1
|
Trybulski R, Kużdżał A, Stanula A, Muracki J, Kawczyński A, Kuczmik W, Wang HK. Acute effects of cold, heat and contrast pressure therapy on forearm muscles regeneration in combat sports athletes: a randomized clinical trial. Sci Rep 2024; 14:22410. [PMID: 39333728 PMCID: PMC11437117 DOI: 10.1038/s41598-024-72412-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/06/2024] [Indexed: 09/29/2024] Open
Abstract
Due to the specific loads that occur in combat sports athletes' forearm muscles, we decided to compare the immediate effect of monotherapy with the use of compressive heat (HT), cold (CT), and alternating therapy (HCT) in terms of eliminating muscle tension, improving muscle elasticity and tissue perfusion and forearm muscle strength. This is a single-blind, randomized, experimental clinical trial. Group allocation was performed using simple 1:1 sequence randomization using the website randomizer.org. The study involved 40 40 combat sports athletes divided into four groups and four therapeutic sessions lasting 20 min. (1) Heat compression therapy session (HT, n = 10) (2) (CT, n = 10), (3) alternating (HCT, n = 10), and sham, control (ShT, n = 10). All participants had measurements of tissue perfusion (PU, [non-reference units]), muscle tension (T-[Hz]), elasticity (E-[arb- relative arbitrary unit]), and maximum isometric force (Fmax [kgf]) of the dominant hand at rest (Rest) after the muscle fatigue protocol (PostFat.5 min), after therapy (PostTh.5 min) and 24 h after therapy (PostTh.24 h). A two-way ANOVA with repeated measures: Group (ColdT, HeatT, ContrstT, ControlT) × Time (Rest, PostFat.5 min, PostTh.5 min, Post.24 h) was used to examine the changes in examined variables. Post-hoc tests with Bonferroni correction and ± 95% confidence intervals (CI) for absolute differences (△) were used to analyze the pairwise comparisons when a significant main effect or interaction was found. The ANOVA for PU, T, E, and Fmax revealed statistically significant interactions of Group by Time factors (p < 0.0001), as well as main effects for the Group factors (p < 0.0001; except for Fmax). In the PostTh.5 min. Period, significantly (p < 0.001) higher PU values were recorded in the HT (19.45 ± 0.91) and HCT (18.71 ± 0.67) groups compared to the ShT (9.79 ± 0.35) group (△ = 9.66 [8.75; 10.57 CI] > MDC(0.73), and △ = 8.92 [8.01; 9.83 CI] > MDC(0.73), respectively). Also, significantly (p < 0.001) lower values were recorded in the CT (3.69 ± 0.93) compared to the ShT (9.79 ± 0.35) group △ = 6.1 [5.19; 7.01 CI] > MDC(0.73). For muscle tone in the PostTh.5 m period significantly (p < 0.001) higher values were observed in the CT (20.08 ± 0.19 Hz) group compared to the HT (18.61 ± 0.21 Hz), HCT (18.95 ± 0.41 Hz) and ShT (19.28 ± 0.33 Hz) groups (respectively: △ = 1.47 [1.11; 1.83 CI] > MDC(0.845); △ = 1.13 [0.77; 1.49 CI] > MDC(0.845), and △ = 0.8 [0.44; 1.16 CI], < MDC(0.845)). The highest elasticity value in the PostTh.5 m period were observed in the CT (1.14 ± 0.07) group, and it was significantly higher than the values observed in the HT (0.97 ± 0.03, △ = 0.18 [0.11; 0.24 CI] > MDC(0.094), p < 0.001), HCT (0.90 ± 0.04, △ = 0.24 [0.17; 0.31 CI] > MDC(0.094), p < 0.001) and ShT (1.05 ± 0.07, △ = 0.094 [0.03; 0.16 CI] = MDC(0.094), p = 0.003) groups. For Fmax, there were no statistically significant differences between groups at any level of measurement. The results of the influence of the forearm of all three therapy forms on the muscles' biomechanical parameters confirmed their effectiveness. However, the effect size of alternating contrast therapy cannot be confirmed, especially in the PostTh24h period. Statistically significant changes were observed in favor of this therapy in PU and E measurements immediately after therapy (PostTh.5 min). Further research on contrast therapy is necessary.
Collapse
Affiliation(s)
- Robert Trybulski
- Provita Żory Medical Center, Żory, Poland.
- Department of Medical Sciences, The Wojciech Korfanty Upper Silesian Academy, Katowice, Poland.
| | - Adrian Kużdżał
- College of Medical Sciences, Institute of Health Sciences, University of Rzeszów, Rzeszów, Poland
| | - Arkadiusz Stanula
- Laboratory of Sport Performance Analysis, Institute of Sport Sciences, Academy of Physical Education in Katowice, Katowice, Poland
| | - Jarosław Muracki
- Institute of Physical Culture Sciences, Department of Physical Culture and Health, University of Szczecin, Szczecin, Poland
| | - Adam Kawczyński
- Department of Paralympic Sport, Wroclaw University of Health and Sport Sciences, Wrocław, Poland
| | - Wacław Kuczmik
- Department and Clinic of General Surgery, Vascular Surgery, Angiology and Phlebology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Hsing-Kuo Wang
- School and Graduate Institute of Physical Therapy, National Taiwan University, Taipei, Taiwan
- Center of Physical Therapy, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
2
|
Charrier M, Leroux I, Pichon J, Schleder C, Larcher T, Hamel A, Magot A, Péréon Y, Lamirault G, Tremblay JP, Skuk D, Rouger K. Human MuStem cells are competent to fuse with nonhuman primate myofibers in a clinically relevant transplantation context: A proof-of-concept study. J Neuropathol Exp Neurol 2024; 83:684-694. [PMID: 38752570 DOI: 10.1093/jnen/nlae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
Abstract
We previously reported that human muscle-derived stem cells (hMuStem cells) contribute to tissue repair after local administration into injured skeletal muscle or infarcted heart in immunodeficient rodent models. However, extrapolation of these findings to a clinical context is problematic owing to the considerable differences often seen between in vivo findings in humans versus rodents. Therefore, we investigated whether the muscle regenerative behavior of hMuStem cells is maintained in a clinically relevant transplantation context. Human MuStem cells were intramuscularly administered by high-density microinjection matrices into nonhuman primates receiving tacrolimus-based immunosuppression thereby reproducing the protocol that has so far produced the best results in clinical trials of cell therapy in myopathies. Four and 9 weeks after administration, histological analysis of cell injection sites revealed large numbers of hMuStem cell-derived nuclei in all cases. Most graft-derived nuclei were distributed in small myofiber groups in which no signs of a specific immune response were observed. Importantly, hMuStem cells contributed to simian tissue repair by fusing mainly with host myofibers, demonstrating their capacity for myofiber regeneration in this model. Together, these findings obtained in a valid preclinical model provide new insights supporting the potential of hMuStem cells in future cell therapies for muscle diseases.
Collapse
Affiliation(s)
- Marine Charrier
- Oniris, INRAE, PAnTher, Nantes, France
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
- Nantes Université, Nantes, France
| | | | | | | | | | - Antoine Hamel
- Service de Chirurgie Infantile, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France
| | - Armelle Magot
- Centre de Référence Maladies Neuromusculaires AOC, Filnemus, Euro-NMD, Laboratoire d'Explorations Fonctionnelles, Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - Yann Péréon
- Centre de Référence Maladies Neuromusculaires AOC, Filnemus, Euro-NMD, Laboratoire d'Explorations Fonctionnelles, Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | | | - Jacques P Tremblay
- Axe Neurosciences, Research Center of the CHU de Quebec-CHUL and Department of Molecular Medicine, School of Medicine, Laval University, Quebec, Quebec, Canada
| | - Daniel Skuk
- Axe Neurosciences, Research Center of the CHU de Quebec-CHUL and Department of Molecular Medicine, School of Medicine, Laval University, Quebec, Quebec, Canada
| | | |
Collapse
|
3
|
Pryor JL, Sweet D, Rosbrook P, Qiao J, Hess HW, Looney DP. Resistance Training in the Heat: Mechanisms of Hypertrophy and Performance Enhancement. J Strength Cond Res 2024; 38:1350-1357. [PMID: 38775794 DOI: 10.1519/jsc.0000000000004815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
ABSTRACT Pryor, JL, Sweet, D, Rosbrook, P, Qiao, J, Hess, HW, and Looney, DP. Resistance training in the heat: Mechanisms of hypertrophy and performance enhancement. J Strength Cond Res 38(7): 1350-1357, 2024-The addition of heat stress to resistance exercise or heated resistance exercise (HRE) is growing in popularity as emerging evidence indicates altered neuromuscular function and an amplification of several mechanistic targets of protein synthesis. Studies demonstrating increased protein synthesis activity have shown temperature-dependent mammalian target of rapamycin phosphorylation, supplemental calcium release, augmented heat shock protein expression, and altered immune and hormone activity. These intriguing observations have largely stemmed from myotube, isolated muscle fiber, or rodent models using passive heating alone or in combination with immobilization or injury models. A growing number of translational studies in humans show comparable results employing local tissue or whole-body heat with and without resistance exercise. While few, these translational studies are immensely valuable as they are most applicable to sport and exercise. As such, this brief narrative review aims to discuss evidence primarily from human HRE studies detailing the neuromuscular, hormonal, and molecular responses to HRE and subsequent strength and hypertrophy adaptations. Much remains unknown in this exciting new area of inquiry from both a mechanistic and functional perspective warranting continued research.
Collapse
Affiliation(s)
- J Luke Pryor
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York; and
| | - Daniel Sweet
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York; and
| | - Paul Rosbrook
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York; and
| | - JianBo Qiao
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York; and
| | - Hayden W Hess
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York; and
| | - David P Looney
- United States Army Research Institute of Environmental Medicine (USARIEM), Natick, Massachusetts
| |
Collapse
|
4
|
Halloran PF, Madill-Thomsen K, Mackova M, Aliabadi-Zuckermann AZ, Cadeiras M, Crespo-Leiro MG, Depasquale EC, Deng M, Gökler J, Hall SA, Kim DH, Kobashigawa J, Macdonald P, Potena L, Shah K, Stehlik J, Zuckermann A, Reeve J. Molecular states associated with dysfunction and graft loss in heart transplants. J Heart Lung Transplant 2024; 43:508-518. [PMID: 38042442 DOI: 10.1016/j.healun.2023.11.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/23/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023] Open
Abstract
BACKGROUND We explored the changes in gene expression correlating with dysfunction and graft failure in endomyocardial biopsies. METHODS Genome-wide microarrays (19,462 genes) were used to define mRNA changes correlating with dysfunction (left ventricular ejection fraction [LVEF] ≤ 55) and risk of graft loss within 3 years postbiopsy. LVEF data was available for 1,013 biopsies and survival data for 779 patients (74 losses). Molecular classifiers were built for predicting dysfunction (LVEF ≤ 55) and postbiopsy 3-year survival. RESULTS Dysfunction is correlated with dedifferentiation-decreased expression of normal heart transcripts, for example, solute carriers, along with increased expression of inflammation genes. Many genes with reduced expression in dysfunction were matrix genes such as fibulin 1 and decorin. Gene ontology (GO) categories suggested matrix remodeling and inflammation, not rejection. Genes associated with the risk of failure postbiopsy overlapped dysfunction genes but also included genes affecting microcirculation, for example, arginase 2, which reduces NO production, and endothelin 1. GO terms also reflected increased glycolysis and response to hypoxia, but decreased VEGF and angiogenesis pathways. T cell-mediated rejection was associated with reduced survival and antibody-mediated rejection with relatively good survival, but the main determinants of survival were features of parenchymal injury. Both dysfunction and graft loss were correlated with increased biopsy expression of BNP (gene NPPB). Survival probability classifiers divided hearts into risk quintiles, with actuarial 3-year postbiopsy survival >95% for the highest versus 50% for the lowest. CONCLUSIONS Dysfunction in transplanted hearts reflects dedifferentiation, decreased matrix genes, injury, and inflammation. The risk of short-term loss includes these changes but is also associated with microcirculation abnormalities, glycolysis, and response to hypoxia.
Collapse
Affiliation(s)
- Philip F Halloran
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| | | | - Martina Mackova
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | - Mario Deng
- Ronald Reagan UCLA Medical Center, Los Angeles, California
| | - Johannes Gökler
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Daniel H Kim
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | - Peter Macdonald
- The Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Luciano Potena
- Heart Failure and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Keyur Shah
- Department of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Josef Stehlik
- Department of Medicine, University of Utah, Salt Lake City, Utah
| | - Andreas Zuckermann
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Jeff Reeve
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Hassouneh R, Flynn S, Shen S, Tseng M, Bui AT, Pham J, Park D, Matherly S, Bruno D, Lee S, Kumaran V, Patel V, Muthiah M, Sharma A, Siddiqui MS. Impact of Liver Transplantation on Adipose Tissue Compartments and Its Association With Metabolic Sequela. Transplantation 2024; 108:235-241. [PMID: 37439776 DOI: 10.1097/tp.0000000000004704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
BACKGROUND Loss of skeletal muscle can be accompanied by an increase in adipose tissue leading to sarcopenic obesity. There are limited data on how liver transplantation (LT) might impact adipose tissue compartments, particularly among patients with metabolically active disease, such as nonalcoholic steatohepatitis (NASH) and subsequent metabolic sequela. METHODS Skeletal muscle, visceral adipose tissue (VAT), and subcutaneous adipose tissue (SAT) were measured using cross-sectional imaging performed in 190 patients pre-LT, 6 mo post-LT and 12 mo post-LT. Changes in adipose tissue and their impact on metabolic diseases were determined in patients transplanted for NASH versus non-NASH. RESULTS Skeletal muscle, VAT, and SAT were similar in patients with NASH and non-NASH pre-LT despite a higher burden of metabolic diseases in patients with NASH. Following LT, no significant differences between skeletal muscle and SAT were observed in the entire cohort and among patients with NASH (versus non-NASH). LT recipients with the highest muscle mass pre-LT were at the greatest risk for muscle loss post-LT. A time-dependent increase in VAT was noted post-LT, which was more robust among patients with a history of NASH cirrhosis. In adjusted multivariate analysis, NASH versus non-NASH was a strong predictor of post-LT increase in VAT (β-coefficient 3.00, P = 0.04). Pre-LT VAT was an independent predictor of post-LT serum triglycerides (β-coefficient 5.49 ± 2.78, P = 0.05) and low-density lipoprotein cholesterol (β-coefficient 1.80 ± 0.75, P = 0.02). A trend between pre-LT VAT and diabetes was noted but did not reach statistical significance. CONCLUSIONS VAT but not SAT increases rapidly after LT, especially among patients transplanted for NASH cirrhosis and predicts future metabolic burden.
Collapse
Affiliation(s)
- Ramzi Hassouneh
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
- Department of Gastroenterology, Indiana University, Indianapolis, IN
| | - Sean Flynn
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Steve Shen
- Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Michael Tseng
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Anh Tuan Bui
- Department of Statistical Sciences and Operations Research, Virginia Commonwealth University, Richmond, VA
| | - Jay Pham
- Department of Radiology, Virginia Commonwealth University, Richmond, VA
| | - Dan Park
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Scott Matherly
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, VA
| | - David Bruno
- Division of Transplant Surgery, Virginia Commonwealth University, Richmond, VA
| | - Seung Lee
- Division of Transplant Surgery, Virginia Commonwealth University, Richmond, VA
| | - Vinay Kumaran
- Division of Transplant Surgery, Virginia Commonwealth University, Richmond, VA
| | - Vaishali Patel
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, VA
| | - Mark Muthiah
- Department of Gastroenterology and Heptology, National University Hospital, Singapore
| | - Amit Sharma
- Division of Transplant Surgery, Virginia Commonwealth University, Richmond, VA
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
6
|
Roberts MD, McCarthy JJ, Hornberger TA, Phillips SM, Mackey AL, Nader GA, Boppart MD, Kavazis AN, Reidy PT, Ogasawara R, Libardi CA, Ugrinowitsch C, Booth FW, Esser KA. Mechanisms of mechanical overload-induced skeletal muscle hypertrophy: current understanding and future directions. Physiol Rev 2023; 103:2679-2757. [PMID: 37382939 PMCID: PMC10625844 DOI: 10.1152/physrev.00039.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.
Collapse
Affiliation(s)
- Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gustavo A Nader
- Department of Kinesiology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Andreas N Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States
| | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Riki Ogasawara
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Cleiton A Libardi
- MUSCULAB-Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Carlos Ugrinowitsch
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Karyn A Esser
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
7
|
Schaumburger N, Pally J, Moraru II, Kositsawat J, Kuchel GA, Blinov ML. Dynamic model assuming mutually inhibitory biomarkers of frailty suggests bistability with contrasting mobility phenotypes. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1079070. [PMID: 37216041 PMCID: PMC10192762 DOI: 10.3389/fnetp.2023.1079070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/17/2023] [Indexed: 05/24/2023]
Abstract
Bistability is a fundamental biological phenomenon associated with "switch-like" behavior reflecting the capacity of a system to exist in either of two stable states. It plays a role in gene regulation, cell fate switch, signal transduction and cell oscillation, with relevance for cognition, hearing, vision, sleep, gait and voiding. Here we consider a potential role for bistability in the existence of specific frailty states or phenotypes as part of disablement pathways. We use mathematical modeling with two frailty biomarkers (insulin growth factor-1, IGF-1 and interleukin-6, IL-6), which mutually inhibit each other. In our model, we demonstrate that small variations around critical IGF-1 or IL-6 blood levels lead to strikingly different mobility outcomes. We employ deterministic modeling of mobility outcomes, calculating the average trends in population health. Our model predicts the bistability of clinical outcomes: the deterministically-computed likelihood of an individual remaining mobile, becoming less mobile, or dying over time either increases to almost 100% or decreases to almost zero. Contrary to statistical models that attempt to estimate the likelihood of final outcomes based on probabilities and correlations, our model predicts functional outcomes over time based on specific hypothesized molecular mechanisms. Instead of estimating probabilities based on stochastic distributions and arbitrary priors, we deterministically simulate model outcomes over a wide range of physiological parameter values within experimentally derived boundaries. Our study is "a proof of principle" as it is based on a major assumption about mutual inhibition of pathways that is oversimplified. However, by making such an assumption, interesting effects can be described qualitatively. As our understanding of molecular mechanisms involved in aging deepens, we believe that such modeling will not only lead to more accurate predictions, but also help move the field from using mostly studies of associations to mechanistically guided approaches.
Collapse
Affiliation(s)
- Nathan Schaumburger
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT, United States
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT, United States
| | - Joel Pally
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT, United States
| | - Ion I. Moraru
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT, United States
| | | | - George A. Kuchel
- UConn Center on Aging, UConn Health, Farmington, CT, United States
| | - Michael L. Blinov
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT, United States
| |
Collapse
|
8
|
Hanada M, Hidaka M, Soyama A, Tanaka T, Hara T, Matsushima H, Haraguchi M, Kitamura M, Sekino M, Oikawa M, Nagura H, Takeuchi R, Sato S, Takahata H, Eguchi S, Kozu R. Association between hospital acquired disability and post-discharge mortality in patients after living donor liver transplantation. BMC Surg 2022; 22:445. [PMID: 36581830 PMCID: PMC9798581 DOI: 10.1186/s12893-022-01896-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Hospital-acquired disability (HAD) in patients who undergo living donor liver transplantation (LDLT) is expected to worsen physical functions due to inactivity during hospitalization. The aim of this study was to explore whether a decline in activities of daily living from hospital admission to discharge is associated with prognosis in LDLT patients, who once discharged from a hospital. METHODS We retrospectively examined the relationship between HAD and prognosis in 135 patients who underwent LDLT from June 2008 to June 2018, and discharged from hospital once. HAD was defined as a decline of over 5 points in the Barthel Index as an activity of daily living assessment. Additionally, LDLT patients were classified into four groups: low or high skeletal muscle index (SMI) and HAD or non-HAD. Univariate and multivariate Cox proportional hazard models were used to evaluate the association between HAD and survival. RESULTS HAD was identified in 47 LDLT patients (34.8%). The HAD group had a significantly higher all-cause mortality than the non-HAD group (log-rank: p < 0.001), and in the HAD/low SMI group, all-cause mortality was highest between the groups (log-rank: p < 0.001). In multivariable analysis, HAD was an independent risk factor for all-cause mortality (hazard ratio [HR]: 16.54; P < 0.001) and HAD/low SMI group (HR: 16.82; P = 0.002). CONCLUSION HAD was identified as an independent risk factor for all-cause mortality suggesting that it could be a key component in determining prognosis after LDLT. Future larger-scale studies are needed to consider the overall new strategy of perioperative rehabilitation, including enhancement of preoperative physiotherapy programs to improve physical function.
Collapse
Affiliation(s)
- Masatoshi Hanada
- grid.411873.80000 0004 0616 1585Cardiorespiratory Division, Department of Rehabilitation Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan ,grid.174567.60000 0000 8902 2273Department of Cardiopulmonary Rehabilitation Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masaaki Hidaka
- grid.174567.60000 0000 8902 2273Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akihiko Soyama
- grid.174567.60000 0000 8902 2273Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takayuki Tanaka
- grid.174567.60000 0000 8902 2273Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takanobu Hara
- grid.174567.60000 0000 8902 2273Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hajime Matsushima
- grid.174567.60000 0000 8902 2273Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masafumi Haraguchi
- grid.174567.60000 0000 8902 2273Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mineaki Kitamura
- grid.174567.60000 0000 8902 2273Department of Nephrology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Motohiro Sekino
- grid.411873.80000 0004 0616 1585Division of Intensive Care, Nagasaki University Hospital, Nagasaki, Japan
| | - Masato Oikawa
- grid.411873.80000 0004 0616 1585Cardiorespiratory Division, Department of Rehabilitation Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan ,grid.174567.60000 0000 8902 2273Department of Cardiopulmonary Rehabilitation Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroki Nagura
- grid.411873.80000 0004 0616 1585Cardiorespiratory Division, Department of Rehabilitation Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan ,grid.174567.60000 0000 8902 2273Department of Cardiopulmonary Rehabilitation Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Rina Takeuchi
- grid.411873.80000 0004 0616 1585Cardiorespiratory Division, Department of Rehabilitation Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Shuntaro Sato
- grid.411873.80000 0004 0616 1585Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Hideaki Takahata
- grid.411873.80000 0004 0616 1585Cardiorespiratory Division, Department of Rehabilitation Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Susumu Eguchi
- grid.174567.60000 0000 8902 2273Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ryo Kozu
- grid.411873.80000 0004 0616 1585Cardiorespiratory Division, Department of Rehabilitation Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan ,grid.174567.60000 0000 8902 2273Department of Cardiopulmonary Rehabilitation Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
9
|
Schmid S, Heim-Kupr B, Pérez-Schindler J, Mansingh S, Beer M, Mittal N, Ehrenfeuchter N, Handschin C. PGC-1β modulates catabolism and fiber atrophy in the fasting-response of specific skeletal muscle beds. Mol Metab 2022; 66:101643. [PMID: 36400401 PMCID: PMC9723918 DOI: 10.1016/j.molmet.2022.101643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Skeletal muscle is a pivotal organ for the coordination of systemic metabolism, constituting one of the largest storage site for glucose, lipids and amino acids. Tight temporal orchestration of protein breakdown in times of fasting has to be balanced with preservation of muscle mass and function. However, the molecular mechanisms that control the fasting response in muscle are poorly understood. METHODS We now have identified a role for the peroxisome proliferator-activated receptor γ coactivator 1β (PGC-1β) in the regulation of catabolic pathways in this context in muscle-specific loss-of-function mouse models. RESULTS Muscle-specific knockouts for PGC-1β experience mitigated muscle atrophy in fasting, linked to reduced expression of myostatin, atrogenes, activation of AMP-dependent protein kinase (AMPK) and other energy deprivation signaling pathways. At least in part, the muscle fasting response is modulated by a negative effect of PGC-1β on the nuclear factor of activated T-cells 1 (NFATC1). CONCLUSIONS Collectively, these data highlight the complex regulation of muscle metabolism and reveal a new role for muscle PGC-1β in the control of proteostasis in fasting.
Collapse
Affiliation(s)
- Svenia Schmid
- Biozentrum, University of Basel, Spitalstrasse 41, CH-4056 Basel, Switzerland
| | - Barbara Heim-Kupr
- Biozentrum, University of Basel, Spitalstrasse 41, CH-4056 Basel, Switzerland
| | | | - Shivani Mansingh
- Biozentrum, University of Basel, Spitalstrasse 41, CH-4056 Basel, Switzerland
| | - Markus Beer
- Biozentrum, University of Basel, Spitalstrasse 41, CH-4056 Basel, Switzerland
| | - Nitish Mittal
- Biozentrum, University of Basel, Spitalstrasse 41, CH-4056 Basel, Switzerland
| | | | - Christoph Handschin
- Biozentrum, University of Basel, Spitalstrasse 41, CH-4056 Basel, Switzerland.
| |
Collapse
|
10
|
Jaitovich A. Impaired regenerative capacity contributes to skeletal muscle dysfunction in chronic obstructive pulmonary disease. Am J Physiol Cell Physiol 2022; 323:C974-C989. [PMID: 35993519 PMCID: PMC9484993 DOI: 10.1152/ajpcell.00292.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 01/18/2023]
Abstract
Locomotor skeletal muscle dysfunction is a relevant comorbidity of chronic obstructive pulmonary disease (COPD) and is strongly associated with worse clinical outcomes including higher mortality. Over the last decades, a large body of literature helped characterize the process, defining the disruptive muscle phenotype caused by COPD that involves reduction in muscle mass, force-generation capacity, fatigue-tolerance, and regenerative potential following injury. A major limitation in the field has been the scarcity of well-calibrated animal models to conduct mechanistic research based on loss- and gain-of-function studies. This article provides an overall description of the process, the tools available to mechanistically investigate it, and the potential role of mitochondrially driven metabolic signals on the regulation muscle regeneration after injury in COPD. Finally, a description of future avenues to further expand on the area is proposed based on very recent evidence involving mitochondrial metabolic cues affecting myogenesis.
Collapse
Affiliation(s)
- Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
11
|
Alhajri N, Rustom M, Adegbile A, Ahmed W, Kilidar S, Afify N. Deciphering the Basis of Molecular Biology of Selected Cardiovascular Diseases: A View on Network Medicine. Int J Mol Sci 2022; 23:ijms231911421. [PMID: 36232723 PMCID: PMC9569471 DOI: 10.3390/ijms231911421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death across the world. For decades, researchers have been studying the causes of cardiovascular disease, yet many of them remain undiscovered or poorly understood. Network medicine is a recently expanding, integrative field that attempts to elucidate this issue by conceiving of disease as the result of disruptive links between multiple interconnected biological components. Still in its nascent stages, this revolutionary application of network science facilitated a number of important discoveries in complex disease mechanisms. As methodologies become more advanced, network medicine harbors the potential to expound on the molecular and genetic complexities of disease to differentiate how these intricacies govern disease manifestations, prognosis, and therapy. This is of paramount importance for confronting the incredible challenges of current and future cardiovascular disease research. In this review, we summarize the principal molecular and genetic mechanisms of common cardiac pathophysiologies as well as discuss the existing knowledge on therapeutic strategies to prevent, halt, or reverse these pathologies.
Collapse
Affiliation(s)
- Noora Alhajri
- Department of Internal Medicine, Cleveland Clinic Abu Dhabi (CCAD), Abu Dhabi P.O. Box 112412, United Arab Emirates
- Correspondence:
| | - Mohammad Rustom
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Adedayo Adegbile
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Weshah Ahmed
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Salsabeel Kilidar
- Department of Emergency Medicine, Sheikh Shakhbout Medical City SSMC, Abu Dhabi P.O. Box 11001, United Arab Emirates
| | - Nariman Afify
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
12
|
Miura E, Watanabe M. Coexpression of calcineurin A and B subunits in various subcellular and synaptic compartments of cerebellar neurons and glia with particular abundance at parallel fiber-Purkinje cell synapses. Neurosci Res 2022; 180:13-22. [PMID: 35247520 DOI: 10.1016/j.neures.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 02/21/2022] [Accepted: 02/27/2022] [Indexed: 10/19/2022]
Abstract
Calcineurin (CN) is a Ca2+/calmodulin-dependent serine/threonine protein phosphatase consisting of catalytic CNA and regulatory CNB subunits, and links activity-dependent Ca2+ signals to various neural functions. Here we studied CN expression in the mouse brain by producing subunit-specific probes and antibodies. Of five CN subunits. CNAα, CNAβ, and CNB1 mRNAs were predominantly expressed over the brain from early embryonic to adult stage, and all were high in the telencephalon and cerebellum. Protein localization was examined in the cerebellum by immunofluorescence with cellular and terminal markers and by preembedding silver-enhanced immunogold microscopy. CNB1 and CNAβ were co-distributed in subcellular and synaptic elements of various cerebellar neurons and glia, whereas CNAα was exclusive in granule cell elements, including parallel fiber terminals. The present study thus discloses that CNB1 subunit well coexists with one or two CNA subunits in various cerebellar compartments. Moreover, high CN contents are provided to parallel fiber-Purkinje cell synapses, i.e., CNAα, CNAβ, and CNB1 in their presynaptic side and CNAβ and CNB1 in their postsynaptic side. These findings will be the anatomical basis, at least partly, for the known regulatory roles of postsynaptic CNs in long-term depression and presynaptic CNs in transmitter release function.
Collapse
Affiliation(s)
- Eriko Miura
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan.
| |
Collapse
|
13
|
Chen C, Cui P, Zhao K, Niu G, Hou S, Zhao D, Zeng H. Down Syndrome Candidate Region 1 Isoform 1L regulated tumor growth by targeting both angiogenesis and tumor cells. Microvasc Res 2022; 140:104305. [PMID: 34958805 PMCID: PMC9295909 DOI: 10.1016/j.mvr.2021.104305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 11/26/2022]
Abstract
Angiogenesis is critical for solid tumor growth beyond its minimal size. Previously, we reported that Down Syndrome Candidate Region 1 isoform 1L (DSCR1-1L) was one of the most up-regulated genes in endothelial cells induced by VEGF and histamine, and regulated endothelial cell proliferation, migration and angiogenesis. However, it was not known whether DSCR1-1L played a role in tumor growth. In this study, we found that DSCR1-1L shRNAs significantly inhibited the growth of transplanted melanoma in mice and its associated tumoral angiogenesis. In the gain of function assay, overexpression of DSCR1-1L cDNA in mouse endothelium is sufficient to significantly increase the tumor initiation induced by carcinogen, the growth of xenografted tumor, and the tumor metastasis in our endothelially-expressed DSCR1-1L transgenic mice, in which angiogenesis was induced. It was the first time to find that DSCR1-1L was also expressed in various tumor cells. DSCR1-1L shRNAs inhibited, but overexpression of DSCR1-1L cDNA increased, the tumor cell proliferation and migration. Most recently, we reported that DSCR1-1L modulated angiogenesis by down-regulation of VE-cadherin expression. Here, we found that DSCR1-1L down-regulated the expression of E-cadherin. Hence, DSCR1-1L is an excellent therapeutic target for cancers by regulation of both the endothelial and tumor cells through down-regulating (V)E-cadherin. DSCR1-1L shRNAs have the potential to be developed for clinical application.
Collapse
Affiliation(s)
- Chen Chen
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Surgery of Breast and Thyroid, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Pengfei Cui
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Pancreatic Disease Institute, Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Kevin Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Gengming Niu
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shiqiang Hou
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
14
|
Attwaters M, Hughes SM. Cellular and molecular pathways controlling muscle size in response to exercise. FEBS J 2022; 289:1428-1456. [PMID: 33755332 DOI: 10.1111/febs.15820] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022]
Abstract
From the discovery of ATP and motor proteins to synaptic neurotransmitters and growth factor control of cell differentiation, skeletal muscle has provided an extreme model system in which to understand aspects of tissue function. Muscle is one of the few tissues that can undergo both increase and decrease in size during everyday life. Muscle size depends on its contractile activity, but the precise cellular and molecular pathway(s) by which the activity stimulus influences muscle size and strength remain unclear. Four correlates of muscle contraction could, in theory, regulate muscle growth: nerve-derived signals, cytoplasmic calcium dynamics, the rate of ATP consumption and physical force. Here, we summarise the evidence for and against each stimulus and what is known or remains unclear concerning their molecular signal transduction pathways and cellular effects. Skeletal muscle can grow in three ways, by generation of new syncytial fibres, addition of nuclei from muscle stem cells to existing fibres or increase in cytoplasmic volume/nucleus. Evidence suggests the latter two processes contribute to exercise-induced growth. Fibre growth requires increase in sarcolemmal surface area and cytoplasmic volume at different rates. It has long been known that high-force exercise is a particularly effective growth stimulus, but how this stimulus is sensed and drives coordinated growth that is appropriately scaled across organelles remains a mystery.
Collapse
Affiliation(s)
- Michael Attwaters
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| |
Collapse
|
15
|
Perisetti A, Goyal H, Yendala R, Chandan S, Tharian B, Thandassery RB. Sarcopenia in hepatocellular carcinoma: Current knowledge and future directions. World J Gastroenterol 2022; 28:432-448. [PMID: 35125828 PMCID: PMC8790553 DOI: 10.3748/wjg.v28.i4.432] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/29/2021] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is the second most occurring cancer worldwide and is one of the leading causes of cancer-related deaths. Hepatocellular carcinoma (HCC) is the most common (80%-90%) type among malignant liver cancers. Sarcopenia occurs very early in HCC and can predict and provide an opportunity to improve muscle health before engaging in the treatment options such as loco-regional, systemic, and transplant management. Multiple prognostic stating systems have been developed in HCC, such as Barcelona Clinic Liver Cancer, Child-Pugh score and Albumin-Bilirubin grade. However, the evaluation of patients' performance status is a major limitation of these scoring systems. In this review, we aim to summarize the current knowledge and recent advances about the role of sarcopenia in cirrhosis in general, while focusing specifically on HCC. Additionally, the role of sarcopenia in predicting clinical outcomes and prognostication in HCC patients undergoing loco-regional therapies, liver resection, liver transplantation and systematic therapy has been discussed. A literature review was performed using databases PubMed/MEDLINE, EMBASE, Cochrane, Web of Science, and CINAHL on April 1, 2021, to identify published reports on sarcopenia in HCC. Sarcopenia can independently predict HCC-related mortality especially in patients undergoing treatments such as loco-regional, surgical liver transplantation and systemic therapies. Basic research is focused on evaluating a balance of anabolic and catabolic pathways responsible for muscle health. Early clinical studies have shown promising results in methods to improve sarcopenia in HCC which can potentially increase prognosis in these patients. As sarcopenia occurs very early in HCC, it can predict and provide an opportunity to improve muscle health before engaging in the treatment options such as loco-regional, systemic, and transplant management. Further, sarcopenia measurement can obviate the confounding caused by the abdominal ascites in these patients. The use of sarcopenia can add to the existing scoring systems to better prognosticate the HCC.
Collapse
Affiliation(s)
- Abhilash Perisetti
- Department of Internal Medicine, Gastroenterology and Hepatology Division, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
- Department of Interventional Oncology and Surgical Endoscopy, Parkview Health, Fort Wayne, IN 46825, United States
| | - Hemant Goyal
- Department of Internal Medicine, The Wright Center for Graduate Medical Education, The Wright Center for Graduate Medical Education, Scranton, PA 18501, United States
| | - Rachana Yendala
- Department of Hematology and Oncology, Conway Regional Medical Center, Conway, AR 72034, United States
| | - Saurabh Chandan
- Department of Internal Medicine, Gastroenterology and Hepatology Division, CHI Creighton University Medical Center, Omaha, NE 68107, United States
| | - Benjamin Tharian
- Department of Internal Medicine, Gastroenterology and Hepatology Division, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Ragesh Babu Thandassery
- Department of Medicine, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, United States
| |
Collapse
|
16
|
Chambers PJ, Juracic ES, Fajardo VA, Tupling AR. The role of SERCA and sarcolipin in adaptive muscle remodeling. Am J Physiol Cell Physiol 2022; 322:C382-C394. [PMID: 35044855 DOI: 10.1152/ajpcell.00198.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sarcolipin (SLN) is a small integral membrane protein that regulates the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump. When bound to SERCA, SLN reduces the apparent Ca2+ affinity of SERCA and uncouples SERCA Ca2+ transport from its ATP consumption. As such, SLN plays a direct role in altering skeletal muscle relaxation and energy expenditure. Interestingly, the expression of SLN is dynamic during times of muscle adaptation, where large increases in SLN content are found in response to development, atrophy, overload and disease. Several groups have suggested that increases in SLN, especially in dystrophic muscle, are deleterious to muscle function and exacerbate already abhorrent intracellular Ca2+ levels. However, there is also significant evidence to show that increased SLN content is a beneficial adaptive mechanism which protects the SERCA pump and activates Ca2+ signaling and adaptive remodeling during times of cell stress. In this review, we first discuss the role for SLN in healthy muscle during both development and overload, where SLN has been shown to activate Ca2+ signaling to promote mitochondrial biogenesis, fibre type shifts and muscle hypertrophy. Then, with respect to muscle disease, we summarize the discrepancies in the literature as to whether SLN upregulation is adaptive or maladaptive in nature. This review is the first to offer the concept of SLN hormesis in muscle disease, wherein both too much and too little SLN are detrimental to muscle health. Finally, the underlying mechanisms which activate SLN upregulation are discussed, specifically acknowledging a potential positive feedback loop between SLN and Ca2+ signaling molecules.
Collapse
Affiliation(s)
- Paige J Chambers
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Emma S Juracic
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Val A Fajardo
- Department Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - A Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
17
|
Hou S, Niu G, Liu X, Bourbon PM, Zhang D, Cui P, Zhao K, Zhao D, Zeng H. A novel transcriptional complex on the VE-cadherin promoter regulated the downregulation of VE-cadherin in the Down Syndrome Candidate Region 1 isoform 1L-mediated angiogenesis. Microvasc Res 2021; 138:104209. [PMID: 34146582 PMCID: PMC9295908 DOI: 10.1016/j.mvr.2021.104209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/18/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022]
Abstract
Angiogenesis is critical for many diseases. Previously, we reported that Down Syndrome Candidate Region 1 isoform 1L (DSCR1-1L) was one of the most up-regulated genes in endothelial cells induced by VEGF and histamine, and regulated endothelial cell proliferation and Matrigel angiogenesis in mice. However, it was not known whether DSCR1-1L regulated angiogenesis in vivo and what was the molecular mechanism underlying it. In this study, gene knockdown and overexpression models were established to study the role of DSCR1-1L in angiogenesis in vivo. Further, the downstream regulatory target of DSCR1-1L was explored with molecular biological methods in vascular endothelial cells. We found that DSCR1-1L shRNAs significantly inhibited angiogenesis induced by VEGF in mice (p < 0.0001). In the gain-of-function assay, overexpression of DSCR1-1L cDNA in mouse endothelium of EC-FH-DSCR1-1L transgenic mice was sufficient to induce angiogenesis significantly (p < 0.01). DSCR1-1L regulated angiogenesis in the early stage by down-regulation of the VE-cadherin expression through targeting its transcription, but not mRNA stability. Three DSCR1-1L-targeted DNA elements in the VE-cadherin promoter were identified by promoter reporter assays, among which, a novel specific transcriptional complex was found. The DNA sequence (CTTCTG) in the VE-cadherin promoter was identified to directly interact with proteins by Electrophoresis Mobility Shift Assays and DNase I footprint assay. Hence, DSCR1-1L is an excellent therapeutic target for angiogenic diseases through down-regulating the formation of a novel transcriptional complex on the VE-cadherin promoter. DSCR1-1L shRNAs and cDNA have the potential to be developed for clinical application. Our results also contribute significantly to the field of mechanistic studies.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Down-Regulation
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/genetics
- Melanoma, Experimental/metabolism
- Mice, Nude
- Mice, Transgenic
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- Promoter Regions, Genetic
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Shiqiang Hou
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Gengming Niu
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Xin Liu
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pierre M Bourbon
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dongmei Zhang
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Key Laboratory of Chinese Internal Medicine, Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Pengfei Cui
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Pancreatic Disease Institute, Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Kevin Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
18
|
Borg ML, Massart J, De Castro Barbosa T, Archilla-Ortega A, Smith JAB, Lanner JT, Alsina-Fernandez J, Yaden B, Culver AE, Karlsson HKR, Brozinick JT, Zierath JR. Modified UCN2 peptide treatment improves skeletal muscle mass and function in mouse models of obesity-induced insulin resistance. J Cachexia Sarcopenia Muscle 2021; 12:1232-1248. [PMID: 34342159 PMCID: PMC8517345 DOI: 10.1002/jcsm.12746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/25/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Type 2 diabetes and obesity are often seen concurrently with skeletal muscle wasting, leading to further derangements in function and metabolism. Muscle wasting remains an unmet need for metabolic disease, and new approaches are warranted. The neuropeptide urocortin 2 (UCN2) and its receptor corticotropin releasing factor receptor 2 (CRHR2) are highly expressed in skeletal muscle and play a role in regulating energy balance, glucose metabolism, and muscle mass. The aim of this study was to investigate the effects of modified UCN2 peptides as a pharmaceutical therapy to counteract the loss of skeletal muscle mass associated with obesity and casting immobilization. METHODS High-fat-fed mice (C57Bl/6J; 26 weeks old) and ob/ob mice (11 weeks old) were injected daily with a PEGylated (Compound A) and non-PEGylated (Compound B) modified human UCN2 at 0.3 mg/kg subcutaneously for 14 days. A separate group of chow-fed C57Bl/6J mice (12 weeks old) was subjected to hindlimb cast immobilization and, after 1 week, received daily injections with Compound A. In vivo functional tests were performed to measure protein synthesis rates and skeletal muscle function. Ex vivo functional and molecular tests were performed to measure contractile force and signal transduction of catabolic and anabolic pathways in skeletal muscle. RESULTS Skeletal muscles (extensor digitorum longus, soleus, and tibialis anterior) from high-fat-fed mice treated with Compound A were ~14% heavier than muscles from vehicle-treated mice. Chronic treatment with modified UCN2 peptides altered the expression of structural genes and transcription factors in skeletal muscle in high-fat diet-induced obesity including down-regulation of Trim63 and up-regulation of Nr4a2 and Igf1 (P < 0.05 vs. vehicle). Signal transduction via both catabolic and anabolic pathways was increased in tibialis anterior muscle, with increased phosphorylation of ribosomal protein S6 at Ser235/236 , FOXO1 at Ser256 , and ULK1 at Ser317 , suggesting that UCN2 treatment modulates protein synthesis and degradation pathways (P < 0.05 vs. vehicle). Acutely, a single injection of Compound A in drug-naïve mice had no effect on the rate of protein synthesis in skeletal muscle, as measured via the surface sensing of translation method, while the expression of Nr4a3 and Ppargc1a4 was increased (P < 0.05 vs. vehicle). Compound A treatment prevented the loss of force production from disuse due to casting. Compound B treatment increased time to fatigue during ex vivo contractions of fast-twitch extensor digitorum longus muscle. Compound A and B treatment increased lean mass and rates of skeletal muscle protein synthesis in ob/ob mice. CONCLUSIONS Modified human UCN2 is a pharmacological candidate for the prevention of the loss of skeletal muscle mass associated with obesity and immobilization.
Collapse
Affiliation(s)
- Melissa L Borg
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Julie Massart
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Thais De Castro Barbosa
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Adrià Archilla-Ortega
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Jonathon A B Smith
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Section for Molecular Muscle Physiology and Pathophysiology, Karolinska Institutet, Stockholm, Sweden
| | | | - Benjamin Yaden
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Alexander E Culver
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Håkan K R Karlsson
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Joseph T Brozinick
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, IN, USA
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
Guilherme JPLF, V Shikhova Y, R Dondukovskaya R, A Topanova A, A Semenova E, V Astratenkova I, Ahmetov II. Androgen receptor gene microsatellite polymorphism is associated with muscle mass and strength in bodybuilders and power athlete status. Ann Hum Biol 2021; 48:142-149. [PMID: 33900145 DOI: 10.1080/03014460.2021.1919204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The androgen receptor (AR) gene contains a polymorphic trinucleotide (CAG) microsatellite repeat sequence (short or long alleles) that has been associated with fat-free mass in untrained men, which needs to be replicated in athletic cohorts. AIM The purpose of this study was to explore the AR (CAG)n polymorphism in trained individuals. SUBJECTS AND METHODS A total of 61 professional bodybuilders (40 males, 21 females), 73 elite male sprinters and weightlifters and 186 male controls were enrolled in this study. The influence of the AR (CAG)n polymorphism on muscle mass and strength was assessed in bodybuilders, while the frequencies of AR (CAG)n alleles were compared between power athletes and non-athletes. RESULTS The polymorphism was associated with anthropometric and strength measurements in bodybuilders of both genders. Those with ≥21 CAG repeats (i.e. carriers of long alleles) exhibited greater (p < 0.05) body mass index, absolute muscle mass, arm/thigh circumference and upper/lower limb strength compared to those with <21 CAG repeats. Furthermore, carriers of ≥21 CAG repeats were more frequent among power athletes compared to controls (p = 0.0076). CONCLUSIONS Long alleles of the AR (CAG)n polymorphism were associated with greater muscle mass and strength in bodybuilders, and power athlete status.
Collapse
Affiliation(s)
- João Paulo L F Guilherme
- Laboratory of Applied Nutrition and Metabolism, School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Yulia V Shikhova
- Sports Genetics Laboratory, St Petersburg Research Institute of Physical Culture, St Petersburg, Russia
| | - Rimma R Dondukovskaya
- Sports Genetics Laboratory, St Petersburg Research Institute of Physical Culture, St Petersburg, Russia.,Weider College of Fitness and Bodybuilding, St Petersburg, Russia
| | - Alexandra A Topanova
- Sports Genetics Laboratory, St Petersburg Research Institute of Physical Culture, St Petersburg, Russia.,Institute of Medical Education, Almazov National Medical Research Centre, St Petersburg, Russia
| | - Ekaterina A Semenova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.,Sport Technology Research Center, Volga Region State University of Physical Culture, Sport and Tourism, Kazan, Russia
| | - Irina V Astratenkova
- Sports Genetics Laboratory, St Petersburg Research Institute of Physical Culture, St Petersburg, Russia.,Department of Physiology, St Petersburg State University, St Petersburg, Russia
| | - Ildus I Ahmetov
- Sports Genetics Laboratory, St Petersburg Research Institute of Physical Culture, St Petersburg, Russia.,Department of Physical Education, Plekhanov Russian University of Economics, Moscow, Russia.,Laboratory of Molecular Genetics, Kazan State Medical University, Kazan, Russia.,Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
20
|
Duong N, Sadowski B, Rangnekar AS. The Impact of Frailty, Sarcopenia, and Malnutrition on Liver Transplant Outcomes. Clin Liver Dis (Hoboken) 2021; 17:271-276. [PMID: 33968388 PMCID: PMC8087926 DOI: 10.1002/cld.1043] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/13/2020] [Accepted: 09/20/2020] [Indexed: 02/04/2023] Open
Affiliation(s)
- Nikki Duong
- Department of Internal MedicineMedStar Georgetown University HospitalWashingtonDC
- Division of Gastroenterology, Hepatology, and NutritionVirginia Commonwealth UniversityRichmondVA
- MedStar Georgetown Transplant InstituteGeorgetown University HospitalWashingtonDC
| | - Brett Sadowski
- MedStar Georgetown Transplant InstituteGeorgetown University HospitalWashingtonDC
| | - Amol S. Rangnekar
- MedStar Georgetown Transplant InstituteGeorgetown University HospitalWashingtonDC
| |
Collapse
|
21
|
Bkaily G, Abou Abdallah N, Simon Y, Jazzar A, Jacques D. Vascular smooth muscle remodeling in health and disease. Can J Physiol Pharmacol 2021; 99:171-178. [PMID: 32853532 DOI: 10.1139/cjpp-2020-0399] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In blood vessels, vascular smooth muscle cells (VSMCs) generally exist in two major phenotypes: contractile and non-contractile (synthetic). The contractile phenotype is predominant and includes quiescent or differentiated VSMCs, which function as the regulators of blood vessel diameter and blood flow. According to some literature in the field, contractile VSMCs do not switch to the non-contractile phenotype due to the activation of specific transcription factors that are considered as guardians of the contractile phenotype. However, a vast amount of the literature uses the terms remodeling and phenotype switching of contractile VSMCs interchangeably based mainly on studies dealing with atherosclerosis. The use of the terms remodeling and switching to describe changes in phenotype based on morphological criteria can be confusing. The term remodeling was first used to describe morphological changes in the heart and was soon used to describe phenotype changes of contractile VSMCs based on morphological criteria. The latter were introduced in early studies, and new molecular criteria were later added, including changes in gene expression, which could be irreversible. In this review, we will discuss the different views concerning remodeling and possible switching of contractile VSMCs to a non-contractile phenotype. We conclude that only remodeling of contractile VSMCs may take place upon vascular injury and disease.
Collapse
Affiliation(s)
- Ghassan Bkaily
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
| | - Nadia Abou Abdallah
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
| | - Yanick Simon
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
| | - Ashley Jazzar
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
| | - Danielle Jacques
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada J1H 5N4
| |
Collapse
|
22
|
Ansari MA, Khan FB, Safdari HA, Almatroudi A, Alzohairy MA, Safdari M, Amirizadeh M, Rehman S, Equbal MJ, Hoque M. Prospective therapeutic potential of Tanshinone IIA: An updated overview. Pharmacol Res 2020; 164:105364. [PMID: 33285229 DOI: 10.1016/j.phrs.2020.105364] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/29/2020] [Accepted: 11/29/2020] [Indexed: 01/03/2023]
Abstract
In the past decades, the branch of complementary and alternative medicine based therapeutics has gained considerable attention worldwide. Pharmacological efficacy of various traditional medicinal plants, their products and/or product derivatives have been explored on an increasing scale. Tanshinone IIA (Tan IIA) is a pharmacologically active lipophilic component of Salvia miltiorrhiza extract. Tan IIA shares a history of high repute in Traditional Chinese Medicine. Reckoning with these, the present review collates the pharmacological properties of Tan IIA with a special emphasis on its therapeutic potential against diverse diseases including cardiovascular diseases, cerebrovascular diseases, cancer, diabetes, obesity and neurogenerative diseases. Further, possible applications of various therapeutic preparations of Tan IIA were discussed with special emphasis on nano-based drug delivery formulations. Considering the tremendous advancement in the field of nanomedicine and the therapeutic potential of Tan IIA, the convergence of these two aspects can be foreseen with great promise in clinical application.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institutes for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1881, Dammam 31441, Saudi Arabia
| | - Farheen Badrealam Khan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Haaris Ahsan Safdari
- New Technology Center, University of Warsaw, Stefana Banacha 2c, 02-097 Warszawa, Poland
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Qassim 51431, Saudi Arabia
| | - Mohammad A Alzohairy
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Qassim 51431, Saudi Arabia
| | - Mohammadreza Safdari
- Imam Ali Hospital, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehran Amirizadeh
- Department of Pharmacotherapy, Faculty of Pharmacy, University of Medical Sciences, Khorramabad, Lorestan, Iran
| | - Suriya Rehman
- Department of Epidemic Disease Research, Institutes for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1881, Dammam 31441, Saudi Arabia
| | - Mohammad Javed Equbal
- Biomedical Institute for Regenerative Research, Texas A&M University Commerce, Commerce, TX 75429, United States.
| | - Mehboob Hoque
- Department of Biological Sciences, Aliah University, Kolkata 700 160, India.
| |
Collapse
|
23
|
Cisterna B, Sobolev AP, Costanzo M, Malatesta M, Zancanaro C. Combined Microscopic and Metabolomic Approach to Characterize the Skeletal Muscle Fiber of the Ts65Dn Mouse, A Model of Down Syndrome. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2020; 26:1014-1023. [PMID: 32867866 DOI: 10.1017/s143192762002437x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Down syndrome (DS) is a genetically based disease caused by triplication of chromosome 21. DS is characterized by severe muscle weakness associated with motor deficits; however, understanding the DS-associated skeletal muscle condition is limited. In this study, we used a combined methodological approach involving light and electron microscopy, as well as nuclear magnetic resonance spectroscopy metabolomics, to investigate morphology and composition of the quadriceps muscles in the Ts65Dn mouse, a model of DS, to identify structural and/or functional trisomy-associated alterations. Morphometric analysis demonstrated a larger size of myofibers in trisomic versus euploid mice; however, myofibrils were thinner and contained higher amounts of mitochondria and lipid droplets. In trisomic mice, magnetic resonance spectroscopy showed a tendency to an overall increase in muscle metabolites involved in protein synthesis. These data strongly suggest that in DS, a sarcoplasmic hypertrophy associated with myofibril loss characterizes quadriceps myofibers. In addition, large-sized mitochondria suggestive of impaired fission/fusion events, as well as metabolites modifications suggestive of decreased mitochondrial function, were found in the trisomic muscle. Albeit preliminary, the results provided by this novel approach consistently indicate structural and compositional alterations of the DS skeletal muscle, which are typical of early aging.
Collapse
Affiliation(s)
- Barbara Cisterna
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, VeronaI-37134, Italy
| | - Anatoly P Sobolev
- Magnetic Resonance Laboratory "Annalaura Segre", Institute for Biological Systems, National Research Council, via Salaria km 29.300, Monterotondo, RomeI-00015, Italy
| | - Manuela Costanzo
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, VeronaI-37134, Italy
| | - Manuela Malatesta
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, VeronaI-37134, Italy
| | - Carlo Zancanaro
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, VeronaI-37134, Italy
| |
Collapse
|
24
|
Marasco G, Serenari M, Renzulli M, Alemanni LV, Rossini B, Pettinari I, Dajti E, Ravaioli F, Golfieri R, Cescon M, Festi D, Colecchia A. Clinical impact of sarcopenia assessment in patients with hepatocellular carcinoma undergoing treatments. J Gastroenterol 2020; 55:927-943. [PMID: 32748172 PMCID: PMC7519899 DOI: 10.1007/s00535-020-01711-w] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/15/2020] [Indexed: 02/04/2023]
Abstract
Changes in body composition are associated with poor outcomes in cancer patients including hepatocellular carcinoma (HCC). Sarcopenia, defined as the loss of skeletal muscle mass, quality and function, has been associated with a higher rate of complications and recurrences in patients with cirrhosis and HCC. The assessment of patient general status before HCC treatment, including the presence of sarcopenia, is a key-point for achieving therapy tolerability and to avoid short- and long-term complications leading to poor patients' survival. Thus, we aimed to review the current literature evaluating the role of sarcopenia assessment related to HCC treatments and to critically provide the clinicians with the most recent and valuable evidence. As a result, sarcopenia can be predictive of poor outcomes in patients undergoing liver resection, transplantation and systemic therapies, offering the chance to clinicians to improve the muscular status of these patients, especially those with high-grade sarcopenia at high risk of mortality. Further studies are needed to clarify the predictive value of sarcopenia in other HCC treatment settings and to evaluate its role as an additional staging tool for identifying the most appropriate treatment. Besides, interventional studies aiming at increasing the skeletal muscle mass for reducing complications and increasing the survival in patients with HCC are needed.
Collapse
Affiliation(s)
- Giovanni Marasco
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40126, Bologna, Italy.
| | - Matteo Serenari
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40126, Bologna, Italy
| | - Matteo Renzulli
- Radiology Unit, Sant'Orsola Malpighi Hospital, Via Albertoni 4, 40138, Bologna, Italy
| | - Luigina Vanessa Alemanni
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40126, Bologna, Italy
| | - Benedetta Rossini
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40126, Bologna, Italy
| | - Irene Pettinari
- Radiology Unit, Sant'Orsola Malpighi Hospital, Via Albertoni 4, 40138, Bologna, Italy
| | - Elton Dajti
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40126, Bologna, Italy
| | - Federico Ravaioli
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40126, Bologna, Italy
| | - Rita Golfieri
- Radiology Unit, Sant'Orsola Malpighi Hospital, Via Albertoni 4, 40138, Bologna, Italy
| | - Matteo Cescon
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40126, Bologna, Italy
| | - Davide Festi
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 9, 40126, Bologna, Italy
| | - Antonio Colecchia
- Gastroenterology Unit, University Hospital Borgo Trento, Verona, Italy
| |
Collapse
|
25
|
Le Bacquer O, Lanchais K, Combe K, Van Den Berghe L, Walrand S. Acute rimonabant treatment promotes protein synthesis in C2C12 myotubes through a CB1-independent mechanism. J Cell Physiol 2020; 236:2669-2683. [PMID: 32885412 DOI: 10.1002/jcp.30034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/06/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022]
Abstract
Sarcopenia is an age-related loss of muscle mass associated with changes in skeletal muscle protein homeostasis due to lipid accumulation and anabolic resistance; changes that are also commonly described in obesity. Activation of the endocannabinoid system is associated with the development of obesity and insulin resistance, and with the perturbed skeletal muscle development. Taken together this suggests that endocannabinoids could be regulators of skeletal muscle protein homeostasis. Here we report that rimonabant, an antagonist for the CB1 receptor, can prevent dexamethasone-induced C2C12 myotube atrophy without affecting the mRNA expression of atrogin-1/MAFbx (a marker of proteolysis), which suggests it is involved in the control of protein synthesis. Rimonabant alone stimulates protein synthesis in a time- and dose-dependent manner through mTOR- and intracellular calcium-dependent mechanisms. CB1 agonists are unable to modulate protein synthesis or prevent the effect of rimonabant. Using C2C12 cells stably expressing an shRNA directed against CB1, or HEK293 cells overexpressing HA-tagged CB1, we demonstrated that the effect of rimonabant is unaffected by CB1 expression level. In summary, rimonabant can stimulate protein synthesis in C2C12 myotubes through a CB1-independent mechanism. These results highlight the need to identify non-CB1 receptor(s) mediating the pro-anabolic effect of rimonabant as potential targets for the treatment of sarcopenia, and to design new side-effect-free molecules that consolidate the effect of rimonabant on skeletal muscle protein synthesis.
Collapse
Affiliation(s)
- Olivier Le Bacquer
- Université Clermont Auvergne, INRAE, Unité de Nutrition Humaine (UNH), Clermont-Ferrand, France
| | - Kassandra Lanchais
- Université Clermont Auvergne, INRAE, Unité de Nutrition Humaine (UNH), Clermont-Ferrand, France
| | - Kristell Combe
- Université Clermont Auvergne, INRAE, Unité de Nutrition Humaine (UNH), Clermont-Ferrand, France
| | | | - Stéphane Walrand
- Université Clermont Auvergne, INRAE, Unité de Nutrition Humaine (UNH), Clermont-Ferrand, France.,CHU Clermont-Ferrand, Service de Nutrition Clinique, Hôpital Gabriel Montpied, Clermont-Ferrand, France
| |
Collapse
|
26
|
Regulation of the Mammalian SWI/SNF Family of Chromatin Remodeling Enzymes by Phosphorylation during Myogenesis. BIOLOGY 2020; 9:biology9070152. [PMID: 32635263 PMCID: PMC7407365 DOI: 10.3390/biology9070152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 11/16/2022]
Abstract
Myogenesis is the biological process by which skeletal muscle tissue forms. Regulation of myogenesis involves a variety of conventional, epigenetic, and epigenomic mechanisms that control chromatin remodeling, DNA methylation, histone modification, and activation of transcription factors. Chromatin remodeling enzymes utilize ATP hydrolysis to alter nucleosome structure and/or positioning. The mammalian SWItch/Sucrose Non-Fermentable (mSWI/SNF) family of chromatin remodeling enzymes is essential for myogenesis. Here we review diverse and novel mechanisms of regulation of mSWI/SNF enzymes by kinases and phosphatases. The integration of classic signaling pathways with chromatin remodeling enzyme function impacts myoblast viability and proliferation as well as differentiation. Regulated processes include the assembly of the mSWI/SNF enzyme complex, choice of subunits to be incorporated into the complex, and sub-nuclear localization of enzyme subunits. Together these processes influence the chromatin remodeling and gene expression events that control myoblast function and the induction of tissue-specific genes during differentiation.
Collapse
|
27
|
Growth hormone increases regulator of calcineurin 1-4 (Rcan1-4) mRNA through c-JUN in rat liver. PLoS One 2020; 15:e0235270. [PMID: 32589657 PMCID: PMC7319343 DOI: 10.1371/journal.pone.0235270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/11/2020] [Indexed: 11/23/2022] Open
Abstract
Growth hormone (GH) activates multiple signal transduction pathways. To investigate these pathways, we identified novel genes whose transcription was induced by GH in the liver of hypophysectomized (HPX) rats using the suppression subtractive hybridization technique. We found that regulator of calcineurin 1 (Rcan1) mRNA was upregulated by GH administration. RCAN1 regulates the activity of calcineurin, a Ca/calmodulin-dependent phosphatase. Rcan1 encodes two major transcripts, Rcan1-1 and Rcan1-4, resulting from differential promoter use and first exon choice. We found that a single injection of GH increased the levels of Rcan1-4 mRNA and RCAN1-4 protein transiently, but did not increase Rcan1-1 mRNA in HPX rat liver. Then the molecular mechanism of GH to induce Rcan1-4 transcription was examined in rat hepatoma H4IIE cells. Experiments using inhibitors suggested that c-JUN N-terminal kinase was required for the induction of Rcan1-4 mRNA by GH. GH increased the levels of phosphorylated c-JUN protein and c-Jun mRNA in HPX rat liver. The luciferase and electrophoretic mobility shift assays showed that c-JUN upregulated Rcan1-4 mRNA by binding to the cAMP-responsive element in the upstream of Rcan1 exon 4. These results indicate that GH activates c-JUN to affect the activity of calcineurin by the induction of Rcan1-4 in rat liver.
Collapse
|
28
|
Kim K, Monroe JC, Gavin TP, Roseguini BT. Skeletal muscle adaptations to heat therapy. J Appl Physiol (1985) 2020; 128:1635-1642. [PMID: 32352340 DOI: 10.1152/japplphysiol.00061.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The therapeutic effects of heat have been harnessed for centuries to treat skeletal muscle disorders and other pathologies. However, the fundamental mechanisms underlying the well-documented clinical benefits associated with heat therapy (HT) remain poorly defined. Foundational studies in cultured skeletal muscle and endothelial cells, as well as in rodents, revealed that episodic exposure to heat stress activates a number of intracellular signaling networks and promotes skeletal muscle remodeling. Renewed interest in the physiology of HT in recent years has provided greater understanding of the signals and molecular players involved in the skeletal muscle adaptations to episodic exposures to HT. It is increasingly clear that heat stress promotes signaling mechanisms involved in angiogenesis, muscle hypertrophy, mitochondrial biogenesis, and glucose metabolism through not only elevations in tissue temperature but also other perturbations, including increased intramyocellular calcium and enhanced energy turnover. The few available translational studies seem to indicate that the earlier observations in rodents also apply to human skeletal muscle. Indeed, recent findings revealed that both local and whole-body HT may promote capillary growth, enhance mitochondrial content and function, improve insulin sensitivity and attenuate disuse-induced muscle wasting. This accumulating body of work implies that HT may be a practical treatment to combat skeletal abnormalities in individuals with chronic disease who are unwilling or cannot participate in traditional exercise-training regimens.
Collapse
Affiliation(s)
- Kyoungrae Kim
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana
| | - Jacob C Monroe
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana
| | - Timothy P Gavin
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana
| | - Bruno T Roseguini
- Department of Health and Kinesiology, Purdue University, West Lafayette, Indiana
| |
Collapse
|
29
|
Pierce JR, Martin BJ, Rarick KR, Alemany JA, Staab JS, Kraemer WJ, Hymer WC, Nindl BC. Growth Hormone and Insulin-like Growth Factor-I Molecular Weight Isoform Responses to Resistance Exercise Are Sex-Dependent. Front Endocrinol (Lausanne) 2020; 11:571. [PMID: 32973684 PMCID: PMC7472848 DOI: 10.3389/fendo.2020.00571] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 07/13/2020] [Indexed: 11/25/2022] Open
Abstract
Purpose: To determine if acute resistance exercise-induced increases in growth hormone (GH) and insulin-like growth factor-I (IGF-I) were differentially responsive for one or more molecular weight (MW) isoforms and if these responses were sex-dependent. Methods: College-aged men (n = 10) and women (n = 10) performed an acute resistance exercise test (ARET; 6 sets, 10 repetition maximum (10-RM) squat, 2-min inter-set rest). Serum aliquots from blood drawn Pre-, Mid-, and Post-ARET (0, +15, and +30-min post) were processed using High Performance Liquid Chromatography (HPLC) fractionation and pooled into 3 MW fractions (Fr.A: >60; Fr.B: 30-60; Fr.C: <30 kDa). Results: We observed a hierarchy of serum protein collected among GH fractions across all time points independent of sex (Fr.C > Fr.A > Fr.B, p ≤ 0.03). Sex × time interactions indicated that women experienced earlier and augmented increases in all serum GH MW isoform fraction pools (p < 0.05); however, men demonstrated delayed and sustained GH elevations (p < 0.01) in all fractions through +30-min of recovery. Similarly, we observed a sex-independent hierarchy among IGF-I MW fraction pools (Fr.A > Fr.B > Fr.C, p ≤ 0.01). Furthermore, we observed increases in IGF-I Fr. A (ternary complexes) in men only (p ≤ 0.05), and increases in Fr.C (free/unbound IGF-I) in women only (p ≤ 0.05) vs. baseline, respectively. Conclusions: These data indicate that the processing of GH and IGF-I isoforms from the somatotrophs and hepatocytes are differential in their response to strenuous resistance exercise and reflect both temporal and sex-related differences.
Collapse
Affiliation(s)
- Joseph R. Pierce
- Military Performance Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
- *Correspondence: Joseph R. Pierce
| | - Brian J. Martin
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kevin R. Rarick
- Military Performance Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Joseph A. Alemany
- Military Performance Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Jeffery S. Staab
- Military Performance Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - William J. Kraemer
- Department of Kinesiology, University of Connecticut, Mansfield, CT, United States
| | - Wesley C. Hymer
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, United States
| | - Bradley C. Nindl
- Military Performance Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
30
|
Bunchorntavakul C, Reddy KR. Review article: malnutrition/sarcopenia and frailty in patients with cirrhosis. Aliment Pharmacol Ther 2020; 51:64-77. [PMID: 31701570 DOI: 10.1111/apt.15571] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/20/2019] [Accepted: 10/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Malnutrition/sarcopenia and frailty are common in patients with cirrhosis and are associated with poor outcomes. AIM To provide an overview of data on the importance, assessment and management of malnutrition/sarcopenia and frailty in cirrhosis. METHODS A literature search was conducted in PubMed and other sources, using the search terms "sarcopenia," "muscle," "malnutrition," "cirrhosis," "liver" and "frailty" from inception to April 2019, to identify the relevant studies and international guidelines. RESULTS The prevalence of malnutrition/sarcopenia in cirrhosis is 23%-60%. Frailty generally overlaps with malnutrition/sarcopenia in cirrhosis, leading to increased morbidity and mortality. Rapid nutritional screening assessment should be performed in all patients with cirrhosis, and more specific tests for sarcopenia should be performed in those at high risk. The pathogenesis of malnutrition/sarcopenia in cirrhosis is complex/multifactorial and not just reduction in protein/calorie intake. Hyperammonemia appears to be the main driver of sarcopenia in cirrhosis through several molecular signalling pathways. Nutritional management in malnourished patients with cirrhosis should be undertaken by a multidisciplinary team to achieve adequate protein/calorie intake. While the role of branched-chained amino acids remains somewhat contentious in achieving a global benefit of decreasing mortality- and liver-related events, they, and vitamin supplements, are recommended for those with advanced liver disease. Novel strategies to reverse sarcopenia such as hormone supplementation, long-term ammonia-lowering agents and myostatin antagonists, are currently under investigation. CONCLUSIONS Malnutrition/sarcopenia and frailty are unique, inter-related and multi-dimensional problems in cirrhosis which require special attention, prompt assessment and appropriate management as they significantly impact morbidity and mortality.
Collapse
Affiliation(s)
- Chalermrat Bunchorntavakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Rajavithi Hospital, College of Medicine, Rangsit University, Bangkok, Thailand
| | - K Rajender Reddy
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
31
|
Cui P, Liu X, Zhao K, Hou S, Chen C, Zhao D, Zeng H. The novel axis of YAP1, transcription enhancer factor 3 and Down Syndrome Candidate Region 1 isoform 1L is a common signaling pathway downstream of several angiogenic factors. Microvasc Res 2019; 129:103955. [PMID: 31733305 DOI: 10.1016/j.mvr.2019.103955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/06/2019] [Accepted: 11/11/2019] [Indexed: 01/06/2023]
Abstract
Angiogenesis is a hallmark of many diseases. Previously, we found that Down Syndrome Candidate Region 1 Isoform 1L (DSCR1-1L) was expressed in human tumor vessels, but was not detectable in normal tissues, and played important roles in angiogenesis induced by vascular endothelial growth factor (VEGF-A165). The expressions of DSCR1-1L mRNA and protein induced by VEGF-A165 were regulated via the direct interaction of transcription enhancer factor 3 (TEF3) with DSCR1-1L promoter. However, the function and the regulation of DSCR1-1L in angiogenesis had not been completely understood. In this study, we found that the expressions of DSCR1-1L mRNA and proteins were upregulated by other angiogenic factors, including VEGF-A121, VEGF-E, histamine, PAF, the endothelial cell (EC) growth medium, and the conditional medium obtained from cancer cells, but not by PlGF, bFGF, PDGF, and serotonin. The EC proliferation, migration and elongation induced by histamine and EC growth medium were inhibited by knocking down the mRNA and protein expressions of DSCR1-1L and TEF3. The TEF3 activation was regulated by its interaction with YAP1, and translocation from cytosol to nuclei, but not by increase of protein expression, after the stimulation of VEGF, histamine and EC growth medium. YAP1 regulated the protein expression of DSCR1-1L, the proliferation, migration and elongation of ECs induced by VEGF, histamine and EC growth medium. Taken together, this study identified a novel axis of YAP1, TEF3 and DSCR1-1L that was a common signaling pathway downstream of several angiogenic factors to regulate angiogenesis, suggesting that this pathway is an excellent therapeutic target for angiogenic diseases and cancers. Our results contribute significantly to the field of mechanistic studies.
Collapse
Affiliation(s)
- Pengfei Cui
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Pancreatic Disease Institute, Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xin Liu
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Kevin Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Shiqiang Hou
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen Chen
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Surgery of Breast and Thyroid, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
32
|
Ferreira DMS, Cheng AJ, Agudelo LZ, Cervenka I, Chaillou T, Correia JC, Porsmyr-Palmertz M, Izadi M, Hansson A, Martínez-Redondo V, Valente-Silva P, Pettersson-Klein AT, Estall JL, Robinson MM, Nair KS, Lanner JT, Ruas JL. LIM and cysteine-rich domains 1 (LMCD1) regulates skeletal muscle hypertrophy, calcium handling, and force. Skelet Muscle 2019; 9:26. [PMID: 31666122 PMCID: PMC6822430 DOI: 10.1186/s13395-019-0214-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/30/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Skeletal muscle mass and strength are crucial determinants of health. Muscle mass loss is associated with weakness, fatigue, and insulin resistance. In fact, it is predicted that controlling muscle atrophy can reduce morbidity and mortality associated with diseases such as cancer cachexia and sarcopenia. METHODS We analyzed gene expression data from muscle of mice or human patients with diverse muscle pathologies and identified LMCD1 as a gene strongly associated with skeletal muscle function. We transiently expressed or silenced LMCD1 in mouse gastrocnemius muscle or in mouse primary muscle cells and determined muscle/cell size, targeted gene expression, kinase activity with kinase arrays, protein immunoblotting, and protein synthesis levels. To evaluate force, calcium handling, and fatigue, we transduced the flexor digitorum brevis muscle with a LMCD1-expressing adenovirus and measured specific force and sarcoplasmic reticulum Ca2+ release in individual fibers. Finally, to explore the relationship between LMCD1 and calcineurin, we ectopically expressed Lmcd1 in the gastrocnemius muscle and treated those mice with cyclosporine A (calcineurin inhibitor). In addition, we used a luciferase reporter construct containing the myoregulin gene promoter to confirm the role of a LMCD1-calcineurin-myoregulin axis in skeletal muscle mass control and calcium handling. RESULTS Here, we identify LIM and cysteine-rich domains 1 (LMCD1) as a positive regulator of muscle mass, that increases muscle protein synthesis and fiber size. LMCD1 expression in vivo was sufficient to increase specific force with lower requirement for calcium handling and to reduce muscle fatigue. Conversely, silencing LMCD1 expression impairs calcium handling and force, and induces muscle fatigue without overt atrophy. The actions of LMCD1 were dependent on calcineurin, as its inhibition using cyclosporine A reverted the observed hypertrophic phenotype. Finally, we determined that LMCD1 represses the expression of myoregulin, a known negative regulator of muscle performance. Interestingly, we observed that skeletal muscle LMCD1 expression is reduced in patients with skeletal muscle disease. CONCLUSIONS Our gain- and loss-of-function studies show that LMCD1 controls protein synthesis, muscle fiber size, specific force, Ca2+ handling, and fatigue resistance. This work uncovers a novel role for LMCD1 in the regulation of skeletal muscle mass and function with potential therapeutic implications.
Collapse
Affiliation(s)
- Duarte M S Ferreira
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Arthur J Cheng
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden.,Present Address: Faculty of Health, York University, School of Kinesiology and Health Science, Toronto, Ontario, Canada
| | - Leandro Z Agudelo
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden.,Present Address: Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Igor Cervenka
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Thomas Chaillou
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden.,School of Health Sciences, Örebro University, Örebro, Sweden
| | - Jorge C Correia
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Margareta Porsmyr-Palmertz
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Manizheh Izadi
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden.,Present Address: Karp Research Building, Boston, MA, 02115, USA
| | - Alicia Hansson
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Vicente Martínez-Redondo
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Paula Valente-Silva
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Amanda T Pettersson-Klein
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Jennifer L Estall
- Division of Cardiovascular and Metabolic Disease, Institut de recherches cliniques de Montreal (IRCM), Montreal, QC, Canada
| | - Matthew M Robinson
- Division of Endocrinology, Diabetes and Nutrition, Mayo Clinic, Rochester, MN, 55905, USA
| | - K Sreekumaran Nair
- Division of Endocrinology, Diabetes and Nutrition, Mayo Clinic, Rochester, MN, 55905, USA
| | - Johanna T Lanner
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden
| | - Jorge L Ruas
- Molecular & Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
33
|
Calcineurin Broadly Regulates the Initiation of Skeletal Muscle-Specific Gene Expression by Binding Target Promoters and Facilitating the Interaction of the SWI/SNF Chromatin Remodeling Enzyme. Mol Cell Biol 2019; 39:MCB.00063-19. [PMID: 31308130 PMCID: PMC6751634 DOI: 10.1128/mcb.00063-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/08/2019] [Indexed: 12/22/2022] Open
Abstract
Calcineurin (Cn) is a calcium-activated serine/threonine protein phosphatase that is broadly implicated in diverse cellular processes, including the regulation of gene expression. During skeletal muscle differentiation, Cn activates the nuclear factor of activated T-cell (NFAT) transcription factor but also promotes differentiation by counteracting the negative influences of protein kinase C beta (PKCβ) via dephosphorylation and activation of Brg1, an enzymatic subunit of the mammalian SWI/SNF ATP-dependent chromatin remodeling enzyme. Here we identified four major temporal patterns of Cn-dependent gene expression in differentiating myoblasts and determined that Cn is broadly required for the activation of the myogenic gene expression program. Mechanistically, Cn promotes gene expression through direct binding to myogenic promoter sequences and facilitating the binding of Brg1, other SWI/SNF subunit proteins, and MyoD, a critical lineage determinant for skeletal muscle differentiation. We conclude that the Cn phosphatase directly impacts the expression of myogenic genes by promoting ATP-dependent chromatin remodeling and formation of transcription-competent promoters.
Collapse
|
34
|
Healthy Body Weights With Corticosteroid-free Immunosuppression Is the Best Predictor of Cardiovascular Health in Children After Liver Transplantation. J Pediatr Gastroenterol Nutr 2019; 68:713-719. [PMID: 30676521 DOI: 10.1097/mpg.0000000000002271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Cardiometabolic dysregulation (CMD) influences morbidity and mortality risk in adults post-liver transplantation (LTx). CMD is reported in 10% to 25% of pediatric LTx recipients, but no information regarding the longitudinal expression of CMD is available. The study objective was to examine the longitudinal expression of CMD and associations with body composition and growth in children post-LTx. METHODS A retrospective review was conducted in youth (34 F/30 M) who underwent LTx between 1994 and 2015 at the Stollery Children's Hospital. Primary outcomes included serum markers of CMD (insulin, glucose, hemoglobin A1C [A1C], homeostasis model assessment for insulin resistance [abnormal >3], lipid panel triglycerides, total cholesterol, high-density lipoprotein-cholesterol, low-density lipoprotein-cholesterol) and systolic/diastolic blood pressure (BP: absolute/z scores). RESULTS Mean (±SD) age, weight z, height z, body mass index z was 9.7 ± 3.4 years (3.5-17.9), 0.26 ± 1.03, 0.017 ± 1.2, and 0.41 ± 1.05, respectively. The majority of children had percentage fat mass, percentage fat-free mass within normal reference ranges. Systolic/diastolic BP was within healthy references ranges in 83.1% and 93.5% of children, respectively. Serum insulin (83.4%) and high-density lipoprotein-cholesterol (43.9%) concentrations were low, with abnormal findings of other laboratory markers found in <5% of participants. Abnormal findings for metabolic parameters were independent of weight z, body mass index z, fat mass, and corticosteroids but were positively related to child's age (>9.7 years) and fat-free mass (total, arms). Insulin levels decreased significantly in the first 4 years post-LTx, but no changes in lipid panel, A1C and glucose were noted over 10 years. CONCLUSIONS Pediatric LTx recipients with healthy body weights and corticosteroid-free immunosuppression have a low expression of CMD over 10 years.
Collapse
|
35
|
Jindal A, Jagdish RK. Sarcopenia: Ammonia metabolism and hepatic encephalopathy. Clin Mol Hepatol 2019; 25:270-279. [PMID: 31006226 PMCID: PMC6759436 DOI: 10.3350/cmh.2019.0015] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/07/2019] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia (loss of muscle mass and/or strength) frequently complicates liver cirrhosis and adversely affects the quality of life; cirrhosis related liver decompensation and significantly decreases wait-list and post-liver transplantation survival. The main therapeutic strategies to improve or reverse sarcopenia include dietary interventions (supplemental calorie and protein intake), increased physical activity (supervised resistance and endurance exercises), hormonal therapy (testosterone), and ammonia lowering agents (L-ornithine L-aspartate, branch chain amino acids) as well as mechanistic approaches that target underlying molecular and metabolic abnormalities. Besides other factors, hyperammonemia has recently gained attention and increase sarcopenia by various mechanisms including increased expression of myostatin, increased phosphorylation of eukaryotic initiation factor 2a, cataplerosis of α ketoglutarate, mitochondrial dysfunction, increased reactive oxygen species that decrease protein synthesis and increased autophagy-mediated proteolysis. Sarcopenia contributes to frailty and increases the risk of minimal and overt hepatic encephalopathy.
Collapse
Affiliation(s)
- Ankur Jindal
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rakesh Kumar Jagdish
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
36
|
Mazumder N, Rinella M. Editorial: sarcopenia in liver transplantation-our weakest patients may need the strongest push. Aliment Pharmacol Ther 2019; 49:1100-1101. [PMID: 30920044 PMCID: PMC6483369 DOI: 10.1111/apt.15204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- N Mazumder
- Division of Hepatology, Department of Medicine Feinberg
School of Medicine, Northwestern University, Chicago, Illinois
| | - M Rinella
- Division of Hepatology, Department of Medicine Feinberg
School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
37
|
Ko YJ, Chang H, Lee DH. Analysis of Taurine's Anti-Down Syndrome Potential in Caenorhabditis elegans. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 2:1113-1128. [PMID: 28849527 DOI: 10.1007/978-94-024-1079-2_89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Down syndrome (DS) patients overexpress human DS critical region gene 1 (hDSCR-1), whose translational product inhibits calcineurin-dependent signaling pathways of genetic transcription. Compared to hDSCR-1, C. elegans rcn-1 has 40% sequence similarity and its proteins share an analogous function with hDSCR-1 in regulating calcineurin. Taurine has had a positive effect on DS patients. According to animal research studies, taurine reduces the expression of MCIP1, a calcineurin inhibitory protein, on C2C12 myotubes and fibroblast in mouse. This study utilizes two C. elegans models for DS: rcn-1 overexpression model, displaying a calcineurin-deficient phenotype, and calcineurin loss-of function mutants. C. elegans larvae were treated with taurine to characterize its effect and mechanism in helping DS patients. RCN-1 expression and behavioral changes were examined in rcn-1 overexpression and calcineurin-deficient models at different concentrations of taurine. When treated with taurine, transgenic worms harboring an rcn-1 reporter (RCN-1::GFP) showed a reduced level of rcn-1 mRNA expression and improved behaviors that were comparable to those in the wild type. These results indicate that taurine exerts a down-regulating effect on the expression of rcn-1 and, consequently, a positive effect on the expression of calcineurins. In summary, taurine may improve the DS symptoms by prompting a positive interaction between RCN-1 and calcineurin. Furthermore, these results suggest that novel mechanisms may regulate interactions among taurine, RCN-1 and calcineurin.
Collapse
Affiliation(s)
- Yun Jung Ko
- Department of Life Sciences, University of Seoul, Seoul, 02504, South Korea
| | | | - Dong Hee Lee
- Department of Life Sciences, University of Seoul, Seoul, 02504, South Korea.
| |
Collapse
|
38
|
van der Pijl R, Strom J, Conijn S, Lindqvist J, Labeit S, Granzier H, Ottenheijm C. Titin-based mechanosensing modulates muscle hypertrophy. J Cachexia Sarcopenia Muscle 2018; 9:947-961. [PMID: 29978560 PMCID: PMC6204599 DOI: 10.1002/jcsm.12319] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/30/2018] [Accepted: 05/22/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Titin is an elastic sarcomeric filament that has been proposed to play a key role in mechanosensing and trophicity of muscle. However, evidence for this proposal is scarce due to the lack of appropriate experimental models to directly test the role of titin in mechanosensing. METHODS We used unilateral diaphragm denervation (UDD) in mice, an in vivo model in which the denervated hemidiaphragm is passively stretched by the contralateral, innervated hemidiaphragm and hypertrophy rapidly occurs. RESULTS In wildtype mice, the denervated hemidiaphragm mass increased 48 ± 3% after 6 days of UDD, due to the addition of both sarcomeres in series and in parallel. To test whether titin stiffness modulates the hypertrophy response, RBM20ΔRRM and TtnΔIAjxn mouse models were used, with decreased and increased titin stiffness, respectively. RBM20ΔRRM mice (reduced stiffness) showed a 20 ± 6% attenuated hypertrophy response, whereas the TtnΔIAjxn mice (increased stiffness) showed an 18 ± 8% exaggerated response after UDD. Thus, muscle hypertrophy scales with titin stiffness. Protein expression analysis revealed that titin-binding proteins implicated previously in muscle trophicity were induced during UDD, MARP1 & 2, FHL1, and MuRF1. CONCLUSIONS Titin functions as a mechanosensor that regulates muscle trophicity.
Collapse
Affiliation(s)
- Robbert van der Pijl
- Department of Cellular and Molecular MedicineUniversity of ArizonaTucsonAZUSA
- Dept of PhysiologyVU University Medical CenterAmsterdamThe Netherlands
| | - Joshua Strom
- Department of Cellular and Molecular MedicineUniversity of ArizonaTucsonAZUSA
| | - Stefan Conijn
- Dept of PhysiologyVU University Medical CenterAmsterdamThe Netherlands
| | - Johan Lindqvist
- Department of Cellular and Molecular MedicineUniversity of ArizonaTucsonAZUSA
| | - Siegfried Labeit
- Department of Integrative PathophysiologyMedical Faculty MannheimMannheimGermany
- Myomedix GmbHNeckargemuendGermany
| | - Henk Granzier
- Department of Cellular and Molecular MedicineUniversity of ArizonaTucsonAZUSA
| | - Coen Ottenheijm
- Department of Cellular and Molecular MedicineUniversity of ArizonaTucsonAZUSA
- Dept of PhysiologyVU University Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
39
|
Mager DR, Hager A, Ooi PH, Siminoski K, Gilmour SM, Yap JY. Persistence of Sarcopenia After Pediatric Liver Transplantation Is Associated With Poorer Growth and Recurrent Hospital Admissions. JPEN J Parenter Enteral Nutr 2018; 43:271-280. [DOI: 10.1002/jpen.1414] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 06/04/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Diana R. Mager
- Department of Agricultural, Food & Nutritional SciencesUniversity of Alberta Edmonton Alberta Canada
- Department of PediatricsUniversity of Alberta Edmonton Alberta Canada
| | - Amber Hager
- Department of Agricultural, Food & Nutritional SciencesUniversity of Alberta Edmonton Alberta Canada
| | - Poh Hwa Ooi
- Department of Agricultural, Food & Nutritional SciencesUniversity of Alberta Edmonton Alberta Canada
| | | | - Susan M. Gilmour
- Department of PediatricsUniversity of Alberta Edmonton Alberta Canada
- Division of Pediatric Gastroenterology & Nutrition/Transplant ServicesThe Stollery Children's Hospital, Alberta Health Services Edmonton Alberta Canada
| | - Jason Y.K. Yap
- Department of PediatricsUniversity of Alberta Edmonton Alberta Canada
- Division of Pediatric Gastroenterology & Nutrition/Transplant ServicesThe Stollery Children's Hospital, Alberta Health Services Edmonton Alberta Canada
| |
Collapse
|
40
|
Zowalaty AEE, Ye X. Seipin deficiency leads to defective parturition in mice. Biol Reprod 2018; 97:378-386. [PMID: 29088395 DOI: 10.1093/biolre/iox088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/10/2017] [Indexed: 01/28/2023] Open
Abstract
Seipin is an integral endoplasmic reticulum membrane protein encoded by Berardinelli-Seip congenital lipodystrophy type 2 (BSCL2/Bscl2) gene. Seipin deficiency results in lipodystrophy, diabetes, muscle hypertrophy, and male infertility in both human and mouse. Seipin function in female reproduction is unknown. Bscl2-/- dams had normal embryo implantation and body weight gain during pregnancy but reduced delivery rates from 2nd to 4th pregnancies and reduced numbers of pups delivered from 1st to 4th pregnancies. Characterization of first pregnancy revealed increased gestation period and parturition problems, including uterine prolapse, difficulty in delivery, undelivered fetuses, and undelivered tissues in Bscl2-/- females. Bscl2-/- uterine weight was comparable to control at 3 weeks old but significantly increased with myometrial hypertrophy at 10 months old. In situ hybridization revealed relatively low level of Bscl2 mRNA expression in myometrium throughout pregnancy and postpartum but high level of expression in uterine luminal epithelium, suggesting that systemic effect (e.g. elevated glucose and insulin levels) rather than local seipin-deficiency in myometrium might be a main contributing factor to myometrial hypertrophy. On near-term gestation day 18.5 (D18.5), Bscl2-/- females had normal levels of serum progesterone and 17β-estradiol, indicating functional ovary and placenta. Proliferating Cell Nuclear Antigen (PCNA) staining showed minimal myometrial cell proliferation in both D18.5 Bscl2+/+ and Bscl2-/- uteri. There was strong LC3 immunostaining in Bscl2+/+ and Bscl2-/- peripartum myometrium and increased LC3 staining in Bscl2-/- peripartum uterine luminal epithelium, suggesting a potential role of seipin in regulating autophagy in uterine luminal epithelium but not myometrium. This study demonstrates an association of seipin with myometrium and parturition.
Collapse
Affiliation(s)
- Ahmed E El Zowalaty
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America.,Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, United States of America
| | - Xiaoqin Ye
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America.,Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
41
|
Navarro-Marquez M, Torrealba N, Troncoso R, Vásquez-Trincado C, Rodriguez M, Morales PE, Villalobos E, Eura Y, Garcia L, Chiong M, Klip A, Jaimovich E, Kokame K, Lavandero S. Herpud1 impacts insulin-dependent glucose uptake in skeletal muscle cells by controlling the Ca2+-calcineurin-Akt axis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1653-1662. [DOI: 10.1016/j.bbadis.2018.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 01/08/2023]
|
42
|
Dehkhoda F, Lee CMM, Medina J, Brooks AJ. The Growth Hormone Receptor: Mechanism of Receptor Activation, Cell Signaling, and Physiological Aspects. Front Endocrinol (Lausanne) 2018; 9:35. [PMID: 29487568 PMCID: PMC5816795 DOI: 10.3389/fendo.2018.00035] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/29/2018] [Indexed: 01/02/2023] Open
Abstract
The growth hormone receptor (GHR), although most well known for regulating growth, has many other important biological functions including regulating metabolism and controlling physiological processes related to the hepatobiliary, cardiovascular, renal, gastrointestinal, and reproductive systems. In addition, growth hormone signaling is an important regulator of aging and plays a significant role in cancer development. Growth hormone activates the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway, and recent studies have provided a new understanding of the mechanism of JAK2 activation by growth hormone binding to its receptor. JAK2 activation is required for growth hormone-mediated activation of STAT1, STAT3, and STAT5, and the negative regulation of JAK-STAT signaling comprises an important step in the control of this signaling pathway. The GHR also activates the Src family kinase signaling pathway independent of JAK2. This review covers the molecular mechanisms of GHR activation and signal transduction as well as the physiological consequences of growth hormone signaling.
Collapse
Affiliation(s)
- Farhad Dehkhoda
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Christine M. M. Lee
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Johan Medina
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Brooks
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
43
|
Kristina Parr M, Müller-Schöll A. Pharmacology of doping agents—mechanisms promoting muscle hypertrophy. AIMS MOLECULAR SCIENCE 2018. [DOI: 10.3934/molsci.2018.2.131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
44
|
Bandyopadhyay A, Dewangan VK, Vajanthri KY, Poddar S, Mahto SK. Easy and affordable method for rapid prototyping of tissue models in vitro using three-dimensional bioprinting. Biocybern Biomed Eng 2018. [DOI: 10.1016/j.bbe.2017.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Chow YL, Iwata Y, Sato F. Dihydrosanguinarine Enhances Glucose Uptake in Mouse 3T3-L1 Cells. ACS OMEGA 2017; 2:6916-6925. [PMID: 29202114 PMCID: PMC5705173 DOI: 10.1021/acsomega.7b01134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/05/2017] [Indexed: 06/07/2023]
Abstract
Recently, more studies have aimed at identifying selective peroxisome proliferator-activated receptor gamma (PPARγ) modulators that transactivate the expression of PPARγ-dependent genes as partial agonists to improve diabetic symptoms with fewer side effects compared to classic PPARγ agonists such as thiazolidinediones. We found that dihydrosanguinarine (DHS) treatment induced preadipocyte differentiation and lipid droplet accumulation in 3T3-L1 cells, but this effect is weaker than that elicited by the full PPARγ agonist troglitazone. Furthermore, this effect was reduced by the addition of a PPARγ antagonist, indicating the involvement of PPARγ signaling. Our results suggest that the stimulatory effects of DHS on adipocyte differentiation and insulin sensitivity are mediated by suppressing adenosine monophosphate-activated protein kinase (AMPK) alpha, upregulating the expression of PPARγ and its target genes (particularly Glut-4 and adiponectin) and reducing PPARγ phosphorylation. DHS significantly enhanced the glucose uptake in 3T3-L1 adipocytes without observable cytotoxicity at the effective concentration (5 μM) applied.
Collapse
|
46
|
Abstract
Skeletal muscle is the largest tissue in the body and loss of its function or its regenerative properties results in debilitating musculoskeletal disorders. Understanding the mechanisms that drive skeletal muscle formation will not only help to unravel the molecular basis of skeletal muscle diseases, but also provide a roadmap for recapitulating skeletal myogenesis in vitro from pluripotent stem cells (PSCs). PSCs have become an important tool for probing developmental questions, while differentiated cell types allow the development of novel therapeutic strategies. In this Review, we provide a comprehensive overview of skeletal myogenesis from the earliest premyogenic progenitor stage to terminally differentiated myofibers, and discuss how this knowledge has been applied to differentiate PSCs into muscle fibers and their progenitors in vitro.
Collapse
Affiliation(s)
- Jérome Chal
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA .,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
47
|
Jingting S, Qin X, Yanju S, Ming Z, Yunjie T, Gaige J, Zhongwei S, Jianmin Z. Oxidative and glycolytic skeletal muscles show marked differences in gene expression profile in Chinese Qingyuan partridge chickens. PLoS One 2017; 12:e0183118. [PMID: 28813489 PMCID: PMC5558948 DOI: 10.1371/journal.pone.0183118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/28/2017] [Indexed: 12/17/2022] Open
Abstract
Oxidative and glycolytic myofibers have different structures and metabolic characteristics and their ratios are important in determining poultry meat quality. However, the molecular mechanisms underlying their differences are unclear. In this study, global gene expression profiling was conducted in oxidative skeletal muscle (obtained from the soleus, or SOL) and glycolytic skeletal muscle (obtained from the extensor digitorum longus, or EDL) of Chinese Qingyuan partridge chickens, using the Agilent Chicken Gene Expression Chip. A total of 1224 genes with at least 2-fold differences were identified (P < 0.05), of which 654 were upregulated and 570 were downregulated in SOL. GO, KEGG pathway, and co-expressed gene network analyses suggested that PRKAG3, ATP2A2, and PPARGC1A might play important roles in myofiber composition. The function of PPARGC1A gene was further validated. PPARGC1A mRNA expression levels were higher in SOL than in EDL muscles throughout the early postnatal development stages. In myoblast cells, shRNA knockdown of PPARGC1A significantly inhibited some muscle development and transition-related genes, including PPP3CA, MEF2C, and SM (P < 0.01 or P < 0.05), and significantly upregulated the expression of FWM (P < 0.05). Our study demonstrates strong transcriptome differences between oxidative and glycolytic myofibers, and the results suggest that PPARGC1A is a key gene involved in chicken myofiber composition and transition.
Collapse
Affiliation(s)
- Shu Jingting
- Key laboratory for poultry genetics and breeding of Jiangsu province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, Jiangsu, China
| | - Xiao Qin
- Key laboratory for poultry genetics and breeding of Jiangsu province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, Jiangsu, China
| | - Shan Yanju
- Key laboratory for poultry genetics and breeding of Jiangsu province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, Jiangsu, China
| | - Zhang Ming
- Key laboratory for poultry genetics and breeding of Jiangsu province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, Jiangsu, China
| | - Tu Yunjie
- Key laboratory for poultry genetics and breeding of Jiangsu province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, Jiangsu, China
| | - Ji Gaige
- Key laboratory for poultry genetics and breeding of Jiangsu province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, Jiangsu, China
| | - Sheng Zhongwei
- Key laboratory for poultry genetics and breeding of Jiangsu province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, Jiangsu, China
| | - Zou Jianmin
- Key laboratory for poultry genetics and breeding of Jiangsu province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, Jiangsu, China
| |
Collapse
|
48
|
Elam MB, Majumdar G, Mozhui K, Gerling IC, Vera SR, Fish-Trotter H, Williams RW, Childress RD, Raghow R. Patients experiencing statin-induced myalgia exhibit a unique program of skeletal muscle gene expression following statin re-challenge. PLoS One 2017; 12:e0181308. [PMID: 28771594 PMCID: PMC5542661 DOI: 10.1371/journal.pone.0181308] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/29/2017] [Indexed: 01/21/2023] Open
Abstract
Statins, the 3-hydroxy-3-methyl-glutaryl (HMG)-CoA reductase inhibitors, are widely prescribed for treatment of hypercholesterolemia. Although statins are generally well tolerated, up to ten percent of statin-treated patients experience myalgia symptoms, defined as muscle pain without elevated creatinine phosphokinase (CPK) levels. Myalgia is the most frequent reason for discontinuation of statin therapy. The mechanisms underlying statin myalgia are not clearly understood. To elucidate changes in gene expression associated with statin myalgia, we compared profiles of gene expression in skeletal muscle biopsies from patients with statin myalgia who were undergoing statin re-challenge (cases) versus those of statin-tolerant controls. A robust separation of case and control cohorts was revealed by Principal Component Analysis of differentially expressed genes (DEGs). To identify putative gene expression and metabolic pathways that may be perturbed in skeletal muscles of patients with statin myalgia, we subjected DEGs to Ingenuity Pathways (IPA) and DAVID (Database for Annotation, Visualization and Integrated Discovery) analyses. The most prominent pathways altered by statins included cellular stress, apoptosis, cell senescence and DNA repair (TP53, BARD1, Mre11 and RAD51); activation of pro-inflammatory immune response (CXCL12, CST5, POU2F1); protein catabolism, cholesterol biosynthesis, protein prenylation and RAS-GTPase activation (FDFT1, LSS, TP53, UBD, ATF2, H-ras). Based on these data we tentatively conclude that persistent myalgia in response to statins may emanate from cellular stress underpinned by mechanisms of post-inflammatory repair and regeneration. We also posit that this subset of individuals is genetically predisposed to eliciting altered statin metabolism and/or increased end-organ susceptibility that lead to a range of statin-induced myopathies. This mechanistic scenario is further bolstered by the discovery that a number of single nucleotide polymorphisms (e.g., SLCO1B1, SLCO2B1 and RYR2) associated with statin myalgia and myositis were observed with increased frequency among patients with statin myalgia.
Collapse
Affiliation(s)
- Marshall B. Elam
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
- Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
- * E-mail: (MBE); (RR)
| | - Gipsy Majumdar
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Khyobeni Mozhui
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Ivan C. Gerling
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Santiago R. Vera
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Hannah Fish-Trotter
- Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Robert W. Williams
- Department of Genetics, Genomics and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Richard D. Childress
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Medicine, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
| | - Rajendra Raghow
- Department of Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
- Department of Pharmacology, University of Tennessee Health Sciences Center, Memphis, Tennessee, United States of America
- * E-mail: (MBE); (RR)
| |
Collapse
|
49
|
Fajardo VA, Rietze BA, Chambers PJ, Bellissimo C, Bombardier E, Quadrilatero J, Tupling AR. Effects of sarcolipin deletion on skeletal muscle adaptive responses to functional overload and unload. Am J Physiol Cell Physiol 2017; 313:C154-C161. [PMID: 28592414 DOI: 10.1152/ajpcell.00291.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 05/30/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022]
Abstract
Overexpression of sarcolipin (SLN), a regulator of sarco(endo)plasmic reticulum Ca2+-ATPases (SERCAs), stimulates calcineurin signaling to enhance skeletal muscle oxidative capacity. Some studies have shown that calcineurin may also control skeletal muscle mass and remodeling in response to functional overload and unload stimuli by increasing myofiber size and the proportion of slow fibers. To examine whether SLN might mediate these adaptive responses, we performed soleus and gastrocnemius tenotomy in wild-type (WT) and Sln-null (Sln-/-) mice and examined the overloaded plantaris and unloaded/tenotomized soleus muscles. In the WT overloaded plantaris, we observed ectopic expression of SLN, myofiber hypertrophy, increased fiber number, and a fast-to-slow fiber type shift, which were associated with increased calcineurin signaling (NFAT dephosphorylation and increased stabilin-2 protein content) and reduced SERCA activity. In the WT tenotomized soleus, we observed a 14-fold increase in SLN protein, myofiber atrophy, decreased fiber number, and a slow-to-fast fiber type shift, which were also associated with increased calcineurin signaling and reduced SERCA activity. Genetic deletion of Sln altered these physiological outcomes, with the overloaded plantaris myofibers failing to grow in size and number, and transition towards the slow fiber type, while the unloaded soleus muscles exhibited greater reductions in fiber size and number, and an accelerated slow-to-fast fiber type shift. In both the Sln-/- overloaded and unloaded muscles, these findings were associated with elevated SERCA activity and blunted calcineurin signaling. Thus, SLN plays an important role in adaptive muscle remodeling potentially through calcineurin stimulation, which could have important implications for other muscle diseases and conditions.
Collapse
Affiliation(s)
- Val A Fajardo
- Department of Kinesiology, University of Waterloo, Waterloo Ontario, Canada
| | - Bradley A Rietze
- Department of Kinesiology, University of Waterloo, Waterloo Ontario, Canada
| | - Paige J Chambers
- Department of Kinesiology, University of Waterloo, Waterloo Ontario, Canada
| | | | - Eric Bombardier
- Department of Kinesiology, University of Waterloo, Waterloo Ontario, Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo, Waterloo Ontario, Canada
| | - A Russell Tupling
- Department of Kinesiology, University of Waterloo, Waterloo Ontario, Canada
| |
Collapse
|
50
|
Fajardo VA, Gamu D, Mitchell A, Bloemberg D, Bombardier E, Chambers PJ, Bellissimo C, Quadrilatero J, Tupling AR. Sarcolipin deletion exacerbates soleus muscle atrophy and weakness in phospholamban overexpressing mice. PLoS One 2017; 12:e0173708. [PMID: 28278204 PMCID: PMC5344511 DOI: 10.1371/journal.pone.0173708] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/24/2017] [Indexed: 11/18/2022] Open
Abstract
Sarcolipin (SLN) and phospholamban (PLN) are two small proteins that regulate the sarco(endo)plasmic reticulum Ca2+-ATPase pumps. In a recent study, we discovered that Pln overexpression (PlnOE) in slow-twitch type I skeletal muscle fibers drastically impaired SERCA function and caused a centronuclear myopathy-like phenotype, severe muscle atrophy and weakness, and an 8 to 9-fold upregulation of SLN protein in the soleus muscles. Here, we sought to determine the physiological role of SLN upregulation, and based on its role as a SERCA inhibitor, we hypothesized that it would represent a maladaptive response that contributes to the SERCA dysfunction and the overall myopathy observed in the PlnOE mice. To this end, we crossed Sln-null (SlnKO) mice with PlnOE mice to generate a PlnOE/SlnKO mouse colony and assessed SERCA function, CNM pathology, in vitro contractility, muscle mass, calcineurin signaling, daily activity and food intake, and proteolytic enzyme activity. Our results indicate that genetic deletion of Sln did not improve SERCA function nor rescue the CNM phenotype, but did result in exacerbated muscle atrophy and weakness, due to a failure to induce type II fiber compensatory hypertrophy and a reduction in total myofiber count. Mechanistically, our findings suggest that impaired calcineurin activation and resultant decreased expression of stabilin-2, and/or impaired autophagic signaling could be involved. Future studies should examine these possibilities. In conclusion, our study demonstrates the importance of SLN upregulation in combating muscle myopathy in the PlnOE mice, and since SLN is upregulated across several myopathies, our findings may reveal SLN as a novel and universal therapeutic target.
Collapse
Affiliation(s)
- Val A. Fajardo
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
| | - Daniel Gamu
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
| | - Andrew Mitchell
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
| | - Darin Bloemberg
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
| | - Eric Bombardier
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
| | - Paige J. Chambers
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
| | - Catherine Bellissimo
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
| | - A. Russell Tupling
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada, N2L 3G1
- * E-mail:
| |
Collapse
|