173 results on '"Shihuan Kuang"'
Search Results
2. Peroxisome proliferator‐activated receptor γ coactivator 1‐α overexpression improves angiogenic signalling potential of skeletal muscle‐derived extracellular vesicles
- Author
-
Chris K. Kargl, Brian P. Sullivan, Derek Middleton, Andrew York, Lundon C. Burton, Jeffrey J. Brault, Shihuan Kuang, and Timothy P. Gavin
- Subjects
Nutrition and Dietetics ,Physiology ,Physiology (medical) ,General Medicine - Abstract
What is the central question of this study? Skeletal muscle extracellular vesicles likely act as pro-angiogenic signalling factors: does overexpression of peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) alter skeletal muscle myotube extracellular vesicle release, contents and angiogenic potential? What is the main finding and its importance? Overexpression of PGC-1α results in secretion of extracellular vesicles that elevate measures of angiogenesis and protect against acute oxidative stress in vitro. Skeletal muscle with high levels of PGC-1α expression, commonly associated with exercise induced angiogenesis and high basal capillarization, may secrete extracellular vesicles that support capillary growth and maintenance.Skeletal muscle capillarization is proportional to muscle fibre mitochondrial content and oxidative capacity. Skeletal muscle cells secrete many factors that regulate neighbouring capillary endothelial cells (ECs), including extracellular vesicles (SkM-EVs). Peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) regulates mitochondrial biogenesis and the oxidative phenotype in skeletal muscle. Skeletal muscle PGC-1α also regulates secretion of multiple angiogenic factors, but it is unknown whether PGC-1α regulates SkM-EV release, contents and angiogenic signalling potential. PGC-1α was overexpressed via adenovirus in primary human myotubes. EVs were collected from PGC-1α-overexpressing myotubes (PGC-EVs) as well as from green fluorescent protein-overexpressing myotubes (GFP-EVs), and from untreated myotubes. EV release and select mRNA contents were measured from EVs. Additionally, ECs were treated with EVs to measure angiogenic potential of EVs in normal conditions and following an oxidative stress challenge. PGC-1α overexpression did not impact EV release but did elevate EV content of mRNAs for several antioxidant proteins (nuclear factor erythroid 2-related factor 2, superoxide dismutase 2, glutathione peroxidase). PGC-EV treatment of cultured human umbilical vein endothelial cells (HUVECs) increased their proliferation (+36.6%), tube formation (length: +28.1%; number: +25.7%) and cellular viability (+52.9%), and reduced reactive oxygen species levels (-41%) compared to GFP-EVs. Additionally, PGC-EV treatment protected against tube formation impairments and induction of cellular senescence following acute oxidative stress. Overexpression of PGC-1α in human myotubes increases the angiogenic potential of SkM-EVs. These angiogenic benefits coincided with increased anti-oxidative capacity of recipient HUVECs. High PGC-1α expression in skeletal muscle may prompt the release of SkM-EVs that support vascular redox homeostasis and angiogenesis.
- Published
- 2022
- Full Text
- View/download PDF
3. Effects of heat therapy on skeletal muscle interstitial oxygenation and exercise tolerance in HFpEF rats
- Author
-
Michael Belbis, Bohyun Ro, Luke Schepers, Kun Kim, Kyoungrae Kim, Terence Ryan, Craig Goergen, Shihuan Kuang, Timothy Gavin, Bruno Roseguini, and Daniel Hirai
- Subjects
Physiology - Abstract
Heart failure with preserved ejection fraction (HFpEF) impairs skeletal muscle microvascular function and blood flow thereby reducing exercise tolerance. Limited therapies currently exist to restore physical capacity in HFpEF. Heat therapy has been shown to improve cardiovascular outcomes and exercise tolerance in health and other diseases. PURPOSE: To test the hypothesis that chronic whole-body heat therapy would improve heart function and morphology, body composition, skeletal muscle interstitial oxygenation (PO2) and, therefore, exercise tolerance in a rodent model of HFpEF. METHODS: Male obese ZSF1 rats (5-6 mo) underwent either 8 weeks (6 days/wk) of heat therapy (HT; 39°C; n=10) or control (CON; 22°C; n=8) interventions. Exercise tolerance (treadmill tests to exhaustion), body composition (echoMRI) and heart function and morphology (echocardiography) were assessed pre- and post-intervention. Spinotrapezius PO2 (phosphorescence quenching) was assessed post-intervention from rest to submaximal contractions (1 Hz, 5-7 V, 3 min). RESULTS: Repeated HT prevented the decline in the main outcome of exercise tolerance (PRE:424±17, POST:425±13 s; p>0.05) observed in CON (PRE:444±24, POST:319±24 s; p0.05). There were no differences in isovolumetric relaxation time after treatment (CON PRE:23±2, POST:24±1; HT PRE:21±1, POST:24±2 ms; p>0.05). HT averted the decline in left ventricular ejection fraction (PRE:71.3±1.7, POST:73.7±2.3 %; p>0.05) observed in CON (PRE:75.3±0.9, POST:67.9±4.0 %; p0.05), it was reduced after HT (PRE:0.38±0.01; POST:0.35±0.01 g/mg; p0.05) but was increased after HT (PRE:0.56±0.01, POST:0.60±0.01 g/mg; p ACSM Doctoral Student Research Grant and Purdue University. This is the full abstract presented at the American Physiology Summit 2023 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure 4 from Plk1 Phosphorylation of Orc2 and Hbo1 Contributes to Gemcitabine Resistance in Pancreatic Cancer
- Author
-
Xiaoqi Liu, Elena G. Chiorean, Tony Hazbun, Timothy L. Ratliff, Stephen F. Konieczny, Bennett D. Elzey, Shihuan Kuang, Steven J. Rice, X. Shawn Liu, and Bing Song
- Abstract
PDF file - 30k, knock-down of cFos and MDR1 by RNAi in gemcitabine-resistant cells
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 3 from Plk1 Phosphorylation of Orc2 and Hbo1 Contributes to Gemcitabine Resistance in Pancreatic Cancer
- Author
-
Xiaoqi Liu, Elena G. Chiorean, Tony Hazbun, Timothy L. Ratliff, Stephen F. Konieczny, Bennett D. Elzey, Shihuan Kuang, Steven J. Rice, X. Shawn Liu, and Bing Song
- Abstract
PDF file - 67K, Orc2-A and Hbo1-A mutant cells are sensitive to gemcitabine treatment
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Plk1 Phosphorylation of Orc2 and Hbo1 Contributes to Gemcitabine Resistance in Pancreatic Cancer
- Author
-
Xiaoqi Liu, Elena G. Chiorean, Tony Hazbun, Timothy L. Ratliff, Stephen F. Konieczny, Bennett D. Elzey, Shihuan Kuang, Steven J. Rice, X. Shawn Liu, and Bing Song
- Abstract
Although gemcitabine is the standard chemotherapeutic drug for treatment of pancreatic cancer, almost all patients eventually develop resistance to this agent. Previous studies identified Polo-like kinase 1 (Plk1) as the mediator of gemcitabine resistance, but the molecular mechanism remains unknown. In this study, we show that Plk1 phosphorylation of Orc2 and Hbo1 mediates the resistance to gemcitabine. We show that the level of Plk1 expression positively correlates with gemcitabine resistance, both in pancreatic cancer cells and xenograft tumors. Overexpression of Plk1 increases gemcitabine resistance, while inhibition of Plk1 sensitizes pancreatic cancer cells to gemcitabine treatment. To validate our findings, we show that inhibition of Plk1 sensitizes tumors to gemcitabine treatment in a mouse xenograft study. Mechanistically, we find that Plk1 phosphorylation of Orc2 maintains DNA replication on gemcitabine treatment. Furthermore, Plk1 phosphorylation of Hbo1 transcriptionally increases cFos expression and consequently elevates its target multidrug resistance 1 (MDR1), which was previously reported to confer chemotherapeutic drug resistance. Knockdown of cFos or MDR1 sensitizes gemcitabine-resistant cells to gemcitabine treatment. Finally, pancreatic cancer cells expressing Plk1-unphosphorylatable mutants of Orc2 or Hbo1 are more sensitive to gemcitabine than cells expressing wild-type Orc2 or Hbo1. In short, our study provides a mechanism for Plk1-mediated gemcitabine resistance, suggesting that Plk1 is a promising target for treatment of gemcitabine-resistant pancreatic cancer. Mol Cancer Ther; 12(1); 58–68. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 2 from Plk1 Phosphorylation of Orc2 and Hbo1 Contributes to Gemcitabine Resistance in Pancreatic Cancer
- Author
-
Xiaoqi Liu, Elena G. Chiorean, Tony Hazbun, Timothy L. Ratliff, Stephen F. Konieczny, Bennett D. Elzey, Shihuan Kuang, Steven J. Rice, X. Shawn Liu, and Bing Song
- Abstract
PDF file - 102K, Gemcitabine-resistant tumors are sensitive to inhibition of Plk1
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Table 1 from Plk1 Phosphorylation of Orc2 and Hbo1 Contributes to Gemcitabine Resistance in Pancreatic Cancer
- Author
-
Xiaoqi Liu, Elena G. Chiorean, Tony Hazbun, Timothy L. Ratliff, Stephen F. Konieczny, Bennett D. Elzey, Shihuan Kuang, Steven J. Rice, X. Shawn Liu, and Bing Song
- Abstract
PDF file - 17K, The IC50 values of Gemcitabine in Pancreatic cell lines
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 1 from Plk1 Phosphorylation of Orc2 and Hbo1 Contributes to Gemcitabine Resistance in Pancreatic Cancer
- Author
-
Xiaoqi Liu, Elena G. Chiorean, Tony Hazbun, Timothy L. Ratliff, Stephen F. Konieczny, Bennett D. Elzey, Shihuan Kuang, Steven J. Rice, X. Shawn Liu, and Bing Song
- Abstract
PDF file - 44K, Inhibition of Plk1 sensitizes cells to gemcitabine
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Methods and Figure Legends from Plk1 Phosphorylation of Orc2 and Hbo1 Contributes to Gemcitabine Resistance in Pancreatic Cancer
- Author
-
Xiaoqi Liu, Elena G. Chiorean, Tony Hazbun, Timothy L. Ratliff, Stephen F. Konieczny, Bennett D. Elzey, Shihuan Kuang, Steven J. Rice, X. Shawn Liu, and Bing Song
- Abstract
PDF file - 88K
- Published
- 2023
- Full Text
- View/download PDF
11. Obesity and exercise training alter inflammatory pathway skeletal muscle small extracellular vesicle microRNAs
- Author
-
Brian P. Sullivan, Yaohui Nie, Sheelagh Evans, Chris K. Kargl, Zach R. Hettinger, Ron T. Garner, Monica J. Hubal, Shihuan Kuang, Julianne Stout, and Timothy P. Gavin
- Subjects
Inflammation ,Nutrition and Dietetics ,Physiology ,Interleukin-8 ,General Medicine ,Extracellular Vesicles ,MicroRNAs ,Phosphatidylinositol 3-Kinases ,Physiology (medical) ,Humans ,Obesity ,RNA, Messenger ,Muscle, Skeletal ,Exercise - Abstract
What is the central question of this study? Is 1 week of exercise training sufficient to reduce local and systemic inflammation? Do obesity and short-term concurrent aerobic and resistance exercise training alter skeletal muscle extracellular vesicle (EV) contents? What is the main finding and its importance? Obesity alters skeletal muscle small EV microRNAs targeting inflammatory and growth pathways. Exercise training alters skeletal muscle small EV microRNAs targeting inflammatory pathways, indicative of reduced inflammation. Our findings provide support for the hypotheses that EVs play a vital role in intercellular communication during health and disease and that EVs mediate many of the beneficial effects of exercise.Obesity is associated with chronic inflammation characterized by increased levels of inflammatory cytokines, whereas exercise training reduces inflammation. Small extracellular vesicles (EVs; 30-150 nm) participate in cell-to-cell communication in part through microRNA (miRNA) post-transcriptional regulation of mRNA. We examined whether obesity and concurrent aerobic and resistance exercise training alter skeletal muscle EV miRNA content and inflammatory signalling. Vastus lateralis biopsies were obtained from sedentary individuals with (OB) and without obesity (LN). Before and after 7 days of concurrent aerobic and resistance training, muscle-derived small EV miRNAs and whole-muscle mRNAs were measured. Pathway analysis revealed that obesity alters small EV miRNAs that target inflammatory (SERPINF1, death receptor and Gα
- Published
- 2022
- Full Text
- View/download PDF
12. Author Reply to Peer Reviews of PRMT5 links lipid metabolism to contractile function of skeletal muscles
- Author
-
Kun Ho Kim, Zhihao Jia, Madigan Mckenna Strange, Jingjuan Chen, Jiamin Qiu, Stephanie Nicole Oprescu, Xiyue Chen, Sabriya A Syed, Feng Yue, Bruno Testini Roseguini, Anthony N Imbalzano, Changdeng Hu, and Shihuan Kuang
- Published
- 2023
- Full Text
- View/download PDF
13. Author Reply to Peer Reviews of Atypical peripheral actin band formation via overactivation of RhoA and Non-muscle myosin II in Mitofusin 2 deficient cells
- Author
-
Yueyang Wang, Lee D. Troughton, Fan Xu, Aritra Chatterjee, Han Zhao, Laura P Cifuentes, Ryan B Wagner, Jingjuan Chen, Linlin Li, David M Umulis, Shihuan Kuang, Daniel Suter, Chongli Yuan, Deva Chan, Fang Huang, Patrick W Oakes, and Qing Deng
- Published
- 2023
- Full Text
- View/download PDF
14. Targeted Lipidomics Analysis of Adipose and Skeletal Muscle Tissues by Multiple Reaction Monitoring Profiling
- Author
-
Xiyue Chen, Christina R. Ferreira, and Shihuan Kuang
- Published
- 2023
- Full Text
- View/download PDF
15. Prenatal Serotonin Fluctuation Affects Serotoninergic Development and Related Neural Circuits in Chicken Embryos
- Author
-
Heng-Wei Cheng, Xiaohong Huang, Todd J. Applegate, Shihuan Kuang, and Tsang-long Lin
- Subjects
Hypothalamo-Hypophyseal System ,Serotonin ,Fetus ,General Neuroscience ,Receptor expression ,Neurogenesis ,Dopaminergic ,Pituitary-Adrenal System ,Chick Embryo ,Biology ,Serotonergic ,Fetal Development ,Ventral tegmental area ,medicine.anatomical_structure ,Dopamine ,medicine ,Animals ,Humans ,Female ,Chickens ,Neural development ,Neuroscience ,medicine.drug - Abstract
The placenta is the primary source of serotonin (5-HT) for fetal development, programming fetal neural wiring in humans and other mammals. The fluctuation in maternal 5-HT affects fetal neurogenesis with life-long consequences, however, its mechanisms have not been well known. The chicken embryo, independent of maternal neurohormonal influence, may offer an ideal model for studying the mechanisms of prenatal 5-HT exposure altering postnatal physiological homeostasis and behavioral exhibition. To investigate the fine-tuning of 5-HT to the early embryonic neurodevelopment, 10 µg and 20 µg 5-HT were secretively injected to chicken embryos before incubation. 5-HT exposure mainly affected the neural development in the pons and midbrain, altered the serotoninergic and dopaminergic (DAergic) neuronal morphology, nucleus distribution, and their metabolisms and related gene expressions. The comprehensive effect of 5-HT exposure was not dosage-dependent but the working pathways differed, 10 µg 5-HT exposure reduced 5-HT turnover rate, increased 5-HT 1a receptor expression, and facilitated the ventral tegmental area neuronal development; while 20 µg 5-HT exposure increased the serotoninergic and DAergic neurotransmission and enhanced serotoninergic regulation to the hypothalamus. These findings indicate that the 5-HT exposure effect can be achieved via different paths by modifying the embryonic serotonergic (5-HTergic) and DAergic systems and altering fetal 5-HTergic influence on the thalamocortical circuit and hypothalamic–pituitary–adrenal axis. These results may offer a novel sight for understanding the function of 5-HT during neurodevelopment and raise the possibility for using selective 5-HT reuptake inhibitors to regulate emotional and mental wellness during early pregnancy and possible risks of complications for babies.
- Published
- 2021
- Full Text
- View/download PDF
16. PRMT5 links lipid metabolism to contractile function of skeletal muscles
- Author
-
Kun Ho Kim, Zhihao Jia, Madigan M. Snyder, Jingjuan Chen, Jiamin Qiu, Stephanie N. Oprescu, Xiyue Chen, Sabriya A Syed, Feng Yue, Bruno T. Roseguini, Anthony N. Imbalzano, Changdeng Hu, and Shihuan Kuang
- Abstract
The skeletal muscle plays a key role in systemic energy homeostasis besides its canonical contractile function, but what couples these functions is poorly defined. Protein Arginine MethylTransferase 5 (PRMT5) is a well-known oncoprotein but also expressed in healthy tissues with unclear physiological functions. As adult muscles express high levels ofPrmt5, we generated myocyte-specificPrmt5knockout (Prmt5MKO) mice. We observed reduced muscle mass, oxidative capacity, force production and exercise performance inPrmt5MKOmice. The motor deficiency is associated with scarce lipid droplets in myofibers due to defects in lipid biosynthesis and degradation. First,Prmt5MKOreduced demethylation and stability of Sterol Regulatory Element-Binding Transcription Factor 1a (SREBP1a), a master regulator ofde novolipogenesis. Second,Prmt5MKOimpaired the repressive H4R3Me2s (histone H4 arginine-3 symmetric demethylation) at thePnpla2gene, elevating the level of its encoded protein ATGL, the rate-limiting enzyme catalyzing lipolysis. Accordingly, myocyte-specific double knockout ofPnpla2andPrmt5normalized muscle mass and function. Together, our findings delineate a physiological function of PRMT5 in linking lipid metabolism to contractile function of myofibers.
- Published
- 2022
- Full Text
- View/download PDF
17. Atypical peripheral actin band formation via overactivation of RhoA and Non-muscle myosin II in Mitofusin 2 deficient cells
- Author
-
Yueyang Wang, Lee D. Troughton, Fan Xu, Aritra Chatterjee, Han Zhao, Laura P. Cifuentes, Ryan B. Wagner, Jingjuan Chen, Shihuan Kuang, Daniel M. Suter, Chongli Yuan, Deva Chan, Fang Huang, Patrick W. Oakes, and Qing Deng
- Abstract
Cell spreading and migration play central roles in many physiological and pathophysiological processes. We have previously shown that MFN2 regulates the migration of human neutrophillike cells via suppressing Rac activation. Here, we show that in mouse embryonic fibroblasts, MFN2 suppresses RhoA activation and supports cell polarization. After the initial spreading period, the wild-type cells polarize and migrate, whereas the Mfn2-/- cells maintain a circular shape. Increased cytosolic Ca2+ resulting from the loss of Mfn2 is directly responsible for this phenotype, which can be rescued by expressing an artificial tether to bring mitochondria and ER to close vicinity. Elevated cytosolic Ca2+ activates Ca2+/calmodulin-dependent protein kinase II, RhoA, and Myosin light-chain kinase, causing an over-activation of non-muscle myosin II and a formation of a prominent F-actin ring at the cell periphery and increased cell contractility. The formation of the peripheral actin band alters cell physics and is dependent on substrate rigidity.Our results provide a novel molecular basis to understand how MFN2 regulates distinct signaling pathways in different cells and tissue environments, which is instrumental in understanding and treating MFN2-related diseases.
- Published
- 2022
- Full Text
- View/download PDF
18. Extracellular vesicles released from stress‐induced prematurely senescent myoblasts impair endothelial function and proliferation
- Author
-
Timothy P. Gavin, Christopher K. Kargl, Shihuan Kuang, Zachary R. Hettinger, and Jonathan H. Shannahan
- Subjects
Tube formation ,Senescence ,Nutrition and Dietetics ,Physiology ,Chemistry ,Angiogenesis ,Myoblasts, Skeletal ,Skeletal muscle ,General Medicine ,Cell biology ,Endothelial stem cell ,Extracellular Vesicles ,medicine.anatomical_structure ,Physiology (medical) ,Human Umbilical Vein Endothelial Cells ,medicine ,Humans ,Myocyte ,Progenitor cell ,Cellular Senescence ,Cell Proliferation ,Transforming growth factor - Abstract
NEW FINDINGS What is the central question of this study? What is the impact of stress-induced premature senescence on skeletal muscle myoblast-derived extracellular vesicles (EVs) and myoblast-endothelial cell crosstalk? What is the main finding and its importance? Hydrogen peroxide treatment of human myoblasts induced stress-induced premature senescence (SIPS) and increased the release of exosome-sized EVs (30-150 nm in size) five-fold compared to untreated controls. Treatment of SIPS myoblast-derived EVs on endothelial cells increased senescence markers and decreased proliferation. Gene expression analysis of SIPS myoblast-derived EVs revealed a four-fold increase in senescence factor transforming growth factor-β. These results highlight potential mechanisms by which senescence imparts deleterious effects on the cellular microenvironment. ABSTRACT Cellular senescence contributes to numerous diseases through the release of pro-inflammatory factors as part of the senescence-associated secretory phenotype (SASP). In skeletal muscle, resident muscle progenitor cells (satellite cells) express markers of senescence with advancing age and in response to various pathologies, which contributes to reduced regenerative capacities in vitro. Satellite cells regulate their microenvironment in part through the release of extracellular vesicles (EVs), but the effect of senescence on EV signaling is unknown. Primary human myoblasts were isolated following biopsies of the vastus lateralis from young healthy subjects. Hydrogen peroxide (H2 O2 ) treatment was used to achieve stress-induced premature senescence (SIPS) of myoblasts. EVs secreted by myoblasts with and without H2 O2 treatment were isolated, analysed and used to treat human umbilical vein endothelial cells (HUVECs) to assess senescence and angiogenic impact. H2 O2 treatment of primary human myoblasts in vitro increased markers of senescence (β-galactosidase and p21Cip1 ), decreased proliferation and increased exosome-like EV (30-150 nm) release approximately five-fold. In HUVECs, EV treatment from H2 O2 -treated myoblasts increased markers of senescence (β-galactosidase and transforming growth factor β), decreased proliferation and impaired HUVEC tube formation. Analysis of H2 O2 -treated myoblast-derived EV mRNA revealed a nearly four-fold increase in transforming growth factor β expression. Our novel results highlight the impact of SIPS on myoblast communication and identify a VasoMyo Crosstalk by which SIPS myoblast-derived EVs impair endothelial cell function in vitro.
- Published
- 2021
- Full Text
- View/download PDF
19. Labeling and analyzing lipid droplets in mouse muscle stem cells
- Author
-
Jingjuan Chen, Feng Yue, and Shihuan Kuang
- Subjects
Mice ,General Immunology and Microbiology ,General Neuroscience ,Stem Cells ,Muscle Fibers, Skeletal ,Animals ,Lipid Droplets ,Muscle, Skeletal ,General Biochemistry, Genetics and Molecular Biology ,Fluorescence - Abstract
Lipid droplets are emerging as an important and dynamic organelle whose metabolism controls stem cell behavior. Here we present a comprehensive protocol to visualize and quantify these organelles in mouse muscle satellite cells (MuSCs). This protocol includes steps for BODIPY/LipidSpot610 staining of freshly isolated MuSCs, in vitro cultured myoblasts, and single myofibers to label lipid droplets and subsequent analysis and quantification of fluorescence signals. This protocol can be modified to stain lipid droplets in other cell types of interest. For complete details on the use and execution of this protocol, please refer to Yue et al. (2022).
- Published
- 2022
20. Biomimetic glycosaminoglycan-based scaffolds improve skeletal muscle regeneration in a Murine volumetric muscle loss model
- Author
-
Liangju Kuang, Zhihao Jia, Kun Ho Kim, Meng Deng, Gabrielle Shafer, Paul Lengemann, Naagarajan Narayanan, Victor Bernal-Crespo, and Shihuan Kuang
- Subjects
Myoblast proliferation ,MHC, Myosin heavy chain ,AChR, Acetyl choline receptors ,Chondroitin sulfate ,VML, Volumetric muscle loss ,Hyaluronic acid ,0206 medical engineering ,Biomedical Engineering ,02 engineering and technology ,MyoD ,Article ,EDC, 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide ,Biomaterials ,Myoblasts ,chemistry.chemical_compound ,ECM, Extracellular matrix ,medicine ,lcsh:TA401-492 ,Myocyte ,PEGDA, Poly(ethylene glycol) diacrylate ,Volumetric muscle loss ,lcsh:QH301-705.5 ,CS, Chondroitin Sulfate ,Regeneration (biology) ,GAG, Glycosaminoglycan ,Skeletal muscle ,Hydrogels ,eMHC, embryonic myosin heavy chain ,021001 nanoscience & nanotechnology ,NHS, N-hydroxysuccinimide ,020601 biomedical engineering ,HA, Hyaluronic acid ,MES, 2-(N-morpholino) ethanesulfonic acid ,Cell biology ,medicine.anatomical_structure ,chemistry ,lcsh:Biology (General) ,Skeletal muscle tissue engineering ,Self-healing hydrogels ,lcsh:Materials of engineering and construction. Mechanics of materials ,0210 nano-technology ,C2C12 ,Biotechnology - Abstract
Volumetric muscle loss (VML) injuries characterized by critical loss of skeletal muscle tissues result in severe functional impairment. Current treatments involving use of muscle grafts are limited by tissue availability and donor site morbidity. In this study, we designed and synthesized an implantable glycosaminoglycan-based hydrogel system consisting of thiolated hyaluronic acid (HA) and thiolated chondroitin sulfate (CS) cross-linked with poly(ethylene glycol) diacrylate to promote skeletal muscle regeneration of VML injuries in mice. The HA-CS hydrogels were optimized with suitable biophysical properties by fine-tuning degree of thiol group substitution to support C2C12 myoblast proliferation, myogenic differentiation and expression of myogenic markers MyoD, MyoG and MYH8. Furthermore, in vivo studies using a murine quadriceps VML model demonstrated that the HA-CS hydrogels supported integration of implants with the surrounding host tissue and facilitated migration of Pax7+ satellite cells, de novo myofiber formation, angiogenesis, and innervation with minimized scar tissue formation during 4-week implantation. The hydrogel-treated and autograft-treated mice showed similar functional improvements in treadmill performance as early as 1-week post-implantation compared to the untreated groups. Taken together, our results demonstrate the promise of HA-CS hydrogels as regenerative engineering matrices to accelerate healing of skeletal muscle injuries., Graphical abstract Image 1, Highlights • Synthesized a biomimetic HA-CS hydrogel system with properties tailored for skeletal muscle. • HA-CS hydrogels supported in vitro myoblast cell behavior. • Implanted cell-free HA-CS hydrogels promoted de novo skeletal muscle regeneration. • HA-CS hydrogels enhanced in vivo angiogenesis and innervation. • HA-CS hydrogels promoted functional recovery comparable to autograft transplants.
- Published
- 2021
21. Imaging and Analysis of Isomeric Unsaturated Lipids through Online Photochemical Derivatization of Carbon–Carbon Double Bonds**
- Author
-
Xiaofei Sun, Yingju Li, Pei Su, Shihuan Kuang, Sudhansu K. Dey, Hang Hu, Jiamin Qiu, Julia Laskin, and Daisy Unsihuay
- Subjects
Spectrometry, Mass, Electrospray Ionization ,Double bond ,Electrospray ionization ,010402 general chemistry ,Photochemistry ,Mass spectrometry ,01 natural sciences ,Article ,Catalysis ,chemistry.chemical_compound ,Isomerism ,Lipid oxidation ,Lipidomics ,Derivatization ,chemistry.chemical_classification ,Desorption electrospray ionization ,Photosensitizing Agents ,Singlet Oxygen ,010405 organic chemistry ,Chemistry ,Hydrogen Peroxide ,General Chemistry ,General Medicine ,Lipidome ,Lipids ,Carbon ,0104 chemical sciences ,Isotope Labeling ,lipids (amino acids, peptides, and proteins) - Abstract
Unraveling the complexity of the lipidome requires the development of novel approaches for the structural characterization of lipid species with isomer-level discrimination. Herein, we introduce an online photochemical approach for lipid isomer identification through selective derivatization of double bonds by reaction with singlet oxygen. Lipid hydroperoxide products are generated promptly after laser irradiation. Fragmentation of these species in a mass spectrometer produces diagnostic fragments, which reveal the C=C locations in the unreacted lipids. This approach uses an inexpensive light source and photosensitizer making it easy to incorporate into any lipidomics workflow. We demonstrate the utility of this approach for the shotgun profiling of C=C locations in different lipid classes present in tissue extracts using electrospray ionization (ESI) and ambient imaging of lipid species differing only by the location of C=C bonds using nanospray desorption electrospray ionization (nano-DESI).
- Published
- 2021
- Full Text
- View/download PDF
22. PRMT5 regulates lipid metabolism in skeletal muscle via epigenetic suppression of Pnpla2
- Author
-
Kun Ho Kim, Feng Yue, Zhihao Jia, Jingjuan Chen, Jiamin Qiu, Madigan M. Strange, Xiyue Chen, Bruno Tesini Roseguini, Changdeng Hu, and Shihuan Kuang
- Subjects
Genetics ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2022
- Full Text
- View/download PDF
23. Transcriptome and Mass‐Spectrometry‐Based Lipidome Reveal a Role of PRMT5 in Membrane Transport and Cholesterol Synthesis in White Adipocytes
- Author
-
Xiyue Chen, Zhihao Jia, Shuyi Fang, Feng Yue, Christina R. Ferreria, Jun Wan, and Shihuan Kuang
- Subjects
Genetics ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2022
- Full Text
- View/download PDF
24. Impact of Heat Therapy on Skeletal Muscle Mass and Function in A Mouse Model of Duchenne Muscular Dystrophy
- Author
-
Bohyun Ro, Abigail Deren, Qifan Song, Kun Ho Kim, Shihuan Kuang, and Bruno Roseguini
- Subjects
Genetics ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2022
- Full Text
- View/download PDF
25. Polymeric nanoparticles functionalized with muscle-homing peptides for targeted delivery of phosphatase and tensin homolog inhibitor to skeletal muscle
- Author
-
Shihuan Kuang, Meng Deng, Feng Yue, Jiamin Qiu, and Di Huang
- Subjects
Duchenne muscular dystrophy ,0206 medical engineering ,Phosphatase ,Biomedical Engineering ,02 engineering and technology ,Biochemistry ,Article ,Polyethylene Glycols ,Biomaterials ,Mice ,Drug Delivery Systems ,In vivo ,medicine ,Animals ,Tensin ,Myocyte ,PTEN ,Muscle, Skeletal ,Molecular Biology ,biology ,Chemistry ,PTEN Phosphohydrolase ,Skeletal muscle ,General Medicine ,021001 nanoscience & nanotechnology ,medicine.disease ,020601 biomedical engineering ,medicine.anatomical_structure ,Targeted drug delivery ,Cancer research ,biology.protein ,Nanoparticles ,Peptides ,0210 nano-technology ,Biotechnology - Abstract
Phosphatase and tensin homolog (PTEN) antagonizes muscle growth and repair, and inhibition of PTEN has been shown to improve the pathophysiology and dystrophic muscle function in the mouse model of Duchenne muscular dystrophy (DMD). However, conventional pharmacological delivery of PTEN inhibitors carries a high risk of off-target side effects in other non-muscle organs due to broad targeting spectrums. Here we report a muscle-targeted nanoparticulate platform for cell-specific delivery of a PTEN inhibitor. Poly(lactide-co-glycolide)-b-poly(ethylene glycol) nanoparticles (NPs) are functionalized with a muscle-homing peptide M12 to promote the selective uptake by muscle cells/tissue in vitro and in vivo. Moreover, the NPs are formulated to slowly release the PTEN inhibitor, preventing cytotoxicity associated with direct exposure to the drug and facilitating sustained inhibition of PTEN. This advanced delivery approach taking advantages of polymeric nanomaterials and muscle-homing peptides opens a new avenue for the development of long-term therapeutic strategies in DMD treatment.
- Published
- 2020
- Full Text
- View/download PDF
26. Sustained activation of notch signaling maintains tumor-initiating cells in a murine model of liposarcoma
- Author
-
Menchus Quan, Shihuan Kuang, and Pei-Chieh Tien
- Subjects
0301 basic medicine ,Cancer Research ,Notch signaling pathway ,Liposarcoma ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cancer stem cell ,Cell Line, Tumor ,Adipocyte ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,CD90 ,Serial Passage ,Cell Proliferation ,Receptors, Notch ,Mesenchymal stem cell ,Contact inhibition ,Endoglin ,medicine.disease ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Neoplasm Transplantation ,Signal Transduction - Abstract
Cells in a tumor are heterogeneous, often including a small number of tumor-initiating cells (TICs) and the majority of cancerous and non-cancerous cells. We have previously reported that constitutive activation of Notch signaling in adipocytes of mice leads to dedifferentiated liposarcoma (DDLPS), an aggressive liposarcoma (LPS) with no effective treatment. Here, we explored the role of Notch signaling in cellular heterogeneity of LPS. We performed serial transplantations to enrich for TICs, and derived cells exhibiting sustained Notch activation (mLPS1 cells) and cells with normal Notch activity (mLPS2 cells). Both mLPS1 and mLPS2 cells proliferated rapidly, and neither exhibited contact inhibition. However, only the mLPS1 cells exhibited tumorigenicity and gave rise to LPS upon engraftment into mice. The mLPS1 cells also highly expressed markers of cancer stem cells (Cd133), mesenchymal stem cells (Cd73, Cd90, Cd105, Dlk1) and the long non-coding RNA Rian. By contrast, the mLPS2 cells accumulated lipid droplets and expressed mature adipocyte markers when induced to differentiate. Most importantly, CRISPR-mediated disruption of Notch abrogated the tumorigenic properties of mLPS1 cells. These results reveal a key role of Notch signaling in maintaining TICs in LPS.
- Published
- 2020
- Full Text
- View/download PDF
27. Skeletal muscle IGF-1 is lower at rest and after resistance exercise in humans with obesity
- Author
-
Timothy P. Gavin, Ron T. Garner, Shihuan Kuang, Cathal J. Drohan, Julianne Stout, Yaohui Nie, Jessica A. Weiss, and Brian P. Sullivan
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Muscle Fibers, Skeletal ,Muscle Proteins ,Muscle hypertrophy ,03 medical and health sciences ,Basal (phylogenetics) ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,microRNA ,medicine ,Humans ,Orthopedics and Sports Medicine ,Obesity ,RNA, Messenger ,Insulin-Like Growth Factor I ,Phosphorylation ,Muscle, Skeletal ,Exercise ,Protein kinase B ,PI3K/AKT/mTOR pathway ,business.industry ,Public Health, Environmental and Occupational Health ,Wnt signaling pathway ,Skeletal muscle ,Resistance Training ,030229 sport sciences ,General Medicine ,medicine.anatomical_structure ,Endocrinology ,Female ,business ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Obesity is associated with numerous changes in skeletal muscle including greater muscle mass and muscle fiber cross sectional area (FCSA), yet fasted muscle protein synthesis is lower. Activation of the IGF-1/Akt/mTOR pathway is critical for muscle mass maintenance, muscle hypertrophy, and muscle protein regulation. Resistance exercise (RE) increases muscle mass, FCSA, and IGF-1. Persons with obesity have greater skeletal muscle mass and larger skeletal muscle fiber cross sectional area. The IGF-1/Akt/mTOR pathway is critical for the regulation of skeletal muscle mass. Our study found men and women with obesity have lower skeletal muscle IGF-1 mRNA and protein and higher expression of miR-206 an epigenetic regulator of IGF-1, at rest and following an acute bout of resistance exercise. Despite this, Akt mediated signaling was maintained and maintenance of phosphorylation does not appear to be accounted for by compensatory pathways. Our findings suggest a possible negative feedback mechanism via increased miR-206 and in turn decreased IGF-1 to limit further skeletal muscle hypertrophy in persons with obesity. The current work investigated if: (1) obesity dysregulates basal skeletal muscle IGF-1 pathways; and (2) obesity augments the muscle IGF-1 pathway responses to acute RE. Eight sedentary (no self-reported physical activity), lean (LN) and eight sedentary subjects with obesity (OB) had vastus lateralis biopsies taken at rest, and 15 min and 3 h post-acute RE for the measurement of the IGF-1 pathway and muscle FCSA. Type II FCSA was larger in OB vs. LN. Skeletal muscle IGF-1 mRNA and IGF-1 protein were lower in OB vs. LN at rest and post-exercise. Acute RE increased IGF-1 protein similarly in both groups. The expression of miR-206, a post-transcriptional inhibitor of IGF-1 expression, was higher in OB vs. LN and linked with lower IGF-1 mRNA (r = – 0.54). In spite of greater muscle FCSA, muscle IGF-1 expression was lower in obesity suggesting negative feedback may be limiting muscle mass expansion in obesity.
- Published
- 2020
- Full Text
- View/download PDF
28. Multivesicular body and exosome pathway responses to acute exercise
- Author
-
Timothy P. Gavin, Julianne Stout, Yaohui Nie, Ron T. Garner, Shihuan Kuang, and Jessica S. Solfest
- Subjects
Adult ,Male ,Adolescent ,Physiology ,macromolecular substances ,030204 cardiovascular system & hematology ,Biology ,Exosomes ,Exosome ,Quadriceps Muscle ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Myokine ,medicine ,Humans ,Aerobic exercise ,RNA, Messenger ,Multivesicular Body ,Muscle, Skeletal ,Exercise ,Drosha ,Nutrition and Dietetics ,Multivesicular Bodies ,Skeletal muscle ,General Medicine ,Microvesicles ,Cell biology ,MicroRNAs ,medicine.anatomical_structure ,biology.protein ,030217 neurology & neurosurgery ,Signal Transduction ,Dicer - Abstract
New findings What is the central question of this study? What is the impact of acute aerobic and aerobic + resistance (concurrent) exercise on the regulation of multivesicular body formation in human skeletal muscle? What is the main finding and its importance? Gene expression for proteins associated with multivesicular body biogenesis was increased in response to concurrent exercise, and gene expression of microRNA processing (genetic information) was increased in response to aerobic and concurrent exercise. A greater understanding of the processing of multivesicular bodies in response to acute exercise may lead to novel treatments focused on intercellular communication pathways. Abstract Regular aerobic exercise (AEx) and resistance exercise (REx) promote many beneficial adaptations. Skeletal muscle participates in intercellular communication in part through the release of myokines and extracellular vesicles including exosomes (EXOs), the latter containing mRNA, microRNA (miRNA), lipids and proteins. Exercise-induced regulation of skeletal muscle multivesicular body (MVB) biogenesis leading to EXO formation and release is poorly understood. We hypothesized that acute exercise would increase skeletal muscle MVB biogenesis and EXO release pathways with a greater response to aerobic + resistance exercise (A+REx) than to AEx alone. Twelve sedentary, healthy male subjects exercised on a cycle ergometer for 45 min (AEx) followed by single leg, knee extensor, resistance exercise (A+REx). Vastus lateralis biopsies were obtained at rest and 1 h post-exercise. Key components of the MVB biogenesis, EXO biogenesis and release, and miRNA processing pathways were analysed. Clathrin and Alix mRNA (MVB biogenesis) were increased by A+REx, while DICER and exportin mRNA (miRNA processing) were increased by AEx and A+REx. There were positive relationships between MVBs and miRNA processing genes following both AEx and A+REx consistent with coordinated regulation of these interrelated processes (Alix mRNA increased with Drosha, exportin and Dicer mRNA). Acute exercise increases the regulation of components of MVB and EXO pathways as well as miRNA processing components. A greater understanding of the production and packaging of skeletal muscle MVBs, EXOs and mature miRNA could lead to novel treatments focused on intercellular communication.
- Published
- 2020
- Full Text
- View/download PDF
29. ACSS3 in brown fat drives propionate catabolism and its deficiency leads to autophagy and systemic metabolic dysfunction
- Author
-
Zhihao Jia, Xiyue Chen, Jingjuan Chen, Lijia Zhang, Stephanie N. Oprescu, Nanjian Luo, Yan Xiong, Feng Yue, and Shihuan Kuang
- Subjects
Mice, Knockout ,Disease Models, Animal ,Mice ,Adipocytes, Brown ,Adipose Tissue, Brown ,Adipocytes, White ,Coenzyme A Ligases ,Molecular Medicine ,Medicine (miscellaneous) ,Animals ,Propionates - Abstract
Propionate is a gut microbial metabolite that has been reported to have controversial effects on metabolic health. Here we show that propionate is activated by acyl-CoA synthetase short-chain family member 3 (ACSS3), located on the mitochondrial inner membrane in brown adipocytes. Knockout of Acss3 gene (Acss3
- Published
- 2021
30. Chchd10 is dispensable for myogenesis but critical for adipose browning
- Author
-
Wei Xia, Jiamin Qiu, Ying Peng, Madigan M. Snyder, Lijie Gu, Kuilong Huang, Nanjian Luo, Feng Yue, and Shihuan Kuang
- Subjects
Cell Biology ,Developmental Biology - Abstract
The Chchd10 gene encodes a coiled-coil-helix-coiled-coil-helix-domain containing protein predicted to function in the mitochondrion and nucleus. Mutations of Chchd10 are associated with ALS, dementia and myopathy in humans and animal models, but how knockout of Chchd10 (Chchd10KO) affects various tissues especially skeletal muscle and adipose tissues remains unclear. Here we show that Chchd10 expression increases as myoblasts and preadipocytes differentiate. During myogenesis, CHCHD10 interacts with TAR DNA binding protein 43 (TDP-43) in regenerating myofibers in vivo and in newly differentiated myotubes ex vivo. Surprisingly, Chchd10KO mice had normal skeletal muscle development, growth and regeneration, with moderate defects in grip strength and motor performance. Chchd10KO similarly had no effects on development of brown and white adipose tissues (WAT). However, Chchd10KO mice had blunted response to acute cold and attenuated cold-induced browning of WAT, with markedly reduced UCP1 levels. Together, these results demonstrate that Chchd10 is dispensable for normal myogenesis and adipogenesis but is required for normal motility and cold-induced, mitochondrion-dependent browning of adipocytes. The data also suggest that human CHCHD10 mutations cause myopathy through a gain-of-function mechanism.
- Published
- 2021
31. Effects of obesity and acute resistance exercise on skeletal muscle angiogenic communication pathways
- Author
-
Ron T. Garner, Jessica A. Weiss, Yaohui Nie, Brian P. Sullivan, Christopher K. Kargl, Cathal J. Drohan, Shihuan Kuang, Julianne Stout, and Timothy P. Gavin
- Subjects
Vascular Endothelial Growth Factor A ,MicroRNAs ,Nutrition and Dietetics ,Physiology ,Physiology (medical) ,Humans ,Neovascularization, Physiologic ,Resistance Training ,General Medicine ,Obesity ,RNA, Messenger ,Muscle, Skeletal - Abstract
What is the central question of this study? Do obesity and acute resistance exercise alter the regulation of muscle intercellular communication pathways consistent with inadequate compensatory angiogenesis in response to muscle loading present in individuals with obesity? What is the main finding and its importance? Obesity is associated with differences in both pro- and anti-angiogenic signalling consistent with lower muscle capillarization. Acute resistance exercise increases the release of skeletal muscle small extracellular vesicles independent of body mass. These results identify new cellular factors associated with impaired angiogenesis in obesity and the positive effects of acute resistance exercise in lean and obese skeletal muscle.Obesity (OB) impairs cell-to-cell communication signalling. Small extracellular vesicles (EVs), which include exosomes, are released by skeletal muscle and participate in cell-to-cell communication, including the regulation of angiogenesis. Resistance exercise (REx) increases muscle fibre size and capillarization. Although obesity increases muscle fibre size, there is an inadequate increase in capillarization such that capillary density is reduced. It was hypothesized that REx-induced angiogenic signalling and EV biogenesis would be lower with obesity. Sedentary lean (LN) and OB subjects (n = 8 per group) performed three sets of single-leg knee-extension REx at 80% of maximum. Muscle biopsies were obtained at rest, 15 min and 3 h postexercise and analysed for angiogenic and EV biogenesis mRNA and protein. In OB subjects, muscle fibre size was ∼20% greater and capillary density with type II fibres ∼25% lower compared with LN subjects (P 0.001). In response to REx, the increase in VEGF mRNA (pro-angiogenic) was similar (3-fold) between groups, while thrombospondin-1 (TSP-1) mRNA (anti-angiogenic) increased ∼2.5-fold in OB subjects only (P = 0.010). miR-130a (pro-angiogenic) was ∼1.4-fold (P = 0.011) and miR-503 (anti-angiogenic) ∼1.8-fold (P = 0.017) greater in OB compared with LN subjects at all time points. In both groups, acute REx decreased the EV surface protein Alix by ∼50%, consistent with the release of exosomes (P = 0.016). Acute REx appears to induce the release of skeletal muscle small EVs independent of body mass. However, with obesity there is predominantly impaired angiogenic signalling, consistent with inadequate angiogenesis in response to basal muscle hypertrophy.
- Published
- 2021
32. Proto-oncogene FAM83A contributes to casein kinase 1–mediated mitochondrial maintenance and white adipocyte differentiation
- Author
-
Kuilong, Huang, Zhihao, Jia, Haoran, Li, Ying, Peng, Xiaochang, Chen, Nanjian, Luo, Tongxing, Song, Yingqian, Wang, Xin'e, Shi, Shihuan, Kuang, and Gongshe, Yang
- Subjects
Mice ,Adipogenesis ,Casein Kinase I ,3T3-L1 Cells ,Adipocytes, White ,Proto-Oncogenes ,Animals ,Cell Differentiation ,Cell Biology ,Molecular Biology ,Biochemistry ,Mitochondria ,Neoplasm Proteins - Abstract
Family with sequence similarity 83 A (FAM83A) is a newly discovered proto-oncogene that has been shown to play key roles in various cancers. However, the function of FAM83A in other physiological processes is not well known. Here, we report a novel function of FAM83A in adipocyte differentiation. We used an adipocyte-targeting fusion oligopeptide (FITC-ATS-9R) to deliver a FAM83A-sgRNA/Cas9 plasmid to knockdown Fam83a (ATS/sg-FAM83A) in white adipose tissue in mice, which resulted in reduced white adipose tissue mass, smaller adipocytes, and mitochondrial damage that was aggravated by a high-fat diet. In cultured 3T3-L1 adipocytes, we found loss or knockdown of Fam83a significantly repressed lipid droplet formation and downregulated the expression of lipogenic genes and proteins. Furthermore, inhibition of Fam83a decreased mitochondrial ATP production through blockage of the electron transport chain, associated with enhanced apoptosis. Mechanistically, we demonstrate FAM83A interacts with casein kinase 1 (CK1) and promotes the permeability of the mitochondrial outer membrane. Furthermore, loss of Fam83a in adipocytes hampered the formation of the TOM40 complex and impeded CK1-driven lipogenesis. Taken together, these results establish FAM83A as a critical regulator of mitochondria maintenance during adipogenesis.
- Published
- 2022
- Full Text
- View/download PDF
33. LETMD1 is required for mitochondrial structure and thermogenic function of brown adipocytes
- Author
-
Kun Ho Kim, Ying Peng, Jiamin Qiu, Stephanie N. Oprescu, Pengpeng Bi, Renjie Shang, Lijia Zhang, Shawn S. Donkin, Madigan M. Snyder, Feng Yue, Jingjuan Chen, and Shihuan Kuang
- Subjects
Receptors, Cell Surface ,Biology ,Mitochondrion ,medicine.disease_cause ,Biochemistry ,Mice ,Downregulation and upregulation ,Adipose Tissue, Brown ,Gene expression ,Brown adipose tissue ,Genetics ,medicine ,Animals ,Obesity ,Molecular Biology ,Mice, Knockout ,Oncogene Proteins ,Thermogenesis ,Thermogenin ,Cell biology ,Mitochondria ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Adipocytes, Brown ,Bacterial outer membrane ,Oxidative stress ,Biotechnology - Abstract
Obesity and metabolic disorders caused by energy surplus pose an increasing concern within the global population. Brown adipose tissue (BAT) dissipates energy through mitochondrial non-shivering thermogenesis, thus representing a powerful agent against obesity. Here we explore the novel role of a mitochondrial outer membrane protein, LETM1-domain containing 1 (LETMD1), in BAT. We generated a knockout (Letmd1KO ) mouse model and analyzed BAT morphology, function and gene expression under various physiological conditions. While the Letmd1KO mice are born normally and have normal morphology and body weight, they lose multilocular brown adipocytes completely and have diminished mitochondrial abundance, DNA copy number, cristae structure, and thermogenic gene expression in the intrascapular BAT, associated with elevated reactive oxidative stress. In consequence, the Letmd1KO mice fail to maintain body temperature in response to acute cold exposure without food and become hypothermic within 4 h. Although the cold-exposed Letmd1KO mice can maintain body temperature in the presence of food, they cannot upregulate expression of uncoupling protein 1 (UCP1) and convert white to beige adipocytes, nor can they respond to adrenergic stimulation. These results demonstrate that LETMD1 is essential for mitochondrial structure and function, and thermogenesis of brown adipocytes.
- Published
- 2021
34. Polymeric Carriers for Controlled Drug Delivery in Obesity Treatment
- Author
-
Meng Deng, Shihuan Kuang, and Di Huang
- Subjects
medicine.medical_specialty ,Polymers ,business.industry ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,medicine.disease ,Controlled release ,Obesity ,Article ,03 medical and health sciences ,Human health ,Drug Delivery Systems ,0302 clinical medicine ,Endocrinology ,Pharmacotherapy ,Antiobesity drugs ,Drug delivery ,medicine ,Humans ,Anti-Obesity Agents ,Metabolic syndrome ,Adverse effect ,Intensive care medicine ,business - Abstract
The global rise in the prevalence of obesity and affiliated metabolic syndrome poses a significant threat to human health. Various approaches, including bariatric surgery and pharmacotherapy, have been used in the clinical setting for obesity treatment; however, these conventional options remain ineffective and carry risks of adverse effects. Therefore, treatments with higher efficacy and specificity are urgently required. Emerging drug delivery systems use polymeric materials and chemical strategies to improve therapeutic efficacy and specificity through stabilization and spatiotemporally controlled release of antiobesity agents. In this review, we provide insights into current treatments for obesity with a focus on recent developments of polymeric carriers for enhanced antiobesity drug delivery.
- Published
- 2019
- Full Text
- View/download PDF
35. Skeletal muscle‐derived exosomes regulate endothelial cell functions via reactive oxygen species‐activated nuclear factor‐κB signalling
- Author
-
Yoriko Sato, Chao Wang, Christopher K. Kargl, Yaohui Nie, Shihuan Kuang, Ron T. Garner, Timothy P. Gavin, and Christopher J. Gilpin
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Physiology ,Angiogenesis ,Muscle Fibers, Skeletal ,Neovascularization, Physiologic ,030204 cardiovascular system & hematology ,Exosomes ,Exosome ,Cell Line ,Mice ,03 medical and health sciences ,Paracrine signalling ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Movement ,Physiology (medical) ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Myocyte ,Muscle, Skeletal ,Cell Proliferation ,Nutrition and Dietetics ,NF-kappa B ,Skeletal muscle ,Mesenchymal Stem Cells ,General Medicine ,Microvesicles ,Cell biology ,Mice, Inbred C57BL ,Vascular endothelial growth factor ,Endothelial stem cell ,MicroRNAs ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,chemistry ,Reactive Oxygen Species ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
New findings What is the central question of this study? Capillary rarefaction is found in diabetic and aged muscle, whereas exercise increases skeletal muscle angiogenesis. The association implies a crosstalk between muscle cells and endothelial cells. The underlying mechanisms mediating the crosstalk between these cells remains to be elucidated fully. What is the main finding and its importance? Endothelial cell functions are regulated by skeletal muscle cell-derived exosomes via a vascular endothelial growth factor-independent pathway. This study reveals a new mechanism mediating the crosstalk between skeletal muscle cells and endothelial cells. Abstract Loss of skeletal muscle capillarization, known as capillary rarefaction, is found in type 2 diabetes, chronic heart failure and healthy ageing and is associated with impaired delivery of substrates to the muscle. However, the interaction and communication of skeletal muscle with endothelial cells in the regulation of capillaries surrounding the muscle remains elusive. Exosomes are a type of secreted extracellular vesicle containing mRNAs, proteins and, especially, microRNAs that exert paracrine and endocrine effects. In this study, we investigated whether skeletal muscle-derived exosomes (SkM-Exo) regulate the endothelial cell functions of angiogenesis. We demonstrated that C2C12 myotube-derived exosomes improved endothelial cell functions, assessed by the proliferation, migration and tube formation of human umbilical vein endothelial cells (HUVECs), which were increased by 20, 23 and 40%, respectively, after SkM-Exo exposure. The SkM-Exo failed to activate HUVEC vascular endothelial growth factor (VEGF) signalling. The SkM-Exo increased HUVEC reactive oxygen species and activated the nuclear factor-κB pathway, suggesting that SkM-Exo-induced angiogenesis was mediated by a VEGF-independent pathway. In addition, several angiogenic microRNAs were packaged in SkM-Exo, with miR-130a being particularly enriched and successfully transferred from SkM-Exo to HUVECs. Delivery of miRNAs into endothelial cells might explain the enhancement of reactive oxygen species production and angiogenesis by SkM-Exo. The potential angiogenic effect of SkM-Exo could provide an effective therapy for promoting skeletal muscle angiogenesis in diseases characterized by capillary rarefaction or inadequate angiogenesis.
- Published
- 2019
- Full Text
- View/download PDF
36. Heat therapy improves soleus muscle force in a model of ischemia-induced muscle damage
- Author
-
Caitlin A. Casey, Bruno T. Roseguini, Bohyun Ro, Shihuan Kuang, Blake A. Reid, Kyoungrae Kim, and Qifan Song
- Subjects
Male ,medicine.medical_specialty ,Hot Temperature ,Physiology ,medicine.medical_treatment ,Ischemia ,030204 cardiovascular system & hematology ,Muscle damage ,Mice ,Peripheral Arterial Disease ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Muscle fibre ,Muscle, Skeletal ,Hydrotherapy ,Soleus muscle ,Chemistry ,Skeletal muscle ,Hindlimb ischemia ,medicine.disease ,Heat therapy ,Heat stress ,Femoral Artery ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,030217 neurology & neurosurgery ,Muscle Contraction - Abstract
Leg muscle ischemia in patients with peripheral artery disease (PAD) leads to alterations in skeletal muscle morphology and reduced leg strength. We tested the hypothesis that exposure to heat therapy (HT) would improve skeletal muscle function in a mouse model of ischemia-induced muscle damage. Male 42-wk-old C57Bl/6 mice underwent ligation of the femoral artery and were randomly assigned to receive HT (immersion in a water bath at 37°C, 39°C, or 41°C for 30 min) or a control intervention for 3 wk. At the end of the treatment, the animals were anesthetized and the soleus and extensor digitorum longus (EDL) muscles were harvested for the assessment of contractile function and examination of muscle morphology. A subset of animals was used to examine the impact of a single HT session on the expression of genes involved in myogenesis and the regulation of muscle mass. Relative soleus muscle mass was significantly higher in animals exposed to HT at 39°C compared with the control group (control: 0.36 ± 0.01 mg/g versus 39°C: 0.40 ± 0.01 mg/g, P = 0.024). Maximal absolute force of the soleus was also significantly higher in animals treated with HT at 37°C and 39°C (control: 274.7 ± 6.6 mN; 37°C: 300.1 ± 7.7 mN; 39°C: 299.5 ± 10 mN, P < 0.05). In the soleus, but not the EDL muscle, a single session of HT enhanced the mRNA expression of myogenic factors as well as of both positive and negative regulators of muscle mass. These findings suggest that the beneficial effects of HT are muscle specific and dependent on the treatment temperature in a model of PAD. NEW & NOTEWORTHY This is the first study to comprehensively examine the impact of temperature and muscle fiber type composition on the adaptations to repeated heat stress in a model of ischemia-induced muscle damage. Exposure to heat therapy (HT) at 37°C and 39°C, but not at 41°C, improved force development of the isolated soleus muscle. These results suggest that HT may be a practical therapeutic tool to restore muscle mass and strength in patients with peripheral artery disease.
- Published
- 2019
- Full Text
- View/download PDF
37. Methyltransferase‐like 21e inhibits 26S proteasome activity to facilitate hypertrophy of type IIb myofibers
- Author
-
Shihuan Kuang, Chao Wang, Keping Hu, Yaohui Nie, Anna C. Ratliff, Jingjuan Chen, Justine V. Arrington, Feng Yue, Xiaobo Sun, Christine A. Hrycyna, Bin Zhang, Wen Jin, Yan Xiong, and W. Andy Tao
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Immunoprecipitation ,Blotting, Western ,Muscle Fibers, Skeletal ,Myostatin ,Real-Time Polymerase Chain Reaction ,Biochemistry ,Muscle hypertrophy ,Bortezomib ,Myoblasts ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Myofibrils ,Genetics ,medicine ,Animals ,Myocyte ,Muscle, Skeletal ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,biology ,Sequence Analysis, RNA ,Chemistry ,Research ,Cell Differentiation ,Methyltransferases ,Muscle atrophy ,Cell biology ,Blot ,Muscular Atrophy ,030104 developmental biology ,Real-time polymerase chain reaction ,Proteasome ,Mutation ,biology.protein ,Female ,medicine.symptom ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Skeletal muscles contain heterogeneous myofibers that are different in size and contractile speed, with type IIb myofiber being the largest and fastest. Here, we identify methyltransferase-like 21e (Mettl21e), a member of newly classified nonhistone methyltransferases, as a gene enriched in type IIb myofibers. The expression of Mettl21e was strikingly up-regulated in hypertrophic muscles and during myogenic differentiation in vitro and in vivo. Knockdown (KD) of Mettl21e led to atrophy of cultured myotubes, and targeted mutation of Mettl21e in mice reduced the size of IIb myofibers without affecting the composition of myofiber types. Mass spectrometry and methyltransferase assay revealed that Mettl21e methylated valosin-containing protein (Vcp/p97), a key component of the ubiquitin-proteasome system. KD or knockout of Mettl21e resulted in elevated 26S proteasome activity, and inhibition of proteasome activity prevented atrophy of Mettl21e KD myotubes. These results demonstrate that Mettl21e functions to maintain myofiber size through inhibiting proteasome-mediated protein degradation.—Wang, C., Zhang, B., Ratliff, A. C., Arrington, J., Chen, J., Xiong, Y., Yue, F., Nie, Y., Hu, K., Jin, W., Tao, W. A., Hrycyna, C. A., Sun, X., Kuang, S. Methyltransferase-like 21e inhibits 26S proteasome activity to facilitate hypertrophy of type IIb myofibers.
- Published
- 2019
- Full Text
- View/download PDF
38. Effects of acute aerobic and concurrent exercise on skeletal muscle metabolic enzymes in untrained men
- Author
-
Julianne Stout, Yaohui Nie, Shihuan Kuang, Timothy P. Gavin, Jessica A. Weiss, Ron T. Garner, and Jessica S. Solfest
- Subjects
Mitochondrial enzymes ,Knee extensors ,Chemistry ,Repetition maximum ,Resistance training ,Skeletal muscle ,VO2 max ,030229 sport sciences ,030204 cardiovascular system & hematology ,03 medical and health sciences ,Crystallography ,0302 clinical medicine ,medicine.anatomical_structure ,Metabolic enzymes ,Post exercise ,medicine ,Orthopedics and Sports Medicine - Abstract
Acute exercise can increase skeletal muscle citrate synthase (CS) enzyme activity and resting skeletal muscle mitochondrial enzyme activity has been linked to maximal oxygen consumption ( $$\dot {V}{{\text{O}}_{2\hbox{max} }}$$ ). We investigated: (1) if acute aerobic exercise (AEx) increases muscle metabolic enzyme activities other than CS; (2) if the addition of acute resistance exercise (REx) enhances the response to AEx (A + REx); and (3) if post-exercise muscle metabolic enzyme activity was related to $$\dot {V}{{\text{O}}_{2\hbox{max} }}$$ . Twelve young, sedentary men completed 45 min of two-legged cycle ergometry at 55% of $$\dot {V}{{\text{O}}_{2\hbox{max} }}$$ and 3 sets of 8–12 repetitions of one-leg knee extensor at 55% of 1 repetition maximum (1-RM). Vastus lateralis biopsies were taken prior to and 1 h post AEx and A + REx for the measurement of phosphofructokinase (PFK), 3-l-hydroxyacyl CoA dehydrogenase (β-HAD), succinate dehydrogenase (SDH) and CS. As a group, there was no effect of acute AEx or A + REx on muscle PFK, β-HAD, CS, and SDH activities. Post exercise muscle PFK, β-HAD, CS, and SDH activities were related to higher $$\dot {V}{{\text{O}}_{2\hbox{max} }}$$ (r = 0.62–0.74). With participants grouped by $$\dot {V}{{\text{O}}_{2\hbox{max} }}$$ (LOW, 50th %), acute exercise-induced changes in muscle PFK, β-HAD, CS, and SDH were greater in NORM compared to LOW. These findings suggest acute exercise muscle metabolic enzyme activities are predictive of $$\dot {V}{{\text{O}}_{2\hbox{max} }}$$ and possibly supportive of higher $$\dot {V}{{\text{O}}_{2\hbox{max} }}$$ . Also, low $$\dot {V}{{\text{O}}_{2\hbox{max} }}$$ (below 30th percentile) appears to impair skeletal muscle metabolic enzyme responses to acute exercise.
- Published
- 2019
- Full Text
- View/download PDF
39. Impact of heat therapy on recovery after eccentric exercise in humans
- Author
-
Bruno T. Roseguini, Qifan Song, Timothy P. Gavin, Shihuan Kuang, and Kyoungrae Kim
- Subjects
Adult ,Male ,Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Hot Temperature ,Knee Joint ,Physiology ,medicine.medical_treatment ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Physical medicine and rehabilitation ,Physiology (medical) ,Intervention (counseling) ,medicine ,Humans ,Muscle Strength ,RNA, Messenger ,Muscle, Skeletal ,Exercise ,business.industry ,030229 sport sciences ,musculoskeletal system ,Functional recovery ,Heat therapy ,Thigh ,Torque ,Eccentric exercise ,Muscle Fatigue ,Angiogenesis Inducing Agents ,Female ,business ,030217 neurology & neurosurgery - Abstract
The purpose of this study was to investigate the effects of heat therapy (HT) on functional recovery, the skeletal muscle expression of angiogenic factors, macrophage content, and capillarization after eccentric exercise in humans. Eleven untrained individuals (23.8 ± 0.6 yr) performed 300 bilateral maximal eccentric contractions of the knee extensors. One randomly selected thigh was treated with five daily 90-min sessions of HT, whereas the opposite thigh received a thermoneutral intervention. Peak isokinetic torque of the knee extensors was assessed at baseline and daily for 4 days and fatigue resistance was assessed at baseline and 1 and 4 days after the eccentric exercise session. Muscle biopsies were obtained 2 wk before and 1 and 5 days after the eccentric exercise bout. There were no differences between thighs in the overall recovery profile of peak torque. However, the thigh exposed to HT had greater fatigue resistance than the thigh exposed to the thermoneutral intervention. The change from baseline in mRNA expression of vascular endothelial growth factor (VEGF) was higher at day 1 in the thigh exposed to HT. Protein levels of VEGF and angiopoietin 1 were also significantly higher in the thigh treated with HT. The number of capillaries around type II fibers decreased similarly in both thighs at day 5. Exposure to HT had no impact on macrophage content. These results suggest that HT accelerates the recovery of fatigue resistance after eccentric exercise and promotes the expression of angiogenic factors in human skeletal muscle. NEW & NOTEWORTHY We investigated whether exposure to local heat therapy (HT) accelerates recovery after a bout of eccentric exercise in humans. Compared with a thermoneutral control intervention, HT improved fatigue resistance of the knee extensors and enhanced the expression of the angiogenic mediators vascular endothelial growth factor and angiopoietin 1. These results suggest that HT hastens functional recovery and enhances the expression of regulatory factors involved in muscle repair after eccentric exercise in humans.
- Published
- 2019
- Full Text
- View/download PDF
40. Effect of PGC1‐α Overexpression on Cardiotoxin‐Induced Damage and Repair of Human Myotubes
- Author
-
Shihuan Kuang, Allison Ellis, Christopher K. Kargl, Timothy P. Gavin, and Brian P. Sullivan
- Subjects
Cardiotoxin ,Myogenesis ,Chemistry ,Genetics ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2021
- Full Text
- View/download PDF
41. Skeletal Muscle Extracellular Vesicles Regulate Endothelial Cells in a Fiber Type Dependent Manner
- Author
-
Zhihao Jia, Timothy P. Gavin, Jonathan H. Shannahan, Chris K. Kargl, Shihuan Kuang, and Autumn Yang
- Subjects
medicine.anatomical_structure ,Dependent manner ,Chemistry ,Genetics ,medicine ,Skeletal muscle ,A fibers ,Molecular Biology ,Biochemistry ,Extracellular vesicles ,Biotechnology ,Cell biology - Published
- 2021
- Full Text
- View/download PDF
42. 193 Single Cell RNA-sequencing Reveals a Role of Lipid Metabolism in Muscle Satellite Cells
- Author
-
Feng Yue, Stephanie N. Oprescu, and Shihuan Kuang
- Subjects
biology ,Chemistry ,Cell ,RNA ,Lipid metabolism ,General Medicine ,biology.organism_classification ,Cell biology ,medicine.anatomical_structure ,Oral Presentations ,Genetics ,medicine ,Animal Science and Zoology ,Satellite (biology) ,Food Science - Abstract
Single Cell RNA-sequencing (scRNA-seq) is a powerful technique to deconvolute gene expression of various subset of cells intermingled within a complex tissue, such as the skeletal muscle. We first used scRNA-seq to understand dynamics of cell populations and their gene expression during muscle regeneration in murine limb muscles. This leads to the identification of a subset of satellite cells (the resident stem cells of skeletal muscles) with immune gene signatures in regenerating muscles. Next, we used scRNA-seq to examine gene expression dynamics of satellite cells at various status: quiescence, activation, proliferation, differentiation and self-renewal. This analysis uncovers stage-dependent changes in expression of genes related to lipid metabolism. Further analyses lead to the discovery of previously unappreciated dynamics of lipid droplets in satellite cells; and demonstrate that the abundance of the lipid droplets in newly divided satellite daughter cells is linked to cell fate segregation into differentiation versus self-renewal. Perturbation of lipid droplet dynamics through blocking lipolysis disrupts cell fate homeostasis and impairs muscle regeneration. Finally, we show that lipid metabolism regulates the function of satellite cells through two mechanisms. On one hand, lipid metabolism functions as an energy source through fatty acid oxidation (FAO), and blockage of FAO reduces energy production that is critical for satellite cell function. On the other hand, lipid metabolism generates bioactive molecules that influence signaling transduction and gene expression. In this scenario, lipid metabolism and FAO regulate the intracellular levels of acetyl-coA and selective acetylation of PAX7, a pivotal transcriptional factor underlying function of satellite cells. These results together reveal for the first time a critical role of lipid metabolism and lipid droplet dynamics in muscle satellite cell fate determination and regenerative function; and underscore a potential role of dietary fatty acids in satellite cell-dependent muscle development, growth and regeneration.
- Published
- 2021
- Full Text
- View/download PDF
43. P28.02 Beyond PACIFIC: Outcomes and Toxicity According to Durvalumab Dosing Schedule Every 2 versus 4 Weeks
- Author
-
Eric Berthelet, Barbara Melosky, Shihuan Kuang, Cheryl Ho, Aria Shokoohi, T. Zhang, Sophie Sun, M. Denault, Mitchell Liu, Bonnie Leung, and Janessa Laskin
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Schedule ,Durvalumab ,Oncology ,business.industry ,Toxicity ,Emergency medicine ,Medicine ,Dosing ,business - Published
- 2021
- Full Text
- View/download PDF
44. Reduced electron transport chain complex I protein abundance and function in Mfn2‐deficient myogenic progenitors lead to oxidative stress and mitochondria swelling
- Author
-
Nanjian Luo, Yongju Zhao, Feng Yue, Shihuan Kuang, Jingjuan Chen, Qing Deng, and Zhihao Jia
- Subjects
Male ,0301 basic medicine ,Mitochondrial DNA ,Protein subunit ,MFN2 ,Mitochondrion ,Muscle Development ,DNA, Mitochondrial ,Biochemistry ,Article ,GTP Phosphohydrolases ,Mice ,03 medical and health sciences ,Mitofusin-2 ,Oxygen Consumption ,0302 clinical medicine ,Physical Conditioning, Animal ,Genetics ,Animals ,Myocyte ,Muscle, Skeletal ,Molecular Biology ,Mice, Knockout ,Electron Transport Complex I ,Chemistry ,Myogenesis ,Stem Cells ,Mitochondria, Muscle ,Cell biology ,Oxidative Stress ,030104 developmental biology ,Electron Transport Chain Complex Proteins ,Gene Expression Regulation ,mitochondrial fusion ,Female ,Reactive Oxygen Species ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Mitochondrial remodeling through fusion and fission is crucial for progenitor cell differentiation but its role in myogenesis is poorly understood. Here, we characterized the function of mitofusin 2 (Mfn2), a mitochondrial outer membrane protein critical for mitochondrial fusion, in muscle progenitor cells (myoblasts). Mfn2 expression is upregulated during myoblast differentiation in vitro and muscle regeneration in vivo. Targeted deletion of Mfn2 gene in myoblasts (Mfn2(MKO)) increases oxygen-consumption rates (OCR) associated with the maximal respiration and spare respiratory capacity, and increased levels of reactive oxygen species (ROS). Skeletal muscles of Mfn2(MKO) mice exhibit robust mitochondrial swelling with normal mitochondrial DNA content. Additionally, mitochondria isolated from Mfn2(MKO) muscles have reduced OCR at basal state and for complex I respiration, associated with decreased levels of complex I proteins NDUFB8 (NADH ubiquinone oxidoreductase subunit B8) and NDUFS3 (NADH ubiquinone oxidoreductase subunit S3). However, Mfn2(MKO) has no obvious effects on myoblast differentiation, muscle development and function, and muscle regeneration. These results demonstrate a novel role of Mfn2 in regulating mitochondrial complex I protein abundance and respiratory functions in myogenic progenitors and myofibers.
- Published
- 2021
- Full Text
- View/download PDF
45. Chemically-defined generation of human hemogenic endothelium and definitive hematopoietic progenitor cells
- Author
-
Yun Chang, Ramizah Syahirah, Stephanie N. Oprescu, Xuepeng Wang, Juhyung Jung, Scott H. Cooper, Sandra Torregrosa-Allen, Bennett D. Elzey, Alan Y. Hsu, Lauren N. Randolph, Yufei Sun, Shihuan Kuang, Hal E. Broxmeyer, Qing Deng, Xiaojun Lian, and Xiaoping Bao
- Subjects
Hemangioblasts ,Biophysics ,Cell Differentiation ,Bioengineering ,Hematopoietic Stem Cells ,Article ,Hematopoiesis ,Biomaterials ,Mice ,Mechanics of Materials ,Mesonephros ,Ceramics and Composites ,Animals ,Humans ,Zebrafish - Abstract
Human hematopoietic stem cells (HSCs), which arise from aorta-gonad-mesonephros (AGM), are widely used to treat blood diseases and cancer. However, a technique for their robust generation in vitro is still missing. Here we show temporal manipulation of Wnt signaling is sufficient and essential to induce AGM-like hematopoiesis from human pluripotent stem cells. TGFβ inhibition at the stage of aorta-like SOX17(+)CD235a(−) hemogenic endothelium yielded AGM-like hematopoietic progenitors, which closely resembled primary cord blood HSCs at the transcriptional level and contained diverse lineage-primed progenitor populations via single cell RNA-sequencing analysis. Notably, the resulting definitive cells presented lymphoid and myeloid potential in vitro; and could home to a definitive hematopoietic site in zebrafish and rescue bloodless zebrafish after transplantation. Engraftment and multilineage repopulating activities were also observed in mouse recipients. Together, our work provided a chemically-defined and feeder-free culture platform for scalable generation of AGM-like hematopoietic progenitor cells, leading to enhanced production of functional blood and immune cells for various therapeutic applications.
- Published
- 2022
- Full Text
- View/download PDF
46. Phosphatase orphan 1 inhibits myoblast proliferation and promotes myogenic differentiation
- Author
-
Gongshe Yang, Shihuan Kuang, Feng Yue, Jingjuan Chen, Wei Xia, Ying Peng, and Kuilong Huang
- Subjects
0301 basic medicine ,Myoblast proliferation ,Cell signaling ,Myoblasts, Skeletal ,Phosphatase ,Muscle Development ,Biochemistry ,Article ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,Myocyte ,Animals ,Molecular Biology ,Myogenin ,Cell Proliferation ,Gene knockdown ,Myogenesis ,Chemistry ,Cell Differentiation ,Phosphoric Monoester Hydrolases ,Cell biology ,Mitochondria, Muscle ,030104 developmental biology ,Myogenic regulatory factors ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Myogenesis includes sequential stages of progenitor cell proliferation, myogenic commitment and differentiation, myocyte fusion, and myotube maturation. Different stages of myogenesis are orchestrated and regulated by myogenic regulatory factors and various downstream cellular signaling. Here we identify phosphatase orphan 1 (Phospho1) as a new player in myogenesis. During activation, proliferation, and differentiation of quiescent satellite cells, the expression of Phospho1 gradually increases. Overexpression of Phospho1 inhibits myoblast proliferation but promotes their differentiation and fusion. Conversely, knockdown of Phospho1 accelerates myoblast proliferation but impairs myotube formation. Moreover, knockdown of Phospho1 decreases the OXPHO protein levels and mitochondria density, whereas overexpression of Phospho1 upregulates OXPHO protein levels and promotes mitochondrial oxygen consumption. Finally, we show that Phospho1 expression is controlled by myogenin, which binds to the promoter of Phospho1 to regulate its transcription. These results indicate a key role of Phospho1 in regulating myogenic differentiation and mitochondrial function.
- Published
- 2020
47. Rapid Profiling of Isomeric Unsaturated Lipids Through Online Photochemical Derivatization of C=C Bonds
- Author
-
Shihuan Kuang, Xiaofei Sun, Jiamin Qiu, Pei Su, Julia Laskin, Yingju Li, Sudhansu K. Dey, and Daisy Unsihuay
- Subjects
Desorption electrospray ionization ,chemistry.chemical_compound ,Resolution (mass spectrometry) ,Chemistry ,Electrospray ionization ,Lipidomics ,Structural isomer ,Lipidome ,Photochemistry ,Derivatization ,Mass spectrometry - Abstract
Unraveling the complexity of the lipidome requires the development of novel approaches for the structural characterization of lipid species with isomeric resolution. Herein, we introduce an online photochemical approach for lipid isomer identification through selective derivatization of double bonds by reaction with singlet oxygen. Lipid hydroperoxide products are generated promptly after laser irradiation. Fragmentation of these species in a mass spectrometer produces diagnostic fragments, which reveal the C=C locations in the unreacted lipids. This approach uses an inexpensive light source and photosensitizer making it easy to incorporate into any lipidomics workflow. We demonstrate the utility of this approach for the shotgun profiling of C=C locations in different lipid classes present in tissue extracts using electrospray ionization (ESI) and for spatially-resolved analysis of lipids in tissue sections using nanospray desorption electrospray ionization (nano-DESI). These results provide a path for both rapid profiling and ambient imaging of positional isomers in biological samples.
- Published
- 2020
- Full Text
- View/download PDF
48. In Vitro Evaluation of Clinical Candidates of γ-Secretase Inhibitors: Effects on Notch Inhibition and Promoting Beige Adipogenesis and Mitochondrial Biogenesis
- Author
-
Jiamin Qiu, Shihuan Kuang, Meng Deng, and Di Huang
- Subjects
Cell Survival ,Notch signaling pathway ,Pharmaceutical Science ,Apoptosis ,02 engineering and technology ,Biology ,030226 pharmacology & pharmacy ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,3T3-L1 Cells ,Adipocytes ,medicine ,Animals ,Pharmacology (medical) ,Enzyme Inhibitors ,Cytotoxicity ,Cell Proliferation ,Pharmacology ,Adipogenesis ,Organelle Biogenesis ,Dose-Response Relationship, Drug ,Receptors, Notch ,Organic Chemistry ,Cancer ,Translation (biology) ,Lipid Droplets ,Adipose Tissue, Beige ,021001 nanoscience & nanotechnology ,medicine.disease ,In vitro ,Mitochondrial biogenesis ,Cancer research ,Molecular Medicine ,Anti-Obesity Agents ,Amyloid Precursor Protein Secretases ,0210 nano-technology ,Biogenesis ,Biotechnology - Abstract
PURPOSE: Inhibition of Notch signaling has been recently demonstrated to promote beige adipocyte biogenesis. However, most γ-secretase inhibitors (GSIs) used to achieve pharmacological inhibition of Notch signaling are at the basic research or preclinical stage, limiting the translation of fundamental findings into clinical practice. This present study aimed to evaluate the potential of several clinical candidates of GSIs as browning agents for the treatment of obesity. METHODS: Seven GSIs that are clinical candidates for the treatment of Alzheimer’s disease or cancer were selected and their impacts on Notch inhibition as well as promoting beige biogenesis were compared using in vitro culture of 3T3-L1 preadipocytes. RESULTS: Four compounds (i.e.RO4929097, PF-03084014, LY3039478, and BMS-906024) that efficiently inhibited the expression of Notch target genes in 3T3-L1 preadipocytes were identified. Moreover, these compounds were optimized for dose-dependent effects at three gradient concentrations (0.5, 1, and 10 μM) to promote beige adipogenesis and mitochondrial biogenesis in 3T3-L1 preadipocytes without causing severe cytotoxicity. CONCLUSIONS: Our findings not only highlight the potential of cross-therapeutic application of these GSIs for obesity treatment via inhibition of γ-secretase-mediated processing of Notch signaling, but also provide important experimental evidence to support further design and development of clinically translatable Notch-inhibiting drug delivery systems.
- Published
- 2020
- Full Text
- View/download PDF
49. Exosomal Secretion of Adipose Tissue during Various Physiological States
- Author
-
Menchus Quan and Shihuan Kuang
- Subjects
Cell ,Pharmaceutical Science ,Adipose tissue ,Inflammation ,02 engineering and technology ,Biology ,Exosomes ,030226 pharmacology & pharmacy ,Energy homeostasis ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Adipocyte ,Neoplasms ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Obesity ,Exosomal secretion ,Pharmacology ,Metabolic Syndrome ,Secretory Pathway ,Organic Chemistry ,021001 nanoscience & nanotechnology ,Microvesicles ,Cell biology ,Insulin receptor ,medicine.anatomical_structure ,chemistry ,Adipose Tissue ,biology.protein ,Molecular Medicine ,medicine.symptom ,0210 nano-technology ,Biotechnology ,Signal Transduction - Abstract
Exosomes are secreted extracellular vesicles containing a wide array of biologically active components. Recent studies have demonstrated that exosomes serve as an important vehicle for extracellular communication and exert systemic effects on the physiology of organisms. Adipose tissues (ATs) play a key role in balancing systemic energy homeostasis as a central hub for fatty acid metabolism. At the same time, proper endocrine function of ATs has also been shown to be crucial for regulating physiological and metabolic health. The endocrine function of ATs is partially mediated by AT-derived exosomes that regulate metabolic homeostasis, such as insulin signaling, lipolysis, and inflammation. During the pathogenesis of obesity, metabolic syndrome, and cancer, exosomes shed by the resident cells in ATs may also have a role in regulating the progression of these diseases along with associated pathologies. In this review, we summarize the contents of AT-derived exosomes and their effects on various cell populations along with possible underlying molecular mechanisms. We further discuss the potential applications of exosomes as a drug delivery tool and therapeutic target.
- Published
- 2020
50. PTEN Inhibition Ameliorates Muscle Degeneration and Improves Muscle Function in a Mouse Model of Duchenne Muscular Dystrophy
- Author
-
Feng Yue, Shihuan Kuang, Naagarajan Narayanan, Changyou Song, Jiamin Qiu, Zhihao Jia, Stephanie N. Oprescu, Zhengrong Yuan, Bruno T. Roseguini, Meng Deng, and Di Huang
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Duchenne muscular dystrophy ,Inflammation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Atrophy ,Lipid biosynthesis ,Internal medicine ,Drug Discovery ,Genetics ,medicine ,Animals ,PTEN ,Myocyte ,Tensin ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,biology ,business.industry ,Muscles ,medicine.disease ,Muscular Dystrophy, Duchenne ,Muscular Atrophy ,Endocrinology ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Original Article ,medicine.symptom ,business ,Dystrophin - Abstract
Duchenne Muscular Dystrophy (DMD) is caused by mutation of the muscle membrane protein dystrophin and characterized by severe degeneration of myofibers, progressive muscle wasting and loss of mobility, ultimately cardiorespiratory failure and premature death. Here we report that skeletal muscle-specific knockout (KO) of Phosphatase and tensin homolog (Pten) gene in an animal model of DMD (mdx mice) alleviates myofiber degeneration and restores muscle function without increasing tumor incidences. Specifically, Pten KO normalizes myofiber size and prevents muscular atrophy, and improves grip strength and exercise performance of mdx mice. Pten KO also reduces fibrosis and inflammation; and ameliorates muscle pathology in mdx mice. Moreover, we found that Pten KO upregulates extracellular matrix and basement membrane components positively correlated to wound healing, but suppresses negative regulators of wound healing and lipid biosynthesis; and restores the integrity of muscle basement membrane in mdx mice. Importantly, pharmacological inhibition of PTEN similarly ameliorates muscle pathology and improves muscle integrity and function in mdx mice. Our finding provides evidence that PTEN inhibition may represent a potential therapeutic strategy to restore muscle function in DMD.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.