1
|
Fu X, Zhang F, Dong X, Pu L, Feng Y, Xu Y, Gao F, Liang T, Kang J, Sun H, Hong T, Liu Y, Zhou H, Jiang J, Yin D, Hu X, Wang DZ, Ding J, Chen J. Adapting cytoskeleton-mitochondria patterning with myocyte differentiation by promyogenic PRR33. Cell Death Differ 2024:10.1038/s41418-024-01363-w. [PMID: 39147882 DOI: 10.1038/s41418-024-01363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Coordinated cytoskeleton-mitochondria organization during myogenesis is crucial for muscle development and function. Our understanding of the underlying regulatory mechanisms remains inadequate. Here, we identified a novel muscle-enriched protein, PRR33, which is upregulated during myogenesis and acts as a promyogenic factor. Depletion of Prr33 in C2C12 represses myoblast differentiation. Genetic deletion of Prr33 in mice reduces myofiber size and decreases muscle strength. The Prr33 mutant mice also exhibit impaired myogenesis and defects in muscle regeneration in response to injury. Interactome and transcriptome analyses reveal that PRR33 regulates cytoskeleton and mitochondrial function. Remarkably, PRR33 interacts with DESMIN, a key regulator of cytoskeleton-mitochondria organization in muscle cells. Abrogation of PRR33 in myocytes substantially abolishes the interaction of DESMIN filaments with mitochondria, leading to abnormal intracellular accumulation of DESMIN and mitochondrial disorganization/dysfunction in myofibers. Together, our findings demonstrate that PRR33 and DESMIN constitute an important regulatory module coordinating mitochondrial organization with muscle differentiation.
Collapse
Affiliation(s)
- Xuyang Fu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Feng Zhang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Xiaoxuan Dong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Linbin Pu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Yan Feng
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yang Xu
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Feng Gao
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tian Liang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Jianmeng Kang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Hongke Sun
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tingting Hong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yunxia Liu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hongmei Zhou
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jun Jiang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Deling Yin
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xinyang Hu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Da-Zhi Wang
- University of South Florida Health Heart Institute, Center for Regenerative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33602, USA
| | - Jian Ding
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Jinghai Chen
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| |
Collapse
|
2
|
Fallahtafti F, Samson K, Salamifar Z, Johanning J, Pipinos I, Myers SA. Enhancing walking performance in patients with peripheral arterial disease: An intervention with ankle-foot orthosis. Int J Cardiol 2024; 407:131992. [PMID: 38527630 DOI: 10.1016/j.ijcard.2024.131992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
Lower extremity peripheral artery disease (PAD) is a cardiovascular condition manifesting from narrowed or blocked arteries supplying the legs. Gait is impaired in patients with PAD. Recent evidence suggests that walking with carbon fiber ankle foot orthoses (AFOs) can improve patient mobility and delay claudication time. This study aimed to employ advanced biomechanical gait analysis to evaluate the impact of AFO intervention on gait performance among patients with PAD. Patients with claudication had hip, knee, and ankle joint kinetics and kinematics assessed using a cross-over intervention design. Participants walked over the force platforms with and without AFOs while kinematic data was recorded with motion analysis cameras. Kinetics and kinematics were combined to quantify torques and powers during the stance period of the gait cycle. The AFOs effectively reduced the excessive ankle plantar flexion and knee extension angles, bringing the patients' joint motions closer to those observed in healthy individuals. After 3 months of the AFO intervention, the hip range of motion decreased, likely due to changes occurring within the ankle chain. With the assistance of the AFOs, the biological power generation required from the ankle and hip during the push-off phase of walking decreased. Wearing AFOs resulted in increased knee flexor torque during the loading response phase of the gait. Based on this study, AFOs may allow patients with PAD to maintain or improve gait performance. More investigation is needed to fully understand and improve the potential benefits of ankle assistive devices.
Collapse
Affiliation(s)
- Farahnaz Fallahtafti
- Department of Biomechanics, University of Nebraska at Omaha, 6160 University Drive South, Omaha, NE 68182, USA.
| | - Kaeli Samson
- Department of Biostatistics, University of Nebraska Medical Center, 984375 Nebraska Medical Center, Omaha, NE 68198-4375, USA
| | - Zahra Salamifar
- Department of Biomechanics, University of Nebraska at Omaha, 6160 University Drive South, Omaha, NE 68182, USA
| | - Jason Johanning
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68105, USA; Department of Surgery and VA Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Iraklis Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68105, USA; Department of Surgery and VA Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Sara A Myers
- Department of Biomechanics, University of Nebraska at Omaha, 6160 University Drive South, Omaha, NE 68182, USA; Department of Surgery and VA Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA.
| |
Collapse
|
3
|
Wilburn D, Miserlis D, Fletcher E, Papoutsi E, Ismaeel A, Bradley C, Ring A, Wilkinson T, Smith RS, Ferrer L, Haynatzki G, Monteleone P, Banerjee S, Brisbois E, Bohannon WT, Koutakis P. Skeletal muscle desmin alterations following revascularization in peripheral artery disease claudicants. Sci Rep 2024; 14:12609. [PMID: 38824194 PMCID: PMC11144188 DOI: 10.1038/s41598-024-63626-3] [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: 01/11/2024] [Accepted: 05/30/2024] [Indexed: 06/03/2024] Open
Abstract
Peripheral artery disease (PAD) is characterized by varying severity of arterial stenosis, exercise induced claudication, malperfused tissue precluding normal healing and skeletal muscle dysfunction. Revascularization interventions improve circulation, but post-reperfusion changes within the skeletal muscle are not well characterized. This study investigates if revascularization enhanced hemodynamics increases walking performance with concurrent improvement of mitochondrial function and reverses abnormal skeletal muscle morphological features that develop with PAD. Fifty-eight patients completed walking performance testing and muscle biopsy before and 6 months after revascularization procedures. Muscle fiber morphology, desmin structure, and mitochondria respiration assessments before and after the revascularization were evaluated. Revascularization improved limb hemodynamics, walking function, and muscle morphology. Qualitatively not all participants recovered normal structural architecture of desmin in the myopathic myofibers after revascularization. Heterogenous responses in the recovery of desmin structure following revascularization may be caused by other underlying factors not reversed with hemodynamic improvements. Revascularization interventions clinically improve patient walking ability and can reverse the multiple subcellular functional and structural abnormalities in muscle cells. Further study is needed to characterize desmin structural remodeling with improvements in skeletal muscle morphology and function.
Collapse
Affiliation(s)
- Dylan Wilburn
- Department of Biology, Baylor University, B.207 Baylor Science Building, One Bear Place #97388, Waco, TX, 76798-7388, USA
| | - Dimitrios Miserlis
- Department of Surgery and Perioperative Care, University of Texas, Austin, TX, USA
- Department of Surgery, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Emma Fletcher
- Department of Biology, Baylor University, B.207 Baylor Science Building, One Bear Place #97388, Waco, TX, 76798-7388, USA
| | - Evlampia Papoutsi
- Department of Biology, Baylor University, B.207 Baylor Science Building, One Bear Place #97388, Waco, TX, 76798-7388, USA
| | - Ahmed Ismaeel
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Cassandra Bradley
- Department of Biology, Baylor University, B.207 Baylor Science Building, One Bear Place #97388, Waco, TX, 76798-7388, USA
| | - Andrew Ring
- Department of Biology, Baylor University, B.207 Baylor Science Building, One Bear Place #97388, Waco, TX, 76798-7388, USA
| | - Trevor Wilkinson
- Department of Biology, Baylor University, B.207 Baylor Science Building, One Bear Place #97388, Waco, TX, 76798-7388, USA
| | - Robert S Smith
- Department of Surgery, Baylor Scott & White Medical Center, Temple, TX, USA
| | - Lucas Ferrer
- Department of Surgery and Perioperative Care, University of Texas, Austin, TX, USA
| | - Gleb Haynatzki
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter Monteleone
- Department of Internal Medicine, University of Texas, Austin, TX, USA
| | - Subhash Banerjee
- Department of Cardiology, Baylor Scott & White Medical Center, Dallas, TX, USA
| | - Elizabeth Brisbois
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, GA, USA
| | - William T Bohannon
- Department of Surgery, Baylor Scott & White Medical Center, Temple, TX, USA
| | - Panagiotis Koutakis
- Department of Biology, Baylor University, B.207 Baylor Science Building, One Bear Place #97388, Waco, TX, 76798-7388, USA.
| |
Collapse
|
4
|
Bradley CE, Fletcher E, Wilkinson T, Ring A, Ferrer L, Miserlis D, Pacher P, Koutakis P. Mitochondrial fatty acid beta-oxidation: a possible therapeutic target for skeletal muscle lipotoxicity in peripheral artery disease myopathy. EXCLI JOURNAL 2024; 23:523-533. [PMID: 38741727 PMCID: PMC11089102 DOI: 10.17179/excli2024-7004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
Peripheral artery disease (PAD) is an atherosclerotic disease impacting over 200 million individuals and the prevalence increases with age. PAD occurs when plaque builds up within the peripheral arteries, leading to reduced blood flow and oxygen supply to the outer extremities. Individuals who experience PAD suffer from ischemia, which is typically accompanied by significant damage to skeletal muscles. Additionally, this tissue damage affects mitochondria, causing them to become dysregulated and dysfunctional, resulting in decreased metabolic rates. As there is no known cure for PAD, researchers are exploring potential therapeutic targets by examining coexisting cardiovascular conditions and metabolic risk factors, such as the aging process. Among these comorbidities, type-two diabetes mellitus and obesity are particularly common in PAD cases. These conditions, along with aging itself, are associated with an elevated accumulation of ectopic lipids within skeletal muscles, similar to what is observed in PAD. Researchers have attempted to reduce excess lipid accumulation by increasing the rate of fatty acid beta oxidation. Manipulating acetyl coenzyme A carboxylase 2, a key regulatory protein of fatty acid beta oxidation, has been the primary focus of such research. When acetyl coenzyme A carboxylase 2 is inhibited, it interrupts the conversion of acetyl-CoA into malonyl-CoA, resulting in an increase in the rate of fatty acid beta oxidation. By utilizing samples from PAD patients and applying the pharmacological strategies developed for acetyl coenzyme A carboxylase 2 in diabetes and obesity to PAD, a potential new therapeutic avenue may emerge, offering hope for improved quality of life for individuals suffering from PAD.
Collapse
Affiliation(s)
- Cassandra E. Bradley
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| | - Emma Fletcher
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| | - Trevor Wilkinson
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| | - Andrew Ring
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| | - Lucas Ferrer
- Department of Surgery, University of Texas at Austin Dell Medical School, 1601 Trinity St, Room 6708A, Austin, TX 78712, USA
| | - Dimitrios Miserlis
- Department of Surgery, University of Texas at Austin Dell Medical School, 1601 Trinity St, Room 6708A, Austin, TX 78712, USA
| | - Pal Pacher
- National Institutes of Health, Bethesda, MD, USA
| | - Panagiotis Koutakis
- Department of Biology, Baylor University, One Bear Place #97388, Waco, TX 76798, USA
| |
Collapse
|
5
|
Koutakis P, Hernandez H, Miserlis D, Thompson JR, Papoutsi E, Mietus CJ, Haynatzki G, Kim JK, Casale GP, Pipinos II. Oxidative damage in the gastrocnemius predicts long-term survival in patients with peripheral artery disease. NPJ AGING 2024; 10:21. [PMID: 38580664 PMCID: PMC10997596 DOI: 10.1038/s41514-024-00147-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/20/2024] [Indexed: 04/07/2024]
Abstract
Patients with peripheral artery disease (PAD) have increased mortality rates and a myopathy in their affected legs which is characterized by increased oxidative damage, reduced antioxidant enzymatic activity and defective mitochondrial bioenergetics. This study evaluated the hypothesis that increased levels of oxidative damage in gastrocnemius biopsies from patients with PAD predict long-term mortality rates. Oxidative damage was quantified as carbonyl adducts in myofibers of the gastrocnemius of PAD patients. The oxidative stress data were grouped into tertiles and the 5-year, all-cause mortality for each tertile was determined by Kaplan-Meier curves and compared by the Modified Peto test. A Cox-regression model was used to control the effects of clinical characteristics. Results were adjusted for age, sex, race, body mass index, ankle-brachial index, smoking, physical activity, and comorbidities. Of the 240 study participants, 99 died during a mean follow up of 37.8 months. Patients in the highest tertile of oxidative damage demonstrated the highest 5-year mortality rate. The mortality hazard ratios (HR) from the Cox analysis were statistically significant for oxidative damage (lowest vs middle tertile; HR = 6.33; p = 0.0001 and lowest vs highest; HR = 8.37; p < 0.0001). Survival analysis of a contemporaneous population of PAD patients identifies abundance of carbonyl adducts in myofibers of their gastrocnemius as a predictor of mortality rate independently of ankle-brachial index, disease stage and other clinical and myopathy-related covariates.
Collapse
Affiliation(s)
- Panagiotis Koutakis
- Department of Biology, Baylor University, Waco, TX, USA.
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Hernan Hernandez
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dimitrios Miserlis
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Surgery and Perioperative Care, University of Texas at Austin, Austin, TX, USA
| | - Jonathan R Thompson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Evlampia Papoutsi
- Department of Biology, Baylor University, Waco, TX, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Constance J Mietus
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Neurological Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - Gleb Haynatzki
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Julian K Kim
- Department of Biology, Baylor University, Waco, TX, USA
| | - George P Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Iraklis I Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Surgery and VA Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
6
|
Träger AP, Günther JS, Raming R, Paulus LP, Lang W, Meyer A, Kempf J, Caranovic M, Li Y, Wagner AL, Tan L, Danko V, Trollmann R, Woelfle J, Klett D, Neurath MF, Regensburger AP, Eckstein M, Uter W, Uder M, Herrmann Y, Waldner MJ, Knieling F, Rother U. Hybrid ultrasound and single wavelength optoacoustic imaging reveals muscle degeneration in peripheral artery disease. PHOTOACOUSTICS 2024; 35:100579. [PMID: 38312805 PMCID: PMC10835356 DOI: 10.1016/j.pacs.2023.100579] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/18/2023] [Accepted: 11/29/2023] [Indexed: 02/06/2024]
Abstract
Peripheral arterial disease (PAD) leads to chronic vascular occlusion and results in end organ damage in critically perfused limbs. There are currently no clinical methods available to determine the muscular damage induced by chronic mal-perfusion. This monocentric prospective cross-sectional study investigated n = 193 adults, healthy to severe PAD, in order to quantify the degree of calf muscle degeneration caused by PAD using a non-invasive hybrid ultrasound and single wavelength optoacoustic imaging (US/SWL-OAI) approach. While US provides morphologic information, SWL-OAI visualizes the absorption of pulsed laser light and the resulting sound waves from molecules undergoing thermoelastic expansion. US/SWL-OAI was compared to multispectral data, clinical disease severity, angiographic findings, phantom experiments, and histological examinations from calf muscle biopsies. We were able to show that synergistic use of US/SWL-OAI is most likely to map clinical degeneration of the muscle and progressive PAD.
Collapse
Affiliation(s)
- Anna P. Träger
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
| | - Josefine S. Günther
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
| | - Roman Raming
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Lars-Philip Paulus
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Werner Lang
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
| | - Alexander Meyer
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
| | - Julius Kempf
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
| | - Milenko Caranovic
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
| | - Yi Li
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
| | - Alexandra L. Wagner
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Lina Tan
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Vera Danko
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Regina Trollmann
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Daniel Klett
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, D-91054 Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, D-91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), University Hospital Erlangen, Ulmenweg 18, D-91054 Erlangen, Germany
- Erlangen Graduate School in Advanced Optical Technologies (SAOT), Friedrich-Alexander-Universität Erlangen-Nürnberg, Paul-Gordan-Straße 6, D-91052 Erlangen, Germany
| | - Adrian P. Regensburger
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Markus Eckstein
- Department of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstrasse 8-10, D-91054 Erlangen, Germany
| | - Wolfgang Uter
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-Universität Erlangen-Nürrnberg (FAU), Waldstraße 6, D-91054 Erlangen, Germany
| | - Michael Uder
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander, Universität Erlangen-Nürnberg (FAU), Maximiliansplatz 1, D-91054 Erlangen, Germany
| | - Yvonne Herrmann
- Department of Pediatric Cardiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Maximilian J. Waldner
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, D-91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), University Hospital Erlangen, Ulmenweg 18, D-91054 Erlangen, Germany
- Erlangen Graduate School in Advanced Optical Technologies (SAOT), Friedrich-Alexander-Universität Erlangen-Nürnberg, Paul-Gordan-Straße 6, D-91052 Erlangen, Germany
| | - Ferdinand Knieling
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nuremberg (FAU), Loschgestraße 15, D-91054 Erlangen, Germany
| | - Ulrich Rother
- Department of Vascular Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 12, D-91054 Erlangen, Germany
| |
Collapse
|
7
|
Claeyssen C, Bulangalire N, Bastide B, Agbulut O, Cieniewski-Bernard C. Desmin and its molecular chaperone, the αB-crystallin: How post-translational modifications modulate their functions in heart and skeletal muscles? Biochimie 2024; 216:137-159. [PMID: 37827485 DOI: 10.1016/j.biochi.2023.10.002] [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: 04/28/2023] [Revised: 08/04/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
Maintenance of the highly organized striated muscle tissue requires a cell-wide dynamic network through protein-protein interactions providing an effective mechanochemical integrator of morphology and function. Through a continuous and complex trans-cytoplasmic network, desmin intermediate filaments ensure this essential role in heart and in skeletal muscle. Besides their role in the maintenance of cell shape and architecture (permitting contractile activity efficiency and conferring resistance towards mechanical stress), desmin intermediate filaments are also key actors of cell and tissue homeostasis. Desmin participates to several cellular processes such as differentiation, apoptosis, intracellular signalisation, mechanotransduction, vesicle trafficking, organelle biogenesis and/or positioning, calcium homeostasis, protein homeostasis, cell adhesion, metabolism and gene expression. Desmin intermediate filaments assembly requires αB-crystallin, a small heat shock protein. Over its chaperone activity, αB-crystallin is involved in several cellular functions such as cell integrity, cytoskeleton stabilization, apoptosis, autophagy, differentiation, mitochondria function or aggresome formation. Importantly, both proteins are known to be strongly associated to the aetiology of several cardiac and skeletal muscles pathologies related to desmin filaments disorganization and a strong disturbance of desmin interactome. Note that these key proteins of cytoskeleton architecture are extensively modified by post-translational modifications that could affect their functional properties. Therefore, we reviewed in the herein paper the impact of post-translational modifications on the modulation of cellular functions of desmin and its molecular chaperone, the αB-crystallin.
Collapse
Affiliation(s)
- Charlotte Claeyssen
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Nathan Bulangalire
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France; Université de Lille, CHU Lille, F-59000 Lille, France
| | - Bruno Bastide
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 75005, Paris, France
| | - Caroline Cieniewski-Bernard
- University of Lille, University of Artois, University of Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France.
| |
Collapse
|
8
|
Fletcher E, Miserlis D, Sorokolet K, Wilburn D, Bradley C, Papoutsi E, Wilkinson T, Ring A, Ferrer L, Haynatzki G, Smith RS, Bohannon WT, Koutakis P. Diet-induced obesity augments ischemic myopathy and functional decline in a murine model of peripheral artery disease. Transl Res 2023; 260:17-31. [PMID: 37220835 PMCID: PMC11388035 DOI: 10.1016/j.trsl.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/25/2023]
Abstract
Peripheral artery disease (PAD) causes an ischemic myopathy contributing to patient disability and mortality. Most preclinical models to date use young, healthy rodents with limited translatability to human disease. Although PAD incidence increases with age, and obesity is a common comorbidity, the pathophysiologic association between these risk factors and PAD myopathy is unknown. Using our murine model of PAD, we sought to elucidate the combined effect of age, diet-induced obesity and chronic hindlimb ischemia (HLI) on (1) mobility, (2) muscle contractility, and markers of muscle (3) mitochondrial content and function, (4) oxidative stress and inflammation, (5) proteolysis, and (6) cytoskeletal damage and fibrosis. Following 16-weeks of high-fat, high-sucrose, or low-fat, low-sucrose feeding, HLI was induced in 18-month-old C57BL/6J mice via the surgical ligation of the left femoral artery at 2 locations. Animals were euthanized 4-weeks post-ligation. Results indicate mice with and without obesity shared certain myopathic changes in response to chronic HLI, including impaired muscle contractility, altered mitochondrial electron transport chain complex content and function, and compromised antioxidant defense mechanisms. However, the extent of mitochondrial dysfunction and oxidative stress was significantly greater in obese ischemic muscle compared to non-obese ischemic muscle. Moreover, functional impediments, such as delayed post-surgical recovery of limb function and reduced 6-minute walking distance, as well as accelerated intramuscular protein breakdown, inflammation, cytoskeletal damage, and fibrosis were only evident in mice with obesity. As these features are consistent with human PAD myopathy, our model could be a valuable tool to test new therapeutics.
Collapse
Affiliation(s)
- Emma Fletcher
- Department of Biology, Baylor University, Waco, Texas
| | - Dimitrios Miserlis
- Department of Surgery, University of Texas at Austin Dell Medical School, Austin, Texas
| | | | - Dylan Wilburn
- Department of Health, Human Performance and Recreation, Baylor University, Waco, Texas
| | | | | | | | - Andrew Ring
- Department of Biology, Baylor University, Waco, Texas
| | - Lucas Ferrer
- Department of Surgery, University of Texas at Austin Dell Medical School, Austin, Texas
| | - Gleb Haynatzki
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska
| | - Robert S Smith
- Department of Surgery, Baylor Scott & White Medical Center, Temple, Texas
| | - William T Bohannon
- Department of Surgery, Baylor Scott & White Medical Center, Temple, Texas
| | | |
Collapse
|
9
|
Rontoyanni VG, Blears E, Nunez Lopez O, Ogunbileje J, Moro T, Bhattarai N, Randolph AC, Fry CS, Fankhauser GT, Cheema ZF, Murton AJ, Volpi E, Rasmussen BB, Porter C. Skeletal Muscle Bioenergetics in Critical Limb Ischemia and Diabetes. J Surg Res 2023; 288:108-117. [PMID: 36963297 PMCID: PMC10192034 DOI: 10.1016/j.jss.2023.02.015] [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: 07/26/2022] [Revised: 02/06/2023] [Accepted: 02/19/2023] [Indexed: 03/26/2023]
Abstract
INTRODUCTION Mitochondrial dysfunction is implicated in the metabolic myopathy accompanying peripheral artery disease (PAD) and critical limb ischemia (CLI). Type-2 diabetes mellitus (T2DM) is a major risk factor for PAD development and progression to CLI and may also independently be related to mitochondrial dysfunction. We set out to determine the effect of T2DM in the relationship between CLI and muscle mitochondrial respiratory capacity and coupling control. METHODS We studied CLI patients undergoing revascularization procedures or amputation, and non-CLI patients with or without T2DM of similar age. Mitochondrial respiratory capacity and function were determined in lower limb permeabilized myofibers by high-resolution respirometry. RESULTS Fourteen CLI patients (65 ± 10y) were stratified into CLI patients with (n = 8) or without (n = 6) T2DM and were compared to non-CLI patients with (n = 18; 69 ± 5y) or without (n = 19; 71 ± 6y) T2DM. Presence of CLI but not T2DM had a marked impact on all mitochondrial respiratory states in skeletal muscle, adjusted for the effects of sex. Leak respiration (State 2, P < 0.025 and State 4o, P < 0.01), phosphorylating respiration (P < 0.001), and maximal respiration in the uncoupled state (P < 0.001), were all suppressed in CLI patients, independent of T2DM. T2DM had no significant effect on mitochondrial respiratory capacity and function in adults without CLI. CONCLUSIONS Skeletal muscle mitochondrial respiratory capacity was blunted by ∼35% in patients with CLI. T2DM was not associated with muscle oxidative capacity and did not moderate the relationship between muscle mitochondrial respiratory capacity and CLI.
Collapse
Affiliation(s)
| | - Elizabeth Blears
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Omar Nunez Lopez
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - John Ogunbileje
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Tatiana Moro
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Nisha Bhattarai
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Amanda C Randolph
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas
| | - Christopher S Fry
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Grant T Fankhauser
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Zulfiqar F Cheema
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Andrew J Murton
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Elena Volpi
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Blake B Rasmussen
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Craig Porter
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas; Department of Pediatrics, University of Arkansas for Medical Sciences & Arkansas Children's Research Institute, Little Rock, Arkansas.
| |
Collapse
|
10
|
Markina YV, Kirichenko TV, Tolstik TV, Bogatyreva AI, Zotova US, Cherednichenko VR, Postnov AY, Markin AM. Target and Cell Therapy for Atherosclerosis and CVD. Int J Mol Sci 2023; 24:10308. [PMID: 37373454 DOI: 10.3390/ijms241210308] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Cardiovascular diseases (CVD) and, in particular, atherosclerosis, remain the main cause of death in the world today. Unfortunately, in most cases, CVD therapy begins after the onset of clinical symptoms and is aimed at eliminating them. In this regard, early pathogenetic therapy for CVD remains an urgent problem in modern science and healthcare. Cell therapy, aimed at eliminating tissue damage underlying the pathogenesis of some pathologies, including CVD, by replacing it with various cells, is of the greatest interest. Currently, cell therapy is the most actively developed and potentially the most effective treatment strategy for CVD associated with atherosclerosis. However, this type of therapy has some limitations. In this review, we have tried to summarize the main targets of cell therapy for CVD and atherosclerosis in particular based on the analysis using the PubMed and Scopus databases up to May 2023.
Collapse
Affiliation(s)
- Yuliya V Markina
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Taisiya V Tolstik
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Ulyana S Zotova
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | | | - Anton Yu Postnov
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Alexander M Markin
- Petrovsky National Research Center of Surgery, Moscow 119991, Russia
- Peoples' Friendship University of Russia named after Patrice Lumumba (RUDN University), Moscow 117198, Russia
| |
Collapse
|
11
|
Ferrucci L, Candia J, Ubaida-Mohien C, Lyaskov A, Banskota N, Leeuwenburgh C, Wohlgemuth S, Guralnik JM, Kaileh M, Zhang D, Sufit R, De S, Gorospe M, Munk R, Peterson CA, McDermott MM. Transcriptomic and Proteomic of Gastrocnemius Muscle in Peripheral Artery Disease. Circ Res 2023; 132:1428-1443. [PMID: 37154037 PMCID: PMC10213145 DOI: 10.1161/circresaha.122.322325] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Few effective therapies exist to improve lower extremity muscle pathology and mobility loss due to peripheral artery disease (PAD), in part because mechanisms associated with functional impairment remain unclear. METHODS To better understand mechanisms of muscle impairment in PAD, we performed in-depth transcriptomic and proteomic analyses on gastrocnemius muscle biopsies from 31 PAD participants (mean age, 69.9 years) and 29 age- and sex-matched non-PAD controls (mean age, 70.0 years) free of diabetes or limb-threatening ischemia. RESULTS Transcriptomic and proteomic analyses suggested activation of hypoxia-compensatory mechanisms in PAD muscle, including inflammation, fibrosis, apoptosis, angiogenesis, unfolded protein response, and nerve and muscle repair. Stoichiometric proportions of mitochondrial respiratory proteins were aberrant in PAD compared to non-PAD, suggesting that respiratory proteins not in complete functional units are not removed by mitophagy, likely contributing to abnormal mitochondrial activity. Supporting this hypothesis, greater mitochondrial respiratory protein abundance was significantly associated with greater complex II and complex IV respiratory activity in non-PAD but not in PAD. Rate-limiting glycolytic enzymes, such as hexokinase and pyruvate kinase, were less abundant in muscle of people with PAD compared with non-PAD participants, suggesting diminished glucose metabolism. CONCLUSIONS In PAD muscle, hypoxia induces accumulation of mitochondria respiratory proteins, reduced activity of rate-limiting glycolytic enzymes, and an enhanced integrated stress response that modulates protein translation. These mechanisms may serve as targets for disease modification.
Collapse
Affiliation(s)
- Luigi Ferrucci
- National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Julián Candia
- National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | | | - Alexey Lyaskov
- National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Nirad Banskota
- National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Christiaan Leeuwenburgh
- University of Florida, Institute on Aging, Department of Physiology and Aging, Gainesville, FL, USA
| | - Stephanie Wohlgemuth
- University of Florida, Institute on Aging, Department of Physiology and Aging, Gainesville, FL, USA
| | - Jack M. Guralnik
- University of Maryland School of Medicine, Department of Epidemiology and Public Health, Baltimore, MD, USA
| | - Mary Kaileh
- National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Dongxue Zhang
- Northwestern University Feinberg School of Medicine, Department of Neurology, Chicago, IL, USA
| | - Robert Sufit
- Northwestern University Feinberg School of Medicine, Department of Neurology, Chicago, IL, USA
| | - Supriyo De
- National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Myriam Gorospe
- National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Rachel Munk
- National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Charlotte A. Peterson
- Center for Muscle Biology. College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Mary M. McDermott
- Northwestern University Feinberg School of Medicine, Department of Medicine, Chicago, IL, USA
| |
Collapse
|
12
|
Affiliation(s)
- Ishita Jain
- Department of Cardiothoracic Surgery, Stanford University,
CA, US
- Stanford Cardiovascular Institute, Stanford University, CA,
US
- Center for Tissue Regeneration, Repair and Restoration;
Veterans Affairs Palo Alto Health Care System
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University,
CA, US
- Stanford Cardiovascular Institute, Stanford University, CA,
US
- Center for Tissue Regeneration, Repair and Restoration;
Veterans Affairs Palo Alto Health Care System
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University,
CA, US
- Stanford Cardiovascular Institute, Stanford University, CA,
US
- Center for Tissue Regeneration, Repair and Restoration;
Veterans Affairs Palo Alto Health Care System
- Department of Chemical Engineering, Stanford University,
CA, US
| |
Collapse
|
13
|
Bapat GM, Bashir AZ, Malcolm P, Johanning JM, Pipinos II, Myers SA. A biomechanical perspective on walking in patients with peripheral artery disease. Vasc Med 2023; 28:77-84. [PMID: 36759931 PMCID: PMC9997455 DOI: 10.1177/1358863x221146207] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The most common symptom of peripheral artery disease (PAD) is intermittent claudication, which consists of debilitating leg pain during walking. In clinical settings, the presence of PAD is often noninvasively evaluated using the ankle-brachial index and imaging of the arterial supply. Furthermore, various questionnaires and functional tests are commonly used to measure the severity and negative effect of PAD on quality of life. However, these evaluations only provide information on vascular insufficiency and severity of the disease, but not regarding the complex mechanisms underlying walking impairments in patients with PAD. Biomechanical analyses using motion capture and ground reaction force measurements can provide insight into the underlying mechanisms to walking impairments in PAD. This review analyzes the application of biomechanics tools to identify gait impairments and their clinical implications on rehabilitation of patients with PAD. A total of 18 published journal articles focused on gait biomechanics in patients with PAD were studied. This narriative review shows that the gait of patients with PAD is impaired from the first steps that a patient takes and deteriorates further after the onset of claudication leg pain. These results point toward impaired muscle function across the ankle, knee, and hip joints during walking. Gait analysis helps understand the mechanisms operating in PAD and could also facilitate earlier diagnosis, better treatment, and slower progression of PAD.
Collapse
Affiliation(s)
- Ganesh M Bapat
- Department of Mechanical Engineering, BITS Pilani K K Birla Goa Campus, Goa, India
| | - Ayisha Z Bashir
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE, USA
| | - Philippe Malcolm
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE, USA
| | - Jason M Johanning
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Surgery and Research Service, Omaha VA Medical Center, Omaha, NE, USA
| | - Iraklis I Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Surgery and Research Service, Omaha VA Medical Center, Omaha, NE, USA
| | - Sara A Myers
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE, USA.,Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
14
|
Saini SK, Pérez‐Cremades D, Cheng HS, Kosmac K, Peterson CA, Li L, Tian L, Dong G, Wu KK, Bouverat B, Wohlgemuth SE, Ryan T, Sufit RL, Ferrucci L, McDermott MM, Leeuwenburgh C, Feinberg MW. Dysregulated Genes, MicroRNAs, Biological Pathways, and Gastrocnemius Muscle Fiber Types Associated With Progression of Peripheral Artery Disease: A Preliminary Analysis. J Am Heart Assoc 2022; 11:e023085. [PMID: 36300658 PMCID: PMC9673627 DOI: 10.1161/jaha.121.023085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/08/2022] [Indexed: 02/01/2023]
Abstract
Background Peripheral artery disease (PAD) is associated with gastrocnemius muscle abnormalities. However, the biological pathways associated with gastrocnemius muscle dysfunction and their associations with progression of PAD are largely unknown. This study characterized differential gene and microRNA (miRNA) expression in gastrocnemius biopsies from people without PAD compared with those with PAD. Participants with PAD included those with and without PAD progression. Methods and Results mRNA and miRNA sequencing were performed to identify differentially expressed genes, differentially expressed miRNAs, mRNA-miRNA interactions, and associated biological pathways for 3 sets of comparisons: (1) PAD progression (n=7) versus non-PAD (n=7); (2) PAD no progression (n=6) versus non-PAD; and (3) PAD progression versus PAD no progression. Immunohistochemistry was performed to determine gastrocnemius muscle fiber types and muscle fiber size. Differentially expressed genes and differentially expressed miRNAs were more abundant in the comparison of PAD progression versus non-PAD compared with PAD with versus without progression. Among the top significant cellular pathways in subjects with PAD progression were muscle contraction or development, transforming growth factor-beta, growth/differentiation factor, and activin signaling, inflammation, cellular senescence, and notch signaling. Subjects with PAD progression had increased frequency of smaller Type 2a gastrocnemius muscle fibers in exploratory analyses. Conclusions Humans with PAD progression exhibited greater differences in the number of gene and miRNA expression, biological pathways, and Type 2a muscle fiber size compared with those without PAD. Fewer differences were observed between people with PAD without progression and control patients without PAD. Further study is needed to confirm whether the identified transcripts may serve as potential biomarkers for diagnosis and progression of PAD.
Collapse
Affiliation(s)
- Sunil K. Saini
- All India Institute of Medical Sciences, Department of BiophysicsNew DelhiIndia
| | - Daniel Pérez‐Cremades
- Cardiovascular Division, Department of MedicineBrigham and Women’s Hospital and Harvard Medical SchoolBostonMA
- Department of PhysiologyUniversity of Valencia and INCLIVA Biomedical Research InstituteValenciaSpain
| | - Henry S. Cheng
- Cardiovascular Division, Department of MedicineBrigham and Women’s Hospital and Harvard Medical SchoolBostonMA
| | - Kate Kosmac
- Center for Muscle Biology, College of Health SciencesUniversity of KentuckyLexingtonKY
| | - Charlotte A Peterson
- Center for Muscle Biology, College of Health SciencesUniversity of KentuckyLexingtonKY
| | - Lingyu Li
- Department of Preventive Medicine, Northwestern University Feinberg School of MedicineChicagoIL
| | - Lu Tian
- Department of Health Research and Policy, Stanford UniversityStanfordCA
| | - Gengfu Dong
- Department of Applied Physiology & Kinesiology, University of FloridaGainesvilleFL
| | - Kevin K. Wu
- Department of Aging and Geriatric Research, University of Florida, Institute on AgingGainesvilleFL
| | - Brian Bouverat
- Department of Aging and Geriatric Research, University of Florida, Institute on AgingGainesvilleFL
| | - Stephanie E. Wohlgemuth
- Department of Aging and Geriatric Research, University of Florida, Institute on AgingGainesvilleFL
| | - Terence Ryan
- Department of Applied Physiology & Kinesiology, University of FloridaGainesvilleFL
| | - Robert L. Sufit
- Department of Medicine, Northwestern University Feinberg School of MedicineChicagoIL
| | - Luigi Ferrucci
- Division of Intramural Research, National Institute on AgingBaltimoreMD
| | - Mary M. McDermott
- Department of Preventive Medicine, Northwestern University Feinberg School of MedicineChicagoIL
- Department of Medicine, Northwestern University Feinberg School of MedicineChicagoIL
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, University of Florida, Institute on AgingGainesvilleFL
| | - Mark W. Feinberg
- Cardiovascular Division, Department of MedicineBrigham and Women’s Hospital and Harvard Medical SchoolBostonMA
| |
Collapse
|
15
|
Fallahtafti F, Salamifar Z, Hassan M, Rahman H, Pipinos I, Myers SA. Joint Angle Variability Is Altered in Patients with Peripheral Artery Disease after Six Months of Exercise Intervention. ENTROPY (BASEL, SWITZERLAND) 2022; 24:1422. [PMID: 37420442 PMCID: PMC9602135 DOI: 10.3390/e24101422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/23/2022] [Accepted: 10/01/2022] [Indexed: 07/09/2023]
Abstract
Supervised exercise therapy (SET) is a conservative non-operative treatment strategy for improving walking performance in patients with peripheral artery disease (PAD). Gait variability is altered in patients with PAD, but the effect of SET on gait variability is unknown. Forty-three claudicating patients with PAD underwent gait analysis before and immediately after a 6-month SET program. Nonlinear gait variability was assessed using sample entropy, and the largest Lyapunov exponent of the ankle, knee, and hip joint angle time series. Linear mean and variability of the range of motion time series for these three joint angles were also calculated. Two-factor repeated measure analysis of variance determined the effect of the intervention and joint location on linear and nonlinear dependent variables. After SET, walking regularity decreased, while the stability remained unaffected. Ankle nonlinear variability had increased values compared with the knee and hip joints. Linear measures did not change following SET, except for knee angle, in which the magnitude of variations increased after the intervention. A six-month SET program produced changes in gait variability toward the direction of healthy controls, which indicates that in general, SET improved walking performance in individuals with PAD.
Collapse
Affiliation(s)
- Farahnaz Fallahtafti
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE 6160, USA
| | - Zahra Salamifar
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE 6160, USA
| | - Mahdi Hassan
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE 6160, USA
- Department of Surgery and VA Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Hafizur Rahman
- School of Podiatric Medicine, University of Texas Rio Grande Valley, Harlingen, TX 78550, USA
| | - Iraklis Pipinos
- Department of Surgery and VA Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Sara A Myers
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE 6160, USA
- Department of Surgery and VA Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| |
Collapse
|
16
|
Tu H, Qian J, Zhang D, Barksdale AN, Wadman MC, Pipinos II, Li YL. Different responses of skeletal muscles to femoral artery ligation-induced ischemia identified in BABL/c and C57BL/6 mice. Front Physiol 2022; 13:1014744. [PMID: 36187770 PMCID: PMC9523359 DOI: 10.3389/fphys.2022.1014744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/31/2022] [Indexed: 11/21/2022] Open
Abstract
Peripheral arterial disease (PAD) is a common circulatory problem in lower extremities, and the murine ischemic model is used to reproduce human PAD. To compare strain differences of skeletal muscle responses to ischemia, the left femoral artery was blocked by ligation to reduce blood flow to the limb of BALB/c and C57BL/6 mice. After 6 weeks of the femoral artery ligation, the functional and morphological changes of the gastrocnemius muscle were evaluated. BALB/c mice displayed serious muscular dystrophy, including smaller myofibers (524.3 ± 66 µM2), accumulation of adipose-liked tissue (17.8 ± 0.9%), and fibrosis (6.0 ± 0.5%), compared to C57BL/6 mice (1,328.3 ± 76.3 µM2, 0.27 ± 0.09%, and 1.56 ± 0.06%, respectively; p < 0.05). About neuromuscular junctions (NMJs) in the gastrocnemius muscle, 6 weeks of the femoral artery ligation induced more damage in BALB/c mice than that in C57BL/6 mice, demonstrated by the fragment number of nicotinic acetylcholine receptor (nAChR) clusters (8.8 ± 1.3 in BALB/c vs. 2.5 ± 0.7 in C57BL/6 mice, p < 0.05) and amplitude of sciatic nerve stimulated-endplate potentials (EPPs) (9.29 ± 1.34 mV in BALB/c vs. 20.28 ± 1.42 mV in C57BL/6 mice, p < 0.05). More importantly, 6 weeks of the femoral artery ligation significantly weakened sciatic nerve-stimulated skeletal muscle contraction in BALB/c mice, whereas it didn’t alter the skeletal muscle contraction in C57BL/6 mice. These results suggest that the femoral artery ligation in BALB/c mice is a useful animal model to develop new therapeutic approaches to improve limb structure and function in PAD, although the mechanisms about strain differences of skeletal muscle responses to ischemia are unclear.
Collapse
Affiliation(s)
- Huiyin Tu
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Junliang Qian
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Dongze Zhang
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Aaron N. Barksdale
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michael C. Wadman
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Iraklis I. Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Yu-Long Li,
| |
Collapse
|
17
|
Ismaeel A, Fletcher E, Miserlis D, Wechsler M, Papoutsi E, Haynatzki G, Smith RS, Bohannon WT, Koutakis P. Skeletal muscle MiR-210 expression is associated with mitochondrial function in peripheral artery disease patients. Transl Res 2022; 246:66-77. [PMID: 35288364 PMCID: PMC9197925 DOI: 10.1016/j.trsl.2022.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/10/2022] [Accepted: 03/07/2022] [Indexed: 11/19/2022]
Abstract
Previous studies have demonstrated that circulating microRNA (miR)-210 levels are elevated in peripheral artery disease (PAD) patients. MiR-210 is known to be a negative regulator of mitochondrial respiration; however, the relationship between miR-210 and mitochondrial function has yet to be studied in PAD. We aimed to compare skeletal muscle miR-210 expression of PAD patients to non-PAD controls (CON) and to examine the relationship between miR-210 expression and mitochondrial function. Skeletal muscle biopsies from CON (n = 20), intermittent claudication (IC) patients (n = 20), and critical limb ischemia (CLI) patients (n = 20) were analyzed by high-resolution respirometry to measure mitochondrial respiration of permeabilized fibers. Samples were also analyzed for miR-210 expression by real-time PCR. MiR-210 expression was significantly elevated in IC and CLI muscle compared to CON (P = 0.008 and P < 0.001, respectively). Mitochondrial respiration of electron transport chain (ETC) Complexes II (P = 0.001) and IV (P < 0.001) were significantly reduced in IC patients. Further, CLI patients demonstrated significant reductions in respiration during Complexes I (state 2: P = 0.04, state 3: P = 0.003), combined I and II (P < 0.001), II (P < 0.001), and IV (P < 0.001). The expression of the miR-210 targets, cytochrome c oxidase assembly factor heme A: farnesyltransferase (COX10), and iron-sulfur cluster assembly enzyme (ISCU) were down-regulated in PAD muscle. MiR-210 may play a role in the cellular adaptation to hypoxia and may be involved in the metabolic myopathy associated with PAD.
Collapse
Affiliation(s)
- Ahmed Ismaeel
- Department of Biology, Baylor University, Waco, Texas
| | - Emma Fletcher
- Department of Biology, Baylor University, Waco, Texas
| | - Dimitrios Miserlis
- Department of Surgery, University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Marissa Wechsler
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas
| | | | - Gleb Haynatzki
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska
| | - Robert S Smith
- Department of Surgery, Baylor Scott & White Medical Center, Temple, Texas
| | - William T Bohannon
- Department of Surgery, Baylor Scott & White Medical Center, Temple, Texas
| | | |
Collapse
|
18
|
Ferrari R, Cong G, Chattopadhyay A, Xie B, Assaf E, Morder K, Calderon MJ, Watkins SC, Sachdev U. Attenuated cell-cycle division protein 2 and elevated mitotic roles of polo-like kinase 1 characterize deficient myoblast fusion in peripheral arterial disease. Biochem Biophys Res Commun 2022; 609:163-168. [PMID: 35436627 PMCID: PMC10687717 DOI: 10.1016/j.bbrc.2022.03.161] [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: 02/27/2022] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 11/02/2022]
Abstract
INTRODUCTION We propose that MuSC-derived myoblasts in PAD have transcriptomic differences that can highlight underlying causes of ischemia-induced myopathy. METHODS Differentiation capacity among perfused and ischemic human myoblasts was compared. Following next generation sequencing of mRNA, Ingenuity Pathway Analysis (IPA) was performed for canonical pathway enrichment. Live cell imaging and immunofluorescence were performed to determine myocyte fusion index and protein expression based on insights from IPA, specifically concerning cell cycle regulators including cell-division cycle protein 2 (CDC2) and polo-like kinase 1 (PLK1). RESULTS Ischemic myoblasts formed attenuated myotubes indicative of reduced fusion. Additionally, myoblasts from ischemic segments showed significant differences in canonical pathways associated with PLK1 (upregulated) and G2/M DNA damage checkpoint regulation (downregulated). PLK1 inhibition with BI2536 did not affect cell viability in any group over 24 h but deterred fusion more significantly in PAD myoblasts. Furthermore, PLK1 inhibition reduced the expression of checkpoint protein CDC2 in perfused but not ischemic cells. CONCLUSION Differentiating myoblasts derived from ischemic muscle have significant differences in gene expression including those essential to DNA-damage checkpoint regulation and cell cycle progress. DNA-damage checkpoint dysregulation may contribute to myopathy in PAD.
Collapse
Affiliation(s)
- Ricardo Ferrari
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA
| | - Guangzhi Cong
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA; Department of Cardiology, Cardiovascular Institute, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | | | - B Xie
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA
| | - E Assaf
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA
| | - K Morder
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA
| | | | | | - Ulka Sachdev
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA.
| |
Collapse
|
19
|
Global O-GlcNAcylation changes impact desmin phosphorylation and its partition toward cytoskeleton in C2C12 skeletal muscle cells differentiated into myotubes. Sci Rep 2022; 12:9831. [PMID: 35701470 PMCID: PMC9198038 DOI: 10.1038/s41598-022-14033-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Desmin is the guardian of striated muscle integrity, permitting the maintenance of muscle shape and the efficiency of contractile activity. It is also a key mediator of cell homeostasis and survival. To ensure the fine regulation of skeletal muscle processes, desmin is regulated by post-translational modifications (PTMs). It is more precisely phosphorylated by several kinases connecting desmin to intracellular processes. Desmin is also modified by O-GlcNAcylation, an atypical glycosylation. However, the functional consequence of O-GlcNAcylation on desmin is still unknown, nor its impact on desmin phosphorylation. In a model of C2C12 myotubes, we modulated the global O-GlcNAcylation level, and we determined whether the expression, the PTMs and the partition of desmin toward insoluble material or cytoskeleton were impacted or not. We have demonstrated in the herein paper that O-GlcNAcylation variations led to changes in desmin behaviour. In particular, our data clearly showed that O-GlcNAcylation increase led to a decrease of phosphorylation level on desmin that seems to involve CamKII correlated to a decrease of its partition toward cytoskeleton. Our data showed that phosphorylation/O-GlcNAcylation interplay is highly complex on desmin, supporting that a PTMs signature could occur on desmin to finely regulate its partition (i.e. distribution) with a spatio-temporal regulation.
Collapse
|
20
|
Ferrari R, Xie B, Assaf E, Morder K, Scott M, Liao H, Calderon MJ, Ross M, Loughran P, Watkins SC, Pipinos I, Casale G, Tzeng E, McEnaney R, Sachdev U. Inflammatory Caspase Activity Mediates HMGB1 Release and Differentiation in Myoblasts Affected by Peripheral Arterial Disease. Cells 2022; 11:1163. [PMID: 35406727 PMCID: PMC8997414 DOI: 10.3390/cells11071163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/10/2022] Open
Abstract
Introduction: We previously showed that caspase-1 and -11, which are activated by inflammasomes, mediate recovery from muscle ischemia in mice. We hypothesized that similar to murine models, inflammatory caspases modulate myogenicity and inflammation in ischemic muscle disease. Methods: Caspase activity was measured in ischemic and perfused human myoblasts in response to the NLRP3 and AIM2 inflammasome agonists (nigericin and poly(dA:dT), respectively) with and without specific caspase-1 or pan-caspase inhibition. mRNA levels of myogenic markers and caspase-1 were assessed, and protein levels of caspases-1, -4, -5, and -3 were measured by Western blot. Results: When compared to perfused cells, ischemic myoblasts demonstrated attenuated MyoD and myogenin and elevated caspase-1 mRNA. Ischemic myoblasts also had significantly higher enzymatic caspase activity with poly(dA:dT) (p < 0.001), but not nigericin stimulation. Inhibition of caspase activity including caspase-4/-5, but not caspase-1, blocked activation effects of poly(dA:dT). Ischemic myoblasts had elevated cleaved caspase-5. Inhibition of caspase activity deterred differentiation in ischemic but not perfused myoblasts and reduced the release of HMGB1 from both groups. Conclusion: Inflammatory caspases can be activated in ischemic myoblasts by AIM2 and influence ischemic myoblast differentiation and release of pro-angiogenic HMGB1. AIM2 inflammasome involvement suggests a role as a DNA damage sensor, and our data suggest that caspase-5 rather than caspase-1 may mediate the downstream mediator of this pathway.
Collapse
Affiliation(s)
- Ricardo Ferrari
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Bowen Xie
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Edwyn Assaf
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Kristin Morder
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Melanie Scott
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Hong Liao
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Michael J. Calderon
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Mark Ross
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Patricia Loughran
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Simon C. Watkins
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Iraklis Pipinos
- Department of Surgery, University of Nebraska, Omaha, NE 68198, USA; (I.P.); (G.C.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - George Casale
- Department of Surgery, University of Nebraska, Omaha, NE 68198, USA; (I.P.); (G.C.)
| | - Edith Tzeng
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - Ryan McEnaney
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - Ulka Sachdev
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| |
Collapse
|
21
|
Muscle forces and power are significantly reduced during walking in patients with peripheral artery disease. J Biomech 2022; 135:111024. [PMID: 35248803 PMCID: PMC9064980 DOI: 10.1016/j.jbiomech.2022.111024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 11/21/2022]
Abstract
Patients with peripheral artery disease (PAD) have significantly reduced lower extremity muscle strength compared with healthy individuals as measured during isolated, single plane joint motion by isometric and isokinetic strength dynamometers. Alterations to the force contribution of muscles during walking caused by PAD are not well understood. Therefore, this study used simulations with PAD biomechanics data to understand lower extremity muscle functions in patients with PAD during walking and to compare that with healthy older individuals. A total of 12 patients with PAD and 10 age-matched healthy older controls walked across a 10-meter pathway with reflective markers on their lower limbs. Marker coordinates and ground reaction forces were recorded and exported to OpenSim software to perform gait simulations. Walking velocity, joint angles, muscle force, muscle power, and metabolic rate were calculated and compared between patients with PAD and healthy older controls. Our results suggest that patients with PAD walked slower with less hip extension during propulsion. Significant force and power reductions were observed in knee extensors during weight acceptance and in plantar flexors and hip flexors during propulsion in patients with PAD. The estimated metabolic rate of walking during stance was not different between patients with PAD and controls. This study is the first to analyze lower limb muscular responses during walking in patients with PAD using the OpenSim simulation software. The simulation results of this study identified important information about alterations to muscle force and power during walking in those with PAD.
Collapse
|
22
|
Wilburn D, Ismaeel A, Machek S, Fletcher E, Koutakis P. Shared and distinct mechanisms of skeletal muscle atrophy: A narrative review. Ageing Res Rev 2021; 71:101463. [PMID: 34534682 DOI: 10.1016/j.arr.2021.101463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 12/15/2022]
Abstract
Maintenance of skeletal muscle mass and function is an incredibly nuanced balance of anabolism and catabolism that can become distorted within different pathological conditions. In this paper we intend to discuss the distinct intracellular signaling events that regulate muscle protein atrophy for a given clinical occurrence. Aside from the common outcome of muscle deterioration, several conditions have at least one or more distinct mechanisms that creates unique intracellular environments that facilitate muscle loss. The subtle individuality to each of these given pathologies can provide both researchers and clinicians with specific targets of interest to further identify and increase the efficacy of medical treatments and interventions.
Collapse
Affiliation(s)
- Dylan Wilburn
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Ahmed Ismaeel
- Department of Biology, Baylor University, Waco, TX 76706, USA
| | - Steven Machek
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Emma Fletcher
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; Department of Biology, Baylor University, Waco, TX 76706, USA
| | | |
Collapse
|
23
|
Syed MH, Zamzam A, Khan H, Singh K, Forbes TL, Rotstein O, Abdin R, Eikelboom J, Qadura M. Fatty acid binding protein 3 is associated with peripheral arterial disease. JVS Vasc Sci 2021; 1:168-175. [PMID: 34617045 PMCID: PMC8489205 DOI: 10.1016/j.jvssci.2020.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022] Open
Abstract
Background Peripheral arterial disease (PAD) affects more than 150 million people worldwide and is associated with high rates of lower extremity amputation, myocardial infarction, stroke and death. Fatty acid binding protein 3 (FABP3) is released into circulation in patients with skeletal muscle injury. In this pilot study, we investigated a possible association between PAD and blood levels of FABP3. Methods Blood samples were collected from patients with clinical symptoms and diagnostic findings indicative of PAD (PAD group; ankle-brachial index [ABI] <0.9; n = 75) and in those without clinical or diagnostic features of PAD (non-PAD group; ABI >0.9; n = 75) presenting to vascular surgery ambulatory clinics at St. Michael's Hospital. Plasma samples were analyzed by protein multiplex to quantify FABP3 levels. Results PAD patients were found to have higher blood levels of FABP3 compared to patients without PAD (mean 3.90 ± 1.69 vs 2.03 ± 0.78; P < .001). A subgroup analysis demonstrated that the FABP3 levels were increased by almost two-fold in patients with PAD, independent of coronary artery disease (P < .001) or diabetes mellitus status (P < .001). Moreover, a significant negative correlation between FABP3 and the ABI was observed in PAD and patients without PAD matched groups (r = –0.51; P = .001). Last, immunohistochemistry demonstrated elevated expressions of FABP3 within skeletal muscle obtained from patients with the most severe form of PAD, chronic limb-threatening ischemia, when compared with patients without PAD. Conclusions Patients with PAD have elevated plasma levels of FABP3. An increasing severity of PAD is associated with higher FABP3 levels. There is a pressing need for a simple, readily accessible, blood-based biomarker for PAD. In this study, we found elevated levels of FABP3 in patients with PAD. This increase in FABP3 was irrespective of history of coronary artery disease or diabetes. Furthermore, our data suggest that an increasing severity of PAD is associated with higher FABP3 levels. Subsequently, FABP3 may be a potential diagnostic biomarker for PAD. However, further studies are needed to confirm the capability of FABP3 to serve as a valid and reliable biomarker for PAD.
Collapse
Affiliation(s)
- Muzammil H Syed
- Division of Vascular Surgery, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Abdelrahman Zamzam
- Division of Vascular Surgery, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Hamzah Khan
- Division of Vascular Surgery, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Krishna Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Thomas L Forbes
- Division of Vascular Surgery, Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Ori Rotstein
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Rawand Abdin
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - John Eikelboom
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Li C, Nie F, Liu X, Chen M, Chi D, Li S, Pipinos II, Li X. Antioxidative and Angiogenic Hyaluronic Acid-Based Hydrogel for the Treatment of Peripheral Artery Disease. ACS APPLIED MATERIALS & INTERFACES 2021; 13:45224-45235. [PMID: 34519480 DOI: 10.1021/acsami.1c11349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Peripheral arterial disease (PAD) is a progressive atherosclerotic disorder characterized by blockages of the arteries supplying the lower extremities. Ischemia initiates oxidative damage and mitochondrial dysfunction in the legs of PAD patients, causing injury to the tissues of the leg, significant decline in walking performance, leg pain while walking, and in the most severe cases, nonhealing ulcers and gangrene. Current clinical trials based on cells/stem cells, the trophic factor, or gene therapy systems have shown some promising results for the treatment of PAD. Biomaterial matrices have been explored in animal models of PAD to enhance these therapies. However, current biomaterial approaches have not fully met the essential requirements for minimally invasive intramuscular delivery to the leg. Ideally, a biomaterial should present properties to ameliorate oxidative stress/damage and failure of angiogenesis. Recently, we have created a thermosensitive hyaluronic acid (HA) hydrogel with antioxidant capacity and skeletal muscle-matching stiffness. Here, we further optimized HA hydrogels with the cell adhesion peptide RGD to facilitate the development of vascular-like structures in vitro. The optimized HA hydrogel reduced intracellular reactive oxygen species levels and preserved vascular-like structures against H2O2-induced damage in vitro. HA hydrogels also provided prolonged release of the vascular endothelial growth factor (VEGF). After injection into rat ischemic hindlimb muscles, this VEGF-releasing hydrogel reduced lipid oxidation, regulated oxidative-related genes, enhanced local blood flow in the muscle, and improved running capacity of the treated rats. Our HA hydrogel system holds great potential for the treatment of the ischemic legs of patients with PAD.
Collapse
Affiliation(s)
- Cui Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Fujiao Nie
- Hunan Engineering Technology Research Center for the Prevention and Treatment of Otorhinolaryngologic Diseases and Protection of Visual Function with Chinese Medicine, Human University of Chinese Medicine, Changsha, Hunan 410208, China
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Xiaoyan Liu
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Meng Chen
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - David Chi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Shuai Li
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Iraklis I Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Xiaowei Li
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
25
|
da Silva GO, Correia MDA, de Oliveira PLM, Farah BQ, Vianna LC, Puech-Leão P, Wolosker N, Cucato GG, Ritti-Dias RM. Are Vascular Parameters Associated with Walking Impairment in Patients with Claudication? Ann Vasc Surg 2021; 77:31-37. [PMID: 34455045 DOI: 10.1016/j.avsg.2021.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/30/2021] [Accepted: 06/05/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND The mechanisms underlying functional impairments in symptomatic PAD patients are controversial and poorly understood. Endothelial dysfunction and arterial stiffness have been proposed as potential mechanisms related to functional impairment in symptomatic PAD patients, however, more studies are needed to confirm these associations. OBJECTIVE To analyze the association between vascular function and walking impairment in patients with peripheral arterial disease (PAD) and symptoms of claudication. METHODS This was a cross-sectional study that included 68 patients with symptomatic PAD. All patients underwent an objective (Six-minute walk test [6MWT], 4-meter walk test) and a subjective (Walking Impairment Questionnaire [WIQ]) measurement of walking impairment. Vascular parameters measured were pulse-wave velocity (PWV) and flow-mediated dilation (FMD). Multiple linear regression was performed to investigate the association among walking impairment variables with vascular function parameters. RESULTS No significant associations between the claudication onset distance (PWV: b=.060, P = 0.842; FMD: b=-.192, P = 0.456), 6MWT (PWV: b=.007, P = 0..975; FMD: b=.090, P = 0.725), WIQ distance (PWV: b=.337, P = 0.117; FMD: b=-.025, P = 0.895) WIQ speed (PWV: b=.320, P = 0.181; FMD: b=-.028, P = 0.497), WIQ stairs (PWV: b=.256, P = 0.204; FMD: b=-.228, P = 0.230), 4-meter usual walk (PWV: b=-.421, P = 0.107; FMD: b=-.338, P = 0.112), 4-meter fast walk (PWV: b=-.496, P = 0.063; FMD: b=-.371, P = 0.086) and vascular function were found. CONCLUSIONS In symptomatic PAD patients, vascular function is not associated to walking impairment, even when adjusting for comorbid conditions and diabetes.
Collapse
Affiliation(s)
| | | | | | - Breno Quintella Farah
- Physical Education Department, Rural Federal University of Pernambuco, Recife-PE, Brazil.; Graduate Program in Physical Education, Federal University of Pernambuco (UFPE), Recife, Brazil
| | - Lauro C Vianna
- NeuroV̇ASQ̇ - Integrative Physiology Laboratory, Faculty of Physical Education, University of Brasilia, Brasilia-DF, Brazil
| | - Pedro Puech-Leão
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo-SP, Brazil
| | | | | | | |
Collapse
|
26
|
McDermott MM, Dayanidhi S, Kosmac K, Saini S, Slysz J, Leeuwenburgh C, Hartnell L, Sufit R, Ferrucci L. Walking Exercise Therapy Effects on Lower Extremity Skeletal Muscle in Peripheral Artery Disease. Circ Res 2021; 128:1851-1867. [PMID: 34110902 DOI: 10.1161/circresaha.121.318242] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Walking exercise is the most effective noninvasive therapy that improves walking ability in peripheral artery disease (PAD). Biologic mechanisms by which exercise improves walking in PAD are unclear. This review summarizes evidence regarding effects of walking exercise on lower extremity skeletal muscle in PAD. In older people without PAD, aerobic exercise improves mitochondrial activity, muscle mass, capillary density, and insulin sensitivity in skeletal muscle. However, walking exercise increases lower extremity ischemia in people with PAD, and therefore, mechanisms by which this exercise improves walking may differ between people with and without PAD. Compared with people without PAD, gastrocnemius muscle in people with PAD has greater mitochondrial impairment, increased reactive oxygen species, and increased fibrosis. In multiple small trials, walking exercise therapy did not consistently improve mitochondrial activity in people with PAD. In one 12-week randomized trial of people with PAD randomized to supervised exercise or control, supervised treadmill exercise increased treadmill walking time from 9.3 to 15.1 minutes, but simultaneously increased the proportion of angular muscle fibers, consistent with muscle denervation (from 7.6% to 15.6%), while angular myofibers did not change in the control group (from 9.1% to 9.1%). These findings suggest an adaptive response to exercise in PAD that includes denervation and reinnervation, an adaptive process observed in skeletal muscle of people without PAD during aging. Small studies have not shown significant effects of exercise on increased capillary density in lower extremity skeletal muscle of participants with PAD, and there are no data showing that exercise improves microcirculatory delivery of oxygen and nutrients in patients with PAD. However, the effects of supervised exercise on increased plasma nitrite abundance after a treadmill walking test in people with PAD may be associated with improved lower extremity skeletal muscle perfusion and may contribute to improved walking performance in response to exercise in people with PAD. Randomized trials with serial, comprehensive measures of muscle biology, and physiology are needed to clarify mechanisms by which walking exercise interventions improve mobility in PAD.
Collapse
Affiliation(s)
- Mary M McDermott
- Department of Medicine and Preventive Medicine (M.M.M., J.S.), Northwestern University Feinberg School of Medicine
| | - Sudarshan Dayanidhi
- Shirley Ryan Ability Laboratory (S.D.), Northwestern University Feinberg School of Medicine
| | - Kate Kosmac
- Center for Muscle Biology, University of Kentucky (K.K.)
| | - Sunil Saini
- Jawaharlal Nehru University, School of Biotechnology, New Delhi, India (S.S.)
| | - Joshua Slysz
- Department of Medicine and Preventive Medicine (M.M.M., J.S.), Northwestern University Feinberg School of Medicine
| | | | - Lisa Hartnell
- Division of Intramural Research, National Institute on Aging (L.H., L.F.)
| | - Robert Sufit
- Department of Neurology (R.S.), Northwestern University Feinberg School of Medicine
| | - Luigi Ferrucci
- Division of Intramural Research, National Institute on Aging (L.H., L.F.)
| |
Collapse
|
27
|
Yu JA, Wang Z, Yang X, Ma M, Li Z, Nie Q. LncRNA-FKBP1C regulates muscle fiber type switching by affecting the stability of MYH1B. Cell Death Discov 2021; 7:73. [PMID: 33837177 PMCID: PMC8035166 DOI: 10.1038/s41420-021-00463-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/12/2021] [Accepted: 03/25/2021] [Indexed: 01/17/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are well-known to participate in a variety of important regulatory processes in myogenesis. In our previous RNA-seq study (accession number GSE58755), we found that lncRNA-FKBP1C was differentially expressed between White Recessive Rock (WRR) and Xinghua (XH) chicken. Here, we have further demonstrated that lncRNA-FKBP1C interacted directly with MYH1B by biotinylated RNA pull-down assay and RNA immunoprecipitation (RIP). Protein stability and degradation experiments identified that lncRNA-FKBP1C enhanced the protein stability of MYH1B. Overexpression of lncRNA-FKBP1C inhibited myoblasts proliferation, promoted myoblasts differentiation, and participated in the formation of skeletal muscle fibers. LncRNA-FKBP1C could downregulate the fast muscle genes and upregulate slow muscle genes. Conversely, its interference promoted cell proliferation, repressed cell differentiation, and drove the transformation of slow-twitch muscle fibers to fast-twitch muscle fibers. Similar results were observed after knockdown of the MYH1B gene, but the difference was that the MYH1B gene had no effects on fast muscle fibers. In short, these data demonstrate that lncRNA-FKBP1C could bound with MYH1B and enhance its protein stability, thus affecting proliferation, differentiation of myoblasts and conversion of skeletal muscle fiber types.
Collapse
Affiliation(s)
- Jia-Ao Yu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China.,National-Local Joint Engineering Research Center for Livestock Breeding, Guangzhou, China
| | - Zhijun Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China.,National-Local Joint Engineering Research Center for Livestock Breeding, Guangzhou, China
| | - Xin Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China.,National-Local Joint Engineering Research Center for Livestock Breeding, Guangzhou, China
| | - Manting Ma
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China.,National-Local Joint Engineering Research Center for Livestock Breeding, Guangzhou, China
| | - Zhenhui Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China.,National-Local Joint Engineering Research Center for Livestock Breeding, Guangzhou, China
| | - Qinghua Nie
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China. .,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China. .,National-Local Joint Engineering Research Center for Livestock Breeding, Guangzhou, China.
| |
Collapse
|
28
|
Gait variability is affected more by peripheral artery disease than by vascular occlusion. PLoS One 2021; 16:e0241727. [PMID: 33788839 PMCID: PMC8011739 DOI: 10.1371/journal.pone.0241727] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/15/2021] [Indexed: 11/19/2022] Open
Abstract
Background Patients with peripheral artery disease with intermittent claudication (PAD-IC) have altered gait variability from the first step they take, well before the onset of claudication pain. The mechanisms underlying these gait alterations are poorly understood. Aims To determine the effect of reduced blood flow on gait variability by comparing healthy older controls and patients with PAD-IC. We also determined the diagnostic value of gait variability parameters to identify the presence of PAD. Methods A cross-sectional cohort design was used. Thirty healthy older controls and thirty patients with PAD-IC walked on a treadmill at their self-selected speed in pain free walking (normal walking for healthy older controls; prior to claudication onset for PAD) and reduced blood flow (post vascular occlusion with thigh tourniquet for healthy older controls; pain for PAD) conditions. Gait variability was assessed using the largest Lyapunov exponent, approximate entropy, standard deviation, and coefficient of variation of ankle, knee, and hip joints range of motion. Receiver operating characteristics curve analyses of the pain free walking condition were performed to determine the optimal cut-off values for separating individuals with PAD-IC from those without PAD-IC. Results and discussion Patients with PAD-IC have increased amount of variability for knee and hip ranges of motion compared with the healthy older control group. Regarding the main effect of condition, reduced blood flow demonstrated increased amount of variability compared with pain free walking. Significant interactions between group and condition at the ankle show increased values for temporal structure of variability, but a similar amount of variability in the reduced blood flow condition. This demonstrates subtle interactions in the movement patterns remain distinct between PAD-IC versus healthy older controls during the reduced blood flow condition. A combination of gait variability parameters correctly identifies PAD-IC disease 70% of the time or more. Conclusions Gait variability is affected both by PAD and by the mechanical induction of reduced blood flow. Gait variability parameters have potential diagnostic ability, as some measures had 90.0% probability of correctly identifying patients with PAD-IC.
Collapse
|
29
|
Li C, Kitzerow O, Nie F, Dai J, Liu X, Carlson MA, Casale GP, Pipinos II, Li X. Bioengineering strategies for the treatment of peripheral arterial disease. Bioact Mater 2021; 6:684-696. [PMID: 33005831 PMCID: PMC7511653 DOI: 10.1016/j.bioactmat.2020.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/12/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
Peripheral arterial disease (PAD) is a progressive atherosclerotic disorder characterized by narrowing and occlusion of arteries supplying the lower extremities. Approximately 200 million people worldwide are affected by PAD. The current standard of operative care is open or endovascular revascularization in which blood flow restoration is the goal. However, many patients are not appropriate candidates for these treatments and are subject to continuous ischemia of their lower limbs. Current research in the therapy of PAD involves developing modalities that induce angiogenesis, but the results of simple cell transplantation or growth factor delivery have been found to be relatively poor mainly due to difficulties in stem cell retention and survival and rapid diffusion and enzymolysis of growth factors following injection of these agents in the affected tissues. Biomaterials, including hydrogels, have the capability to protect stem cells during injection and to support cell survival. Hydrogels can also provide a sustained release of growth factors at the injection site. This review will focus on biomaterial systems currently being investigated as carriers for cell and growth factor delivery, and will also discuss biomaterials as a potential stand-alone method for the treatment of PAD. Finally, the challenges of development and use of biomaterials systems for PAD treatment will be reviewed.
Collapse
Affiliation(s)
- Cui Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Oliver Kitzerow
- Department of Genetics Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Fujiao Nie
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Jingxuan Dai
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaoyan Liu
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Omaha VA Medical Center, Omaha, NE, 68105, United States
| | - George P. Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Iraklis I. Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaowei Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| |
Collapse
|
30
|
Casale GP, Thompson JR, Carpenter LC, Kim J, Lackner TJ, Mietus CJ, Ha DM, Myers SA, Brunette KE, Li S, Shields C, Willcockson G, Pipinos II. Cytokine signature of inflammation mediated by autoreactive Th-cells, in calf muscle of claudicating patients with Fontaine stage II peripheral artery disease. Transl Res 2021; 228:94-108. [PMID: 32835907 PMCID: PMC7779738 DOI: 10.1016/j.trsl.2020.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/21/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023]
Abstract
Peripheral artery disease (PAD), a severe atherosclerotic condition primarily of the elderly, afflicts 200 million individuals, worldwide, and is associated with lower extremity myopathy. Circulating markers of inflammation have been linked to risk and severity of PAD but the contribution of local inflammation to myopathy remains unknown. We evaluated, by ELISA, calf muscle of PAD patients (N = 23) and control subjects (N = 18) for local expression of inflammatory cytokines including Granulocyte/Monocyte Colony-Stimulating Factor (GM-CSF), Interleukin 17A (IL-17A), Interferon ϒ (IFN-ϒ), tumor necrosis factor α (TNF-α), and Interleukin 6 (IL-6). One or more of these cytokines were expressed in nineteen patients and 2 controls and coordinated expression of GM-CSF, IL-17A, IFN-ϒ, and TNF-α, a signature of activated, MHC Class II dependent autoreactive Th-cells, was unique to 11 patients. GM-CSF is the central driver of tissue-damaging myeloid macrophages. Patients with this cytokine signature had a shorter (P= 0.017) Claudication Onset Distance (17 m) compared with patients lacking the signature (102 m). Transforming Growth Factor β1 (TGFβ1) and Chemokine Ligand 5 (CCL5) were expressed coordinately in all PAD and control muscles, independently of GM-CSF, IL-17A, IFN-ϒ, TNF-α, or IL-6. TGFβ1 and CCL5 and their gene transcripts were increased in PAD muscle, consistent with increased age-associated inflammation in these patients. Serum cytokines were not informative of muscle cytokine expression. We have identified a cytokine profile of autoimmune inflammation in calf muscles of a significant proportion of claudicating PAD patients, in association with decreased limb function, and a second independent profile consistent with increased "inflammaging" in all PAD patients.
Collapse
Affiliation(s)
- George P Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska.
| | - Jonathan R Thompson
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Lauren C Carpenter
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Julian Kim
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Timothy J Lackner
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Constance J Mietus
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Duy M Ha
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sara A Myers
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, Nebraska
| | | | - Shuai Li
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Christina Shields
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Gregory Willcockson
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Iraklis I Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
31
|
Barlow J, Sfyri PP, Mitchell R, Verpoorten S, Scully D, Andreou C, Papadopoulos P, Patel K, Matsakas A. Platelet releasate normalises the compromised muscle regeneration in a mouse model of hyperlipidaemia. Exp Physiol 2021; 106:700-713. [PMID: 33450106 DOI: 10.1113/ep088937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the impact of obesity-independent hyperlipidaemia on skeletal muscle stem cell function of ApoE-deficient (ApoE-/- ) mice? What is the main finding and its importance? Compromised muscle stem cell function accounts for the impaired muscle regeneration in hyperlipidaemic ApoE-/- mice. Importantly, impaired muscle regeneration is normalised by administration of platelet releasate. ABSTRACT Muscle satellite cells are important stem cells for skeletal muscle regeneration and repair after injury. ApoE-deficient mice, an established mouse model of hyperlipidaemia and atherosclerosis, show evidence of oxidative stress-induced lesions and fat infiltration in skeletal muscle followed by impaired repair after injury. However, the mechanisms underpinning attenuated muscle regeneration remain to be fully defined. Key to addressing the latter is to understand the properties of muscle stem cells from ApoE-deficient mice and their myogenic potential. Muscle stem cells from ApoE-deficient mice were cultured both ex vivo (on single fibres) and in vitro (primary myoblasts) and their myogenic capacity was determined. Skeletal muscle regeneration was studied on days 5 and 10 after cardiotoxin injury. ApoE-deficient muscle stem cells showed delayed activation and differentiation on single muscle fibres ex vivo. Impaired proliferation and differentiation profiles were also evident on isolated primary muscle stem cells in culture. ApoE-deficient mice displayed impaired skeletal muscle regeneration after acute injury in vivo. Administration of platelet releasate in ApoE-deficient mice reversed the deficits of muscle regeneration after acute injury to wild-type levels. These findings indicate that muscle stem cell myogenic potential is perturbed in skeletal muscle of a mouse model of hyperlipidaemia. We propose that platelet releasate could be a therapeutic intervention for conditions with associated myopathy such as peripheral arterial disease.
Collapse
Affiliation(s)
- Joseph Barlow
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Pagona Panagiota Sfyri
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Rob Mitchell
- School of Biological Sciences, University of Reading, Reading, UK
| | - Sandrine Verpoorten
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - David Scully
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Charalampos Andreou
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Petros Papadopoulos
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| |
Collapse
|
32
|
McDermott MM, Ferrucci L, Gonzalez-Freire M, Kosmac K, Leeuwenburgh C, Peterson CA, Saini S, Sufit R. Skeletal Muscle Pathology in Peripheral Artery Disease: A Brief Review. Arterioscler Thromb Vasc Biol 2020; 40:2577-2585. [PMID: 32938218 DOI: 10.1161/atvbaha.120.313831] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This brief review summarizes current evidence regarding lower extremity peripheral artery disease (PAD) and lower extremity skeletal muscle pathology. Lower extremity ischemia is associated with reduced calf skeletal muscle area and increased calf muscle fat infiltration and fibrosis on computed tomography or magnetic resonance imaging. Even within the same individual, the leg with more severe ischemia has more adverse calf muscle characteristics than the leg with less severe ischemia. More adverse computed tomography-measured calf muscle characteristics, such as reduced calf muscle density, are associated with higher rates of mobility loss in people with PAD. Calf muscle in people with PAD may also have reduced mitochondrial activity compared with those without PAD, although evidence is inconsistent. Muscle biopsy document increased oxidative stress in PAD. Reduced calf muscle perfusion, impaired mitochondrial activity, and smaller myofibers are associated with greater walking impairment in PAD. Preliminary evidence suggests that calf muscle pathology in PAD may be reversible. In a small uncontrolled trial, revascularization improved both the ankle-brachial index and mitochondrial activity, measured by calf muscle phosphocreatine recovery time. A pilot clinical trial showed that cocoa flavanols increased measures of myofiber health, mitochondrial activity, and capillary density while simultaneously improving 6-minute walk distance in PAD. Calf muscle pathological changes are associated with impaired walking performance in people with PAD, and interventions that both increase calf perfusion and improve calf muscle health are promising therapies to improve walking performance in PAD.
Collapse
Affiliation(s)
- Mary M McDermott
- Department of Medicine and Preventive Medicine (M.M.M.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Luigi Ferrucci
- Division of Intramural Research, National Institute on Aging, Baltimore, MD (L.F.)
| | - Marta Gonzalez-Freire
- Health Research Institute of the Balearic Islands (IdISBa), Vascular and Metabolic Pathologies Group, Spain (M.G.-F.)
| | - Kate Kosmac
- Department of Physical Therapy, University of Kentucky Center for Muscle Biology, Lexington (K.K., C.A.P.)
| | | | - Charlotte A Peterson
- Department of Physical Therapy, University of Kentucky Center for Muscle Biology, Lexington (K.K., C.A.P.)
| | - Sunil Saini
- Department of Aging and Geriatric Research, University of Florida, Gainesville (C.L., S.S.)
| | - Robert Sufit
- Department of Neurology (R.S.), Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
33
|
Chen M, Li C, Nie F, Liu X, Pipinos II, Li X. Synthesis and characterization of a hyaluronic acid-based hydrogel with antioxidative and thermosensitive properties. RSC Adv 2020; 10:33851-33860. [PMID: 35519025 PMCID: PMC9056774 DOI: 10.1039/d0ra07208g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/04/2020] [Indexed: 02/01/2023] Open
Abstract
Peripheral arterial disease (PAD) is initiated by progressive atherosclerotic blockages of the arteries supplying the lower extremities. The most common presentation of PAD is claudication (leg pain and severe walking limitation), with many patients progressing to limb threatening ischemia and amputation. Biomaterial approaches are just beginning to be explored in the therapy of PAD with different materials now being evaluated for the delivery of cells or growth factors in animal models of PAD. A biomaterial matrix optimized for minimally invasive injection in the ischemic leg muscles of patients with PAD is urgently needed. There are several important requirements for optimal delivery, retention, and performance of a biomaterial matrix in the mechanically, histologically, and biochemically dynamic intramuscular environment of the PAD leg. Ideally, the material should have mechanical properties matching those of the recipient muscle, undergo minimal swelling, and should introduce properties that can ameliorate the mechanisms operating in PAD like oxidative stress and damage. Here we have developed an injectable, antioxidative, and thermosensitive hydrogel system based on hyaluronic acid (HA). We first synthesized a unique crosslinker of disulfide-modified poloxamer F127 diacrylate. This crosslinker led to the creation of a thermosensitive HA hydrogel with minimal swelling and muscle-matching mechanical properties. We introduced unique disulfide groups into hydrogels which functioned as an effective reactive oxygen species scavenger, exhibited hydrogen peroxide (H2O2)-responsive degradation, and protected cells against H2O2-induced damage. Our antioxidative thermosensitive HA hydrogel system holds great potential for the treatment of the ischemic legs of patients with PAD.
Collapse
Affiliation(s)
- Meng Chen
- Mary & Dick Holland Regenerative Medicine Program, Department of Neurological Sciences, University of Nebraska Medical Center Omaha NE 68198 USA
| | - Cui Li
- Mary & Dick Holland Regenerative Medicine Program, Department of Neurological Sciences, University of Nebraska Medical Center Omaha NE 68198 USA
| | - Fujiao Nie
- Mary & Dick Holland Regenerative Medicine Program, Department of Neurological Sciences, University of Nebraska Medical Center Omaha NE 68198 USA
| | - Xiaoyan Liu
- Mary & Dick Holland Regenerative Medicine Program, Department of Neurological Sciences, University of Nebraska Medical Center Omaha NE 68198 USA
| | - Iraklis I Pipinos
- Department of Surgery, University of Nebraska Medical Center Omaha NE 68198 USA
| | - Xiaowei Li
- Mary & Dick Holland Regenerative Medicine Program, Department of Neurological Sciences, University of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|
34
|
Pizzimenti M, Meyer A, Charles A, Giannini M, Chakfé N, Lejay A, Geny B. Sarcopenia and peripheral arterial disease: a systematic review. J Cachexia Sarcopenia Muscle 2020; 11:866-886. [PMID: 32648665 PMCID: PMC7432591 DOI: 10.1002/jcsm.12587] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/31/2020] [Accepted: 02/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Patients with lower extremity peripheral arterial disease (PAD) and sarcopenia are a population at risk requiring specific and targeted care. The aim of this review is to gather all relevant studies associating sarcopenia and PAD and to identify the underlying pathophysiological mechanisms as well as potential therapeutic strategies to improve skeletal muscle function. METHODS A systematic review was carried out following the recommendations of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). RESULTS Data extraction allowed the evaluation of 140 publications; 87 met the inclusion criteria; of which 79 were included in the final review, reporting sufficient data for epidemiological and diagnostic criteria, mechanical analysis, and therapeutic approaches. Epidemiological analysis and diagnostic criteria were based on 18 studies following 2362 PAD patients [31.39% (SD 7.61) women], aged 72.42 (SD 2.84); sarcopenia was present in 34.63% (SD 12.86) of the patients. Mechanical and pathway analysis were based on five animal studies and 29 clinical reports, showing significantly altered muscle strength and function in 1352 PAD patients [26.49% (SD 17.32) women], aged 67.67 (SD 5.14) years; impaired muscle histology in 192 PAD patients (9.2% (SD 11.22) women), aged 64.3 (SD 0.99) years; +58.63% (SD 25.48) of oxidative stress in 69 PAD patients [16.96% (SD 8.10) women], aged 63.17 (SD 1.43) years; mitochondriopathy in 153 PAD patients [29.39% (SD 28.27) women], aged 63.50 (SD 1.83) years; +15.58% (SD 7.41) of inflammation in 900 PAD patients [40.77% (SD 3.71) women], aged 74.88 (SD 2.76) years; and altered signalling pathways in 51 PAD patients [34.45% (SD 32.23) women], aged 72.25 (SD 5.25) years. Therapeutic approaches analysis was based on seven animal studies and 21 clinical reports. In total, 884 patients followed an exercise therapy, and 18 received an angiogenesis treatment; 30.84% (SD 17.74) were women. Mean ages of patients studied were 66.85 (SD 3.96). CONCLUSIONS Sarcopenia and lower extremity PAD have musculoskeletal consequences that directly impair patients' quality of life and prognosis. Although PAD is primarily a vascular disease, all etiological factors of sarcopenia identified so far are present in PAD. Indeed, both sarcopenia and PAD are accompanied by oxidative stress, skeletal muscle mitochondrial impairments, inflammation, inhibition of specific pathways regulating muscle synthesis or protection (i.e. IGF-1, RISK, and SAFE), and activation of molecules associated with muscle degradation. To date, besides revascularization, the best therapeutic strategy includes exercise, but approaches targeting the underlying mechanisms still deserve further studies.
Collapse
Affiliation(s)
- Mégane Pizzimenti
- FMTS, Department of Physiology, EA3072 Mitochondria, Oxidative Stress and Muscular ProtectionUniversity of StrasbourgStrasbourgFrance
- Department of Physiology and Functional ExplorationsUniversity Hospital of StrasbourgStrasbourgFrance
| | - Alain Meyer
- FMTS, Department of Physiology, EA3072 Mitochondria, Oxidative Stress and Muscular ProtectionUniversity of StrasbourgStrasbourgFrance
- Department of Physiology and Functional ExplorationsUniversity Hospital of StrasbourgStrasbourgFrance
| | - Anne‐Laure Charles
- FMTS, Department of Physiology, EA3072 Mitochondria, Oxidative Stress and Muscular ProtectionUniversity of StrasbourgStrasbourgFrance
| | - Margherita Giannini
- FMTS, Department of Physiology, EA3072 Mitochondria, Oxidative Stress and Muscular ProtectionUniversity of StrasbourgStrasbourgFrance
- Department of Physiology and Functional ExplorationsUniversity Hospital of StrasbourgStrasbourgFrance
| | - Nabil Chakfé
- FMTS, Department of Physiology, EA3072 Mitochondria, Oxidative Stress and Muscular ProtectionUniversity of StrasbourgStrasbourgFrance
- Department of Vascular Surgery and Kidney TransplantationUniversity Hospital of StrasbourgStrasbourgFrance
| | - Anne Lejay
- FMTS, Department of Physiology, EA3072 Mitochondria, Oxidative Stress and Muscular ProtectionUniversity of StrasbourgStrasbourgFrance
- Department of Vascular Surgery and Kidney TransplantationUniversity Hospital of StrasbourgStrasbourgFrance
| | - Bernard Geny
- FMTS, Department of Physiology, EA3072 Mitochondria, Oxidative Stress and Muscular ProtectionUniversity of StrasbourgStrasbourgFrance
- Department of Physiology and Functional ExplorationsUniversity Hospital of StrasbourgStrasbourgFrance
| |
Collapse
|
35
|
Signorelli SS, Marino E, Scuto S, Di Raimondo D. Pathophysiology of Peripheral Arterial Disease (PAD): A Review on Oxidative Disorders. Int J Mol Sci 2020; 21:ijms21124393. [PMID: 32575692 PMCID: PMC7352779 DOI: 10.3390/ijms21124393] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/13/2020] [Accepted: 06/18/2020] [Indexed: 12/14/2022] Open
Abstract
Peripheral arterial disease (PAD) is an atherosclerotic disease that affects a wide range of the world’s population, reaching up to 200 million individuals worldwide. PAD particularly affects elderly individuals (>65 years old). PAD is often underdiagnosed or underestimated, although specificity in diagnosis is shown by an ankle/brachial approach, and the high cardiovascular event risk that affected the PAD patients. A number of pathophysiologic pathways operate in chronic arterial ischemia of lower limbs, giving the possibility to improve therapeutic strategies and the outcome of patients. This review aims to provide a well detailed description of such fundamental issues as physical exercise, biochemistry of physical exercise, skeletal muscle in PAD, heme oxygenase 1 (HO-1) in PAD, and antioxidants in PAD. These issues are closely related to the oxidative stress in PAD. We want to draw attention to the pathophysiologic pathways that are considered to be beneficial in order to achieve more effective options to treat PAD patients.
Collapse
Affiliation(s)
- Salvatore Santo Signorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95125 Catania, Italy; (E.M.); (S.S.)
- Correspondence: ; Tel.: +39-09-5378-2545
| | - Elisa Marino
- Department of Clinical and Experimental Medicine, University of Catania, 95125 Catania, Italy; (E.M.); (S.S.)
| | - Salvatore Scuto
- Department of Clinical and Experimental Medicine, University of Catania, 95125 Catania, Italy; (E.M.); (S.S.)
| | - Domenico Di Raimondo
- Division of Internal Medicine and Stroke Care, Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (Promise) G. D’Alessandro, University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
36
|
Groennebaek T, Nielsen J, Jespersen NR, Bøtker HE, de Paoli FV, Miller BF, Vissing K. Utilization of biomarkers as predictors of skeletal muscle mitochondrial content after physiological intervention and in clinical settings. Am J Physiol Endocrinol Metab 2020; 318:E886-E889. [PMID: 32255679 DOI: 10.1152/ajpendo.00101.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The measurement of mitochondrial content is essential for bioenergetic research, as it provides a tool to evaluate whether changes in mitochondrial function are strictly due to changes in content or other mechanisms that influence function. In this perspective, we argue that commonly used biomarkers of mitochondrial content may possess limited utility for capturing changes in content with physiological intervention. Moreover, we argue that they may not provide reliable estimates of content in certain pathological situations. Finally, we discuss potential solutions to overcome issues related to the utilization of biomarkers of mitochondrial content. Shedding light on this important issue will hopefully aid conclusions about the mitochondrial structure-function relationship.
Collapse
Affiliation(s)
| | - Joachim Nielsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | | | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | | |
Collapse
|
37
|
Groennebaek T, Billeskov TB, Schytz CT, Jespersen NR, Bøtker HE, Olsen RKJ, Eldrup N, Nielsen J, Farup J, de Paoli FV, Vissing K. Mitochondrial Structure and Function in the Metabolic Myopathy Accompanying Patients with Critical Limb Ischemia. Cells 2020; 9:cells9030570. [PMID: 32121096 PMCID: PMC7140415 DOI: 10.3390/cells9030570] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction has been implicated as a central mechanism in the metabolic myopathy accompanying critical limb ischemia (CLI). However, whether mitochondrial dysfunction is directly related to lower extremity ischemia and the structural and molecular mechanisms underpinning mitochondrial dysfunction in CLI patients is not understood. Here, we aimed to study whether mitochondrial dysfunction is a distinctive characteristic of CLI myopathy by assessing mitochondrial respiration in gastrocnemius muscle from 14 CLI patients (65.3 ± 7.8 y) and 15 matched control patients (CON) with a similar comorbidity risk profile and medication regimen but without peripheral ischemia (67.4 ± 7.4 y). Furthermore, we studied potential structural and molecular mechanisms of mitochondrial dysfunction by measuring total, sub-population, and fiber-type-specific mitochondrial volumetric content and cristae density with transmission electron microscopy and by assessing mitophagy and fission/fusion-related protein expression. Finally, we asked whether commonly used biomarkers of mitochondrial content are valid in patients with cardiovascular disease. CLI patients exhibited inferior mitochondrial respiration compared to CON. This respiratory deficit was not related to lower whole-muscle mitochondrial content or cristae density. However, stratification for fiber types revealed ultrastructural mitochondrial alterations in CLI patients compared to CON. CLI patients exhibited an altered expression of mitophagy-related proteins but not fission/fusion-related proteins compared to CON. Citrate synthase, cytochrome c oxidase subunit IV (COXIV), and 3-hydroxyacyl-CoA dehydrogenase (β-HAD) could not predict mitochondrial content. Mitochondrial dysfunction is a distinctive characteristic of CLI myopathy and is not related to altered organelle content or cristae density. Our results link this intrinsic mitochondrial deficit to dysregulation of the mitochondrial quality control system, which has implications for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Thomas Groennebaek
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (C.T.S.)
| | - Tine Borum Billeskov
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.B.B.); (J.F.)
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Camilla Tvede Schytz
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (C.T.S.)
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, 5230 Odense, Denmark;
| | - Nichlas Riise Jespersen
- Department of Cardiology, Aarhus University Hospital, 8200 Aarhus, Denmark; (N.R.J.); (H.E.B.)
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, 8200 Aarhus, Denmark; (N.R.J.); (H.E.B.)
| | | | - Nikolaj Eldrup
- Department Vascular Surgery, Rigshospitalet, Copenhagen University, 2100 Copenhagen, Denmark;
| | - Joachim Nielsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, 5230 Odense, Denmark;
| | - Jean Farup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.B.B.); (J.F.)
| | - Frank Vincenzo de Paoli
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.B.B.); (J.F.)
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, 8200 Aarhus, Denmark
- Correspondence: (F.V.d.P.); (K.V.); Tel.: +45-87168173
| | - Kristian Vissing
- Department of Public Health, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (C.T.S.)
- Correspondence: (F.V.d.P.); (K.V.); Tel.: +45-87168173
| |
Collapse
|
38
|
Matsui R, Hamburg NM. Eating Chocolate to Improve Muscle Health and Walking Ability in Patients With Peripheral Artery Disease. Circ Res 2020; 126:600-602. [DOI: 10.1161/circresaha.120.316614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Reiko Matsui
- From the Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA
| | - Naomi M. Hamburg
- From the Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, MA
| |
Collapse
|
39
|
Drp1 overexpression induces desmin disassembling and drives kinesin-1 activation promoting mitochondrial trafficking in skeletal muscle. Cell Death Differ 2020; 27:2383-2401. [PMID: 32042098 PMCID: PMC7370230 DOI: 10.1038/s41418-020-0510-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/13/2019] [Accepted: 01/23/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondria change distribution across cells following a variety of pathophysiological stimuli. The mechanisms presiding over this redistribution are yet undefined. In a murine model overexpressing Drp1 specifically in skeletal muscle, we find marked mitochondria repositioning in muscle fibres and we demonstrate that Drp1 is involved in this process. Drp1 binds KLC1 and enhances microtubule-dependent transport of mitochondria. Drp1-KLC1 coupling triggers the displacement of KIF5B from kinesin-1 complex increasing its binding to microtubule tracks and mitochondrial transport. High levels of Drp1 exacerbate this mechanism leading to the repositioning of mitochondria closer to nuclei. The reduction of Drp1 levels decreases kinesin-1 activation and induces the partial recovery of mitochondrial distribution. Drp1 overexpression is also associated with higher cyclin-dependent kinase-1 (Cdk-1) activation that promotes the persistent phosphorylation of desmin at Ser-31 and its disassembling. Fission inhibition has a positive effect on desmin Ser-31 phosphorylation, regardless of Cdk-1 activation, suggesting that induction of both fission and Cdk-1 are required for desmin collapse. This altered desmin architecture impairs mechanotransduction and compromises mitochondrial network stability priming mitochondria transport through microtubule-dependent trafficking with a mechanism that involves the Drp1-dependent regulation of kinesin-1 complex.
Collapse
|
40
|
Pizzimenti M, Riou M, Charles AL, Talha S, Meyer A, Andres E, Chakfé N, Lejay A, Geny B. The Rise of Mitochondria in Peripheral Arterial Disease Physiopathology: Experimental and Clinical Data. J Clin Med 2019; 8:jcm8122125. [PMID: 31810355 PMCID: PMC6947197 DOI: 10.3390/jcm8122125] [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: 11/10/2019] [Revised: 11/27/2019] [Accepted: 11/29/2019] [Indexed: 12/21/2022] Open
Abstract
Peripheral arterial disease (PAD) is a frequent and serious condition, potentially life-threatening and leading to lower-limb amputation. Its pathophysiology is generally related to ischemia-reperfusion cycles, secondary to reduction or interruption of the arterial blood flow followed by reperfusion episodes that are necessary but also—per se—deleterious. Skeletal muscles alterations significantly participate in PAD injuries, and interestingly, muscle mitochondrial dysfunctions have been demonstrated to be key events and to have a prognosis value. Decreased oxidative capacity due to mitochondrial respiratory chain impairment is associated with increased release of reactive oxygen species and reduction of calcium retention capacity leading thus to enhanced apoptosis. Therefore, targeting mitochondria might be a promising therapeutic approach in PAD.
Collapse
Affiliation(s)
- Mégane Pizzimenti
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 «Mitochondria, Oxidative Stress and Muscle Protection», 11 Rue Humann, 67000 Strasbourg, France; (M.P.); (M.R.); (A.-L.C.); (S.T.); (A.M.); (A.L.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Marianne Riou
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 «Mitochondria, Oxidative Stress and Muscle Protection», 11 Rue Humann, 67000 Strasbourg, France; (M.P.); (M.R.); (A.-L.C.); (S.T.); (A.M.); (A.L.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Anne-Laure Charles
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 «Mitochondria, Oxidative Stress and Muscle Protection», 11 Rue Humann, 67000 Strasbourg, France; (M.P.); (M.R.); (A.-L.C.); (S.T.); (A.M.); (A.L.)
| | - Samy Talha
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 «Mitochondria, Oxidative Stress and Muscle Protection», 11 Rue Humann, 67000 Strasbourg, France; (M.P.); (M.R.); (A.-L.C.); (S.T.); (A.M.); (A.L.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Alain Meyer
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 «Mitochondria, Oxidative Stress and Muscle Protection», 11 Rue Humann, 67000 Strasbourg, France; (M.P.); (M.R.); (A.-L.C.); (S.T.); (A.M.); (A.L.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Emmanuel Andres
- Internal Medicine, Diabete and Metabolic Diseases Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France;
| | - Nabil Chakfé
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 «Mitochondria, Oxidative Stress and Muscle Protection», 11 Rue Humann, 67000 Strasbourg, France; (M.P.); (M.R.); (A.-L.C.); (S.T.); (A.M.); (A.L.)
- Vascular Surgery and Kidney Transplantation Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Anne Lejay
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 «Mitochondria, Oxidative Stress and Muscle Protection», 11 Rue Humann, 67000 Strasbourg, France; (M.P.); (M.R.); (A.-L.C.); (S.T.); (A.M.); (A.L.)
- Vascular Surgery and Kidney Transplantation Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
| | - Bernard Geny
- Unistra, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072 «Mitochondria, Oxidative Stress and Muscle Protection», 11 Rue Humann, 67000 Strasbourg, France; (M.P.); (M.R.); (A.-L.C.); (S.T.); (A.M.); (A.L.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’Hôpital, 67091 Strasbourg CEDEX, France
- Correspondence:
| |
Collapse
|
41
|
Caradu C, Couffinhal T, Chapouly C, Guimbal S, Hollier PL, Ducasse E, Bura-Rivière A, Dubois M, Gadeau AP, Renault MA. Restoring Endothelial Function by Targeting Desert Hedgehog Downstream of Klf2 Improves Critical Limb Ischemia in Adults. Circ Res 2019; 123:1053-1065. [PMID: 30355159 DOI: 10.1161/circresaha.118.313177] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Klf (kruppel-like factor) 2 is critical to establish and maintain endothelial integrity. OBJECTIVE Therefore, determining upstream and downstream mediators of Klf2 would lead to alternative therapeutic targets in cardiovascular disease management. METHODS AND RESULTS Here we identify Dhh (desert hedgehog) as a downstream effector of Klf2, whose expression in endothelial cells (ECs) is upregulated by shear stress and decreased by inflammatory cytokines. Consequently, we show that Dhh knockdown in ECs promotes endothelial permeability and EC activation and that Dhh agonist prevents TNF-α (tumor necrosis factor alpha) or glucose-induced EC dysfunction. Moreover, we demonstrate that human critical limb ischemia, a pathological condition linked to diabetes mellitus and inflammation, is associated to major EC dysfunction. By recreating a complex model of critical limb ischemia in diabetic mice, we found that Dhh-signaling agonist significantly improved EC function without promoting angiogenesis, which subsequently improved muscle perfusion. CONCLUSION Restoring EC function leads to significant critical limb ischemia recovery. Dhh appears to be a promising target, downstream of Klf2, to prevent the endothelial dysfunction involved in ischemic vascular diseases.
Collapse
Affiliation(s)
- Caroline Caradu
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Thierry Couffinhal
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Candice Chapouly
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Sarah Guimbal
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Pierre-Louis Hollier
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Eric Ducasse
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | | | - Mathilde Dubois
- Inserm U1045, Centre de recherche Cardio-thoracique, University of Bordeaux, France (M.D.)
| | - Alain-Pierre Gadeau
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Marie-Ange Renault
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| |
Collapse
|
42
|
Fuglestad MA, Hernandez H, Gao Y, Ybay H, Schieber MN, Brunette KE, Myers SA, Casale GP, Pipinos II. A low-cost, wireless near-infrared spectroscopy device detects the presence of lower extremity atherosclerosis as measured by computed tomographic angiography and characterizes walking impairment in peripheral artery disease. J Vasc Surg 2019; 71:946-957. [PMID: 31445826 DOI: 10.1016/j.jvs.2019.04.493] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 04/28/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Patients with peripheral artery disease (PAD) who experience intermittent claudication report a range of symptoms. Patients with symptoms other than classically described intermittent claudication may be at the highest risk for functional decline and mobility loss. Therefore, technologies allowing for characterization of PAD severity are desirable. Near-infrared spectroscopy (NIRS) allows for measurements of muscle heme oxygen saturation (StO2) during exercise. We hypothesized lower extremities affected by PAD would exhibit distinct NIRS profiles as measured by a low-cost, wireless NIRS device and that NIRS during exercise predicts walking limitation. METHODS We recruited 40 patients with PAD and 10 control participants. All patients with PAD completed a computed tomographic angiography, 6-minute walk test, and a standardized treadmill test. Controls completed a 540-second treadmill test for comparison. StO2 measurements were continuously taken from the gastrocnemius during exercise. Variables were analyzed by Fischer's exact, χ2, Wilcoxon rank-sum, and Kruskal-Wallis tests as appropriate. Correlations were assessed by partial Spearman correlation coefficients adjusted for occlusive disease pattern. RESULTS Patients with PAD experienced claudication onset at a median of 108 seconds with a median peak walking time of 288 seconds. The baseline StO2 was similar between PAD and control. The StO2 of PAD and control participants dropped below baseline at a median of 1 and 104 seconds of exercise, respectively (P < .0001). Patients with PAD reached minimum StO2 earlier than control participants (119 seconds vs 522 seconds, respectively; P < .001) and experienced a greater change in StO2 at 1 minute of exercise (-73.2% vs 8.3%; P < .0001) and a greater decrease at minimum exercise StO2 (-83.4% vs -16.1%; P < .0001). For patients with PAD, peak walking time, and 6-minute walking distance correlated with percent change in StO2 at 1 minute of exercise (r = -0.76 and -0.67, respectively; P < .001) and time to minimum StO2 (r = 0.79 and 0.70, respectively; P < .0001). CONCLUSIONS In this initial evaluation of a novel, low-cost NIRS device, lower extremities affected by PAD exhibited characteristic changes in calf muscle StO2, which differentiated them from healthy controls and were strongly correlated with walking impairment. These findings confirm and expand on previous work demonstrating the potential clinical value of NIRS devices and the need for further research investigating the ability of low-cost NIRS technology to evaluate, diagnose, and monitor treatment response in PAD.
Collapse
Affiliation(s)
| | - Hernan Hernandez
- Department of Surgery, University of Nebraska Medical Center, Omaha, Neb
| | - Yue Gao
- Department of Surgery, University of Nebraska Medical Center, Omaha, Neb
| | - Henamari Ybay
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, Neb
| | - Molly N Schieber
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, Neb
| | | | - Sara A Myers
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, Neb
| | - George P Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, Neb
| | - Iraklis I Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, Neb; Department of Biomechanics, University of Nebraska at Omaha, Omaha, Neb; Department of Surgery, Veterans Affairs Medical Center, Omaha, Neb.
| |
Collapse
|
43
|
Kim K, Reid BA, Ro B, Casey CA, Song Q, Kuang S, Roseguini BT. Heat therapy improves soleus muscle force in a model of ischemia-induced muscle damage. J Appl Physiol (1985) 2019; 127:215-228. [PMID: 31161885 DOI: 10.1152/japplphysiol.00115.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Leg muscle ischemia in patients with peripheral artery disease (PAD) leads to alterations in skeletal muscle morphology and reduced leg strength. We tested the hypothesis that exposure to heat therapy (HT) would improve skeletal muscle function in a mouse model of ischemia-induced muscle damage. Male 42-wk-old C57Bl/6 mice underwent ligation of the femoral artery and were randomly assigned to receive HT (immersion in a water bath at 37°C, 39°C, or 41°C for 30 min) or a control intervention for 3 wk. At the end of the treatment, the animals were anesthetized and the soleus and extensor digitorum longus (EDL) muscles were harvested for the assessment of contractile function and examination of muscle morphology. A subset of animals was used to examine the impact of a single HT session on the expression of genes involved in myogenesis and the regulation of muscle mass. Relative soleus muscle mass was significantly higher in animals exposed to HT at 39°C compared with the control group (control: 0.36 ± 0.01 mg/g versus 39°C: 0.40 ± 0.01 mg/g, P = 0.024). Maximal absolute force of the soleus was also significantly higher in animals treated with HT at 37°C and 39°C (control: 274.7 ± 6.6 mN; 37°C: 300.1 ± 7.7 mN; 39°C: 299.5 ± 10 mN, P < 0.05). In the soleus, but not the EDL muscle, a single session of HT enhanced the mRNA expression of myogenic factors as well as of both positive and negative regulators of muscle mass. These findings suggest that the beneficial effects of HT are muscle specific and dependent on the treatment temperature in a model of PAD. NEW & NOTEWORTHY This is the first study to comprehensively examine the impact of temperature and muscle fiber type composition on the adaptations to repeated heat stress in a model of ischemia-induced muscle damage. Exposure to heat therapy (HT) at 37°C and 39°C, but not at 41°C, improved force development of the isolated soleus muscle. These results suggest that HT may be a practical therapeutic tool to restore muscle mass and strength in patients with peripheral artery disease.
Collapse
Affiliation(s)
- Kyoungrae Kim
- Department of Health and Kinesiology, Purdue University , West Lafayette, Indiana
| | - Blake A Reid
- Department of Health and Kinesiology, Purdue University , West Lafayette, Indiana
| | - Bohyun Ro
- Department of Physical Education, Dong-A University , Busan , Korea
| | - Caitlin A Casey
- Department of Health and Kinesiology, Purdue University , West Lafayette, Indiana
| | - Qifan Song
- Department of Statistics, Purdue University , West Lafayette, Indiana
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University , West Lafayette, Indiana
| | - Bruno T Roseguini
- Department of Health and Kinesiology, Purdue University , West Lafayette, Indiana
| |
Collapse
|
44
|
Shah F, Franklin KA, Holmlund T, Levring Jäghagen E, Berggren D, Forsgren S, Stål P. Desmin and dystrophin abnormalities in upper airway muscles of snorers and patients with sleep apnea. Respir Res 2019; 20:31. [PMID: 30764835 PMCID: PMC6376723 DOI: 10.1186/s12931-019-0999-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/04/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The pathophysiology of obstruction and swallowing dysfunction in snores and sleep apnea patients remains unclear. Neuropathy and to some extent myopathy have been suggested as contributing causes. Recently we reported an absence and an abnormal isoform of two cytoskeletal proteins, desmin, and dystrophin, in upper airway muscles of healthy humans. These cytoskeletal proteins are considered vital for muscle function. We aimed to investigate for muscle cytoskeletal abnormalities in upper airways and its association with swallowing dysfunction and severity of sleep apnea. METHODS Cytoskeletal proteins desmin and dystrophin were morphologically evaluated in the uvula muscle of 22 patients undergoing soft palate surgery due to snoring and sleep apnea and in 10 healthy controls. The muscles were analysed with immunohistochemical methods, and swallowing function was assessed using videoradiography. RESULTS Desmin displayed a disorganized pattern in 21 ± 13% of the muscle fibres in patients, while these fibers were not present in controls. Muscle fibres lacking desmin were present in both patients and controls, but the proportion was higher in patients (25 ± 12% vs. 14 ± 7%, p = 0.009). The overall desmin abnormalities were significantly more frequent in patients than in controls (46 ± 18% vs. 14 ± 7%, p < 0.001). In patients, the C-terminus of the dystrophin molecule was absent in 19 ± 18% of the desmin-abnormal muscle fibres. Patients with swallowing dysfunction had 55 ± 10% desmin-abnormal muscle fibres vs. 22 ± 6% in patients without swallowing dysfunction, p = 0.002. CONCLUSION Cytoskeletal abnormalities in soft palate muscles most likely contribute to pharyngeal dysfunction in snorers and sleep apnea patients. Plausible causes for the presence of these abnormalities is traumatic snoring vibrations, tissue stretch or muscle overload.
Collapse
Affiliation(s)
- Farhan Shah
- Department of Integrative Medical Biology, Laboratory of Muscle Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Karl A. Franklin
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Thorbjörn Holmlund
- Department of Clinical Sciences, Otolaryngology, Umeå University, Umeå, Sweden
| | - Eva Levring Jäghagen
- Department of Odontology, Oral and Maxillofacial Radiology, Umeå University, Umeå, Sweden
| | - Diana Berggren
- Department of Clinical Sciences, Otolaryngology, Umeå University, Umeå, Sweden
| | - Sture Forsgren
- Department of Integrative Medical Biology, Laboratory of Muscle Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Per Stål
- Department of Integrative Medical Biology, Laboratory of Muscle Biology, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
45
|
Sfyri PP, Yuldasheva NY, Tzimou A, Giallourou N, Crispi V, Aburima A, Beltran-Alvarez P, Patel K, Mougios V, Swann JR, Kearney MT, Matsakas A. Attenuation of oxidative stress-induced lesions in skeletal muscle in a mouse model of obesity-independent hyperlipidaemia and atherosclerosis through the inhibition of Nox2 activity. Free Radic Biol Med 2018; 129:504-519. [PMID: 30342191 DOI: 10.1016/j.freeradbiomed.2018.10.422] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 09/12/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023]
Abstract
Obesity leading to hyperlipidaemia and atherosclerosis is recognised to induce morphological and metabolic changes in many tissues. However, hyperlipidaemia can occur in the absence of obesity. The impact of the latter scenario on skeletal muscle and liver is not understood sufficiently. In this regard, we used the Apolipoprotein E-deficient (ApoE-/-) mouse model, an established model of hyperlipidaemia and atherosclerosis, that does not become obese when subjected to a high-fat diet, to determine the impact of Western-type diet (WD) and ApoE deficiency on skeletal muscle morphological, metabolic and biochemical properties. To establish the potential of therapeutic targets, we further examined the impact of Nox2 pharmacological inhibition on skeletal muscle redox biology. We found ectopic lipid accumulation in skeletal muscle and the liver, and altered skeletal muscle morphology and intramuscular triacylglycerol fatty acid composition. WD and ApoE deficiency had a detrimental impact in muscle metabolome, followed by perturbed gene expression for fatty acid uptake and oxidation. Importantly, there was enhanced oxidative stress in the skeletal muscle and development of liver steatosis, inflammation and oxidative protein modifications. Pharmacological inhibition of Nox2 decreased reactive oxygen species production and protein oxidative modifications in the muscle of ApoE-/- mice subjected to a Western-type diet. This study provides key evidence to better understand the pathophysiology of skeletal muscle in the context of hyperlipidaemia and atherosclerosis and identifies Nox2 as a potential target for attenuating oxidative stress in skeletal muscle in a mouse model of obesity-independent hyperlipidaemia.
Collapse
Affiliation(s)
- Pagona Panagiota Sfyri
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, United Kingdom
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom
| | - Anastasia Tzimou
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sports Science at Thessaloniki, Aristotle University of Thessaloniki, Greece
| | - Natasa Giallourou
- Department of Surgery and Cancer, Division of Computational and Systems Medicine, Imperial College London, United Kingdom
| | - Vassili Crispi
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, United Kingdom
| | - Ahmed Aburima
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, United Kingdom
| | | | - Ketan Patel
- School of Biological Sciences, University of Reading, United Kingdom
| | - Vassilis Mougios
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sports Science at Thessaloniki, Aristotle University of Thessaloniki, Greece
| | - Jonathan R Swann
- Department of Surgery and Cancer, Division of Computational and Systems Medicine, Imperial College London, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, United Kingdom.
| |
Collapse
|
46
|
Shah F, Forsgren S, Holmlund T, Levring Jäghagen E, Berggren D, Franklin KA, Stål P. Neurotrophic factor BDNF is upregulated in soft palate muscles of snorers and sleep apnea patients. Laryngoscope Investig Otolaryngol 2018; 4:174-180. [PMID: 30828636 PMCID: PMC6383323 DOI: 10.1002/lio2.225] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/13/2018] [Accepted: 09/29/2018] [Indexed: 11/25/2022] Open
Abstract
Objectives Neuromuscular injuries are suggested to contribute to upper airway collapse and swallowing dysfunction in patients with sleep apnea. Neurotrophins, a family of proteins involved in survival, development, and function of neurons, are reported to be upregulated in limb muscle fibers in response to overload and nerve damage. We aimed to investigate the expression of two important neurotrophins, brain‐derived neurotrophic factor (BDNF) and nerve growth factor (NGF), in muscle fibers of uvula from snorers and sleep apnea patients and to compare these findings with pharyngeal function. Methods Uvula muscle biopsies from 22 patients and 10 controls were analyzed for BDNF, NGF, and cytoskeletal protein desmin using immunohistochemistry. Pharyngeal swallowing function was assessed using videoradiography. Results BDNF, but not NGF, was significantly upregulated in a subpopulation of muscle fibers in snoring and sleep apnea patients. Two major immunoreaction patterns for BDNF were observed; a fine grainy point like BDNF staining was displayed in muscle fibers of both patients and controls (41 ± 23 vs. 25 ± 17%, respectively, P = .06), while an abnormal upregulated intense‐dotted or disorganized reaction was mainly observed in patients (8 ± 8 vs. 2 ± 2%, P = .02). The latter fibers, which often displayed an abnormal immunoreaction for desmin, were more frequent in patients with than without swallowing dysfunction (10 ± 8 vs. 3 ± 3%, P = .05). Conclusion BDNF is upregulated in the upper airway muscles of snorers and sleep apnea patients, and especially in patients with swallowing dysfunction. Upregulation of BDNF is suggested to be a response to denervation, reinnervation, and repair of injured muscle fibers. Our findings propose that damaged upper airway muscles might heal following treatment for snoring and sleep apnea. Level of Evidence NA
Collapse
Affiliation(s)
- Farhan Shah
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
| | - Sture Forsgren
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
| | - Thorbjörn Holmlund
- Laboratory of Muscle Biology, the Department of Clinical Sciences, OtolaryngologyUmeå UniversityUmeåSweden
| | - Eva Levring Jäghagen
- Department of Odontology, Oral, and Maxillofacial RadiologyUmeå UniversityUmeåSweden
| | - Diana Berggren
- Laboratory of Muscle Biology, the Department of Clinical Sciences, OtolaryngologyUmeå UniversityUmeåSweden
| | - Karl A. Franklin
- Department of Surgical and Perioperative SciencesUmeå UniversityUmeåSweden
| | - Per Stål
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
| |
Collapse
|
47
|
Begam M, Roche JA. Damaged muscle fibers might masquerade as hybrid fibers - a cautionary note on immunophenotyping mouse muscle with mouse monoclonal antibodies. Eur J Histochem 2018; 62. [PMID: 30043594 PMCID: PMC6060487 DOI: 10.4081/ejh.2018.2896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/06/2018] [Indexed: 11/24/2022] Open
Abstract
We report that, labeling mouse muscle tissue, with mouse monoclonal antibodies specific to slow or fast myosin heavy chain (sMyHC and fMyHC, respectively), can lead to artefactual labeling of damaged muscle fibers, as hybrid fibers (sMyHC+ and fMyHC+). We demonstrate that, such erroneous immunophenotyping of muscle may be avoided, by performing colabeling or serialsection- labeling, to identify damaged fibers. The quadriceps femorismuscle group (QF) in 7-month-old, male, C57BL/6J mice had: 1.21±0.21%, 98.34±1.06%, 0.07±0.01%, and 0.53±0.85% fibers, that were, sMyHC+, fMyHC+, hybrid, and damaged, respectively. All fibers in the tibialis anterior muscle (TA) of 3-month-old, male, C57BL/6J mice were fMyHC+; and at 3 days after injurious eccentric contractions, there was no fiber-type shift, but ~ 18% fibers were damaged.
Collapse
Affiliation(s)
- Morium Begam
- Wayne State University, Department of Health Care Sciences.
| | | |
Collapse
|
48
|
Signolet I, Abraham P, Chupin S, Ammi M, Gueguen N, Letournel F, Picquet J, Baufreton C, Daligault M, Procaccio V, Reynier P, Henni S. Mitochondrial complex I defect resulting from exercise-induced lower limb ischemia in patients with peripheral arterial disease. J Appl Physiol (1985) 2018; 125:938-946. [PMID: 29792553 DOI: 10.1152/japplphysiol.00059.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study aims to compare the structural and mitochondrial alterations between muscle segments affected by exercise-induced ischemia and segments of the same muscle without ischemia, in the same subject. In a prospective analysis, 34 patients presenting either peripheral arterial disease or chronic coronary syndrome without any evidence of peripheral arterial disease were eligible for inclusion based on findings indicating a need for either a femoro-popliteal bypass or a saphenous harvesting for coronary bypass. Before surgery, we assessed the level of exercise-induced ischemia in proximal and distal sections of the thigh by the measurement of transcutaneous oxygen pressure during an exercise treadmill test. Distal and proximal biopsies of the sartorius muscle were procured during vascular surgical procedures to assess mitochondrial function and morphometric parameters of the sartorius myofibers. Comparisons were made between the distal and proximal biopsies, with respect to these parameters. Thirteen of the study patients that initially presented with peripheral arterial disease had evidence of an isolated distal thigh exercise-induced ischemia, associated with a 35% decrease in the mitochondrial complex I enzymatic activity in the distal muscle biopsy. This defect was also associated with a decreased expression of the manganese superoxide dismutase enzyme and with alterations of the shapes of the myofibers. No functional or structural alterations were observed in the patients with coronary syndrome. We validated a specific model ischemia in peripheral arterial disease characterized by muscular alterations. This "Distal-Proximal-Sartorius Model" would be promising to explore the physiopathological consequences specific to chronic ischemia. NEW & NOTEWORTHY We compared proximal versus distal biopsies of the sartorius muscle in patients with superficial femoral artery stenosis or occlusion and proof of, distal only, regional blood flow impairment with exercise oximetry. We identified a decrease in the mitochondrial complex I enzymatic activity and antioxidant system impairment at the distal level only. We validate a model to explore the physiopathological consequences of chronic muscle ischemia.
Collapse
Affiliation(s)
- I Signolet
- Laboratory for Vascular Investigation, University Hospital , Angers , France.,Department of Biochemistry and Genetics, University Hospital , Angers , France
| | - P Abraham
- Laboratory for Vascular Investigation, University Hospital , Angers , France.,Mitovasc Institute, CNRS 6015, INSERM U1083, University of Angers , Angers , France
| | - S Chupin
- Department of Biochemistry and Genetics, University Hospital , Angers , France.,Mitovasc Institute, CNRS 6015, INSERM U1083, University of Angers , Angers , France
| | - M Ammi
- Department of Vascular Surgery, University Hospital , Angers , France
| | - N Gueguen
- Department of Biochemistry and Genetics, University Hospital , Angers , France.,Mitovasc Institute, CNRS 6015, INSERM U1083, University of Angers , Angers , France
| | - F Letournel
- Department of Tissue and Cellular Pathology, University Hospital , Angers , France
| | - J Picquet
- Department of Vascular Surgery, University Hospital , Angers , France
| | - C Baufreton
- Department of Cardiac Surgery, University Hospital , Angers , France
| | - M Daligault
- Department of Vascular Surgery, University Hospital , Angers , France
| | - V Procaccio
- Department of Biochemistry and Genetics, University Hospital , Angers , France.,Mitovasc Institute, CNRS 6015, INSERM U1083, University of Angers , Angers , France
| | - P Reynier
- Department of Biochemistry and Genetics, University Hospital , Angers , France.,Mitovasc Institute, CNRS 6015, INSERM U1083, University of Angers , Angers , France
| | - S Henni
- Laboratory for Vascular Investigation, University Hospital , Angers , France.,Mitovasc Institute, CNRS 6015, INSERM U1083, University of Angers , Angers , France
| |
Collapse
|
49
|
Kawasaki T, Iwasaki T, Yamada M, Yoshida T, Watanabe T. Rapid growth rate results in remarkably hardened breast in broilers during the middle stage of rearing: A biochemical and histopathological study. PLoS One 2018; 13:e0193307. [PMID: 29474396 PMCID: PMC5825104 DOI: 10.1371/journal.pone.0193307] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 02/08/2018] [Indexed: 11/29/2022] Open
Abstract
The high incidence of meat of impaired quality poses a serious problem in the poultry industry. In recent years, the incidence of the pectoralis major muscle that appeared pale colored, remarkably hardened, and exudative, called "wooden breast" or "woody breast" has increased in slaughter houses. In the present study, 19-day-old Ross 308 broiler chickens affected (n = 10) and unaffected (n = 10) with remarkably hardened breast were selected from a commercial broiler farm, and reared to 55 days of age under a controlled environment. Among the affected birds, 5 of 10 birds appeared exhausted with markedly suppressed weight gain and 4 of 10 birds died during the rearing period. In contrast, all unaffected birds survived and most gained weight. Four of 10 unaffected birds lost the ability of back-to-back wing contact by the late stage of rearing. The biochemical analysis of blood plasma samples of 20-day-old birds revealed that creatine kinase and L-aspartate aminotransferase values in most affected birds were higher than those in unaffected birds; however, these values in unaffected birds increased rapidly with lost wing contactability and increasing age. Postmortem examinations revealed that the mean diameter of myofibers in affected birds was smaller than that in unaffected birds. Moreover, symptoms of degenerative and regenerative muscles were observed in most birds in both groups. Among them, a decrease in, or defect of, the characteristic polygonal shape of myofibers was the most common change within the pectoralis major muscles in both groups. The present study demonstrated that broilers affected with remarkably hardened breast during the middle stage of rearing would have suppressed physical status and weight gain, or would die. It was suggested that rapid growth in broilers might be a cause of remarkably hardened breast.
Collapse
Affiliation(s)
- Takeshi Kawasaki
- Department of Food Science and Human Wellness, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
- Research Office Concerning the Health of Humans and Birds, Abashiri, Hokkaido, Japan
- Medical Engineering Course, Graduate School of Engineering, Kitami Institute of Technology, Kitami, Hokkaido, Japan
| | - Tomohito Iwasaki
- Department of Food Science and Human Wellness, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Michi Yamada
- Department of Sustainable Agriculture, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Takashi Yoshida
- Medical Engineering Course, Graduate School of Engineering, Kitami Institute of Technology, Kitami, Hokkaido, Japan
| | | |
Collapse
|
50
|
Frangogiannis NG. Cell therapy for peripheral artery disease. Curr Opin Pharmacol 2018; 39:27-34. [PMID: 29452987 DOI: 10.1016/j.coph.2018.01.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/28/2022]
Abstract
Patients with severe peripheral artery disease (PAD) who are not candidates for revascularization have poor prognosis. Cell therapy using peripheral blood-derived or bone marrow-derived mononuclear cells, mesenchymal stem cells, or marker-specific subsets of bone marrow cells with angiogenic properties may hold promise for no-option PAD patients. Injected cells may exert beneficial actions by enhancing local angiogenesis (either through maturation of endothelial progenitors, or through secretion of angiogenic mediators), or by transducing cytoprotective signals that preserve tissue structure. Despite extensive research, robust clinical evidence supporting the use of cell therapy in patients with critical limb ischemia is lacking. Larger, well-designed placebo-controlled clinical trials did not support the positive results of smaller less rigorous studies. There is a need for high-quality clinical studies to test the effectiveness of cell therapy in PAD patients. Moreover, fundamental cell biological studies are needed to identify the optimal cell types, and to develop strategies that may enhance homing, survival and effectiveness of the injected cells.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|