4408
Author(s):  
Alejandra Ortiz-Ruiz ◽  
Yanira Ruiz-Heredia ◽  
Mehmet Samur ◽  
Pedro Aguilar-Garrido ◽  
Maria Luz Morales ◽  
...  

Introduction Mitochondria controls crucial biological pathways such as proliferation, apoptosis and cell growth. However, the implication of mitochondrial activity in the pathogenesis of Multiple Myeloma (MM) still remains unknown and only a few studies connect the mitochondrial status and MM. We planned to decipher the role of the mitochondria in the MM mechanism of resistance and the potential exploitation of mitochondrial activity as a functional target in the MM therapy. Methods In order to understand the role of mitochondria in MM and its therapeutic exploitation, firstly we explored factors involved in the mitochondrial function (c-Myc, HNRNPK, TFAM, NRF1 and EF-Tu) from 770 MM patients RNAseq CoMMpass℠ data. Furthermore, we performed different studies in our MM 77 patients set: gene expression validation by RT-PCR (n=40), protein expression (COXII) by IHC (n=28); and mitochondrial activity (COX activity) by histoenzymatic-HE assay (n=11). Additionally, we analyzed the impact of bortezomib in the mitochondria regulator CD38 in 50 samples (n=30 RVD, n=20 RD regimens), at diagnosis and 6/9 months follow-up MM patients. We have tested the effect of tigecycline, a mitochondrial inhibitor, in three regimens: monotherapy, pre-treament of tigecycline (48h) with consecutive bortezomib treatment, and in combination with bortezomib in the MM cell lines JJN3, L363 and NCI-H929. To characterize the molecular mechanisms underlying the cytotoxic effect of tigecycline we analysed mitochondria load and activity (MitoTracker green and red) OXPHOS expression by WB and COX2 activity by HE assay. Finally, we followed an in vivo experiment in NSG mice (n=40) engrafted with the JJN3-GFP cell line (1x106) via tail vein and treated by 4 weeks. Analysis of the in vivo imaging and survival curve were obtained. Results The higher expression of factors involved in the mitochondrial function such as: c-Myc, HNRNPK, NRF1 and EF-Tu predict MM poor outcomes (Fig.1A). Furthermore, mitochondrial representative gene and protein expression and activity were found increased in MM relapse stage patients. We showed overexpression of C-Myc, TFAM and EF-Tu on the MM relapsed group (Fig. 1B). Moreover, IHC reveals overexpression of mitochondrial COXII protein in relapse MM patients (p-value ** < 0.001) (Fig. 1C). By functional assays we have demonstrated that gene/protein overexpression drives to an increase of activity (COX HE) in MM at relapse (p-value ***< 0.0001). (Fig. 1D). Moreover, we observed an increase of CD38 expression in patients with RVD regimen, but not without bortezomib (RD regimen) (Fig. 1E). Together these results suggest elevation of mitochondrial activity plays a role in the mechanism of resistance to treatment and/or progression of MM and the consequent relapse of the patients. In vitro studies with tigecyline and bortezomib showed cytotoxic effects in three MM cell lines (IC50 JJN3 11,91 µM; IC50 L363 10,21 µM and NCI-H929 26,37 µM, p-value *< 0.05). Moreover, bortezomib and tigecyline showed high levels of synergism (CI 0,19) (Fig. 1F). In fact, the "conditioning" treatment with tigecyline revert the resistance to bortezomib. The cells treated with tigecycline reflect diminishing in the mitochondria respiration by MitoTracker assays, decrease of COX activity and respiratory chain complexes, suggesting a reduction of mitochondrial activity (Fig. 1G). These molecular effects are exacerbated by the tigecycline and bortezomib combination. However, bortezomib monotherapy not decrease or inclusive, increase, all the molecular mechanisms of mitochondria studied. Finally, mice groups treated with tigecycline alone or in combination with bortezomib reported a better survival and lower JJN3-GFP infiltration (p-value *< 0.05) (Fig. 1H). Conclusion To sum up, these findings highlight new vulnerabilities in myeloma cells, suggesting a potential therapeutic target in the treatment of the disease. The metabolic activation of myeloma cells with the collaboration of CD38 and/or c-Myc overexpression or his regulators (e.g. HNRNPK) in response to bortezomib treatment lead an increase of mitochondria respiration. These data confirm the important role of mitochondria in the loss of efficacy in inhibitors of proteasome treatment. Thus, mitochondrial respiration emerges as a novel target in bortezomib relapsed MM patients, and, potentially, in multiple c-Myc, HNRNPK and CD38 overexpression neoplasms. Disclosures Munshi: Adaptive: Consultancy; Oncopep: Consultancy; Janssen: Consultancy; Takeda: Consultancy; Amgen: Consultancy; Celgene: Consultancy; Abbvie: Consultancy.


2019 ◽  
Vol 2 (2) ◽  
pp. e201900392 ◽  
Author(s):  
Sophia von Stockum ◽  
Alvaro Sanchez-Martinez ◽  
Samantha Corrà ◽  
Joy Chakraborty ◽  
Elena Marchesan ◽  
...  

Aberrant mitochondrial dynamics disrupts mitochondrial function and contributes to disease conditions. A targeted RNA interference screen for deubiquitinating enzymes (DUBs) affecting protein levels of multifunctional mitochondrial fusion protein Mitofusin (MFN) identified USP8 prominently influencing MFN levels. Genetic and pharmacological inhibition of USP8 normalized the elevated MFN protein levels observed in PINK1 and Parkin-deficient models. This correlated with improved mitochondrial function, locomotor performance and life span, and prevented dopaminergic neurons loss in Drosophila PINK1 KO flies. We identified a novel target antagonizing pathologically elevated MFN levels, mitochondrial dysfunction, and dopaminergic neuron loss of a Drosophila model of mitochondrial dysfunction.


2021 ◽  
Author(s):  
Jia Song ◽  
Lingshu Wang ◽  
Xinghong Guo ◽  
Qin He ◽  
Chen Cui ◽  
...  

Abstract Background: Dysregulation of α-cells results in hyperglycemia and hyperglucagonemia in type 2 diabetes mellitus (T2DM). Mesenchymal stem cell (MSC)-based therapy increases oxygen consumption of islets and enhances insulin secretion. However, the underlying mechanism for the protective role of MSCs in α- cell mitochondrial dysfunction remains unclear. Here, we evaluated the efficacy and molecular mechanisms of human umbilical cord MSCs (hucMSCs) on α-cell mitochondrial function and glucagon secretion in T2DM.Methods: hucMSCs were used to treat two kinds of T2DM mice and αTC1-6 cells to explore the role of hucMSCs in improving α-cell mitochondrial dysfunction and hyperglucagonemia. Plasma and supernatant glucagon were detected by enzyme-linked immunosorbent assay (ELISA). Mitochondrial function of α-cells was assessed by the Seahorse Analyzer. To investigate the underlying mechanisms, Sirtuin 1 (SIRT1), Forkhead box O3a (FoxO3a), glucose transporter type1 (GLUT1), and glucokinase (GCK) were assessed by western blotting analysis.Results: In vivo, hucMSC infusion improved glucose and insulin tolerance, as well as hyperglycemia and hyperglucagonemia in T2DM mice. Meanwhile, hucMSC intervention rescued islet structure and decreased α- to β-cell ratio. Consistently, glucagon secretion from αTC1-6 cells was inhibited by hucMSCs in vitro. Meanwhile, hucMSC treatment activated intracellular SIRT1/FoxO3a signaling, promoted glucose uptake and activation, alleviated mitochondrial dysfunction, and enhanced ATP production. However, transfection of SIRT1 small interfering RNA (siRNA) or the application of SIRT1 inhibitor EX-527 weakened the therapeutic effects of hucMSCs on mitochondrial function and glucagon secretion.Conclusions: Our observations indicate that hucMSCs mitigate mitochondrial dysfunction and glucagon hypersecretion of α-cells in T2DM via SIRT1/FoxO3a signaling, which provides novel evidence demonstrating the potential for hucMSCs in treating T2DM.


2016 ◽  
Vol 2016 ◽  
pp. 1-11 ◽  
Author(s):  
Le Li ◽  
Creed M. Stary

Astrocytes and microglia play crucial roles in the response to cerebral ischemia and are effective targets for stroke therapy in animal models. MicroRNAs (miRs) are important posttranscriptional regulators of gene expression that function by inhibiting the translation of select target genes. In astrocytes, miR expression patterns regulate mitochondrial function in response to oxidative stressviatargeting of Bcl2 and heat shock protein 70 family members. Mitochondria play an active role in microglial activation, and miRs regulate the microglial neuroinflammatory response. As endogenous miR expression patterns can be altered with exogenous mimics and inhibitors, miR-targeted therapies represent a viable intervention to optimize glial mitochondrial function and improve clinical outcome following cerebral ischemia. In the present article, we review the role that astrocytes and microglia play in neuronal function and fate following ischemic stress, discuss the relevance of mitochondria in the glial response to injury, and present current evidence implicating miRs as critical regulators in the glial mitochondrial response to cerebral ischemia.


Sign in / Sign up

Export Citation Format

Share Document