1
|
Davuluri S, Lood C, Chung L. Calcinosis in systemic sclerosis. Curr Opin Rheumatol 2024; 36:360-369. [PMID: 37830924 DOI: 10.1097/bor.0000000000000900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
PURPOSE OF REVIEW To provide updated information on the prevalence, pathogenesis, diagnostics, and therapeutics of calcinosis cutis associated with systemic sclerosis (SSc). RECENT FINDINGS Observational studies show ethnic and geographical differences in the prevalence of calcinosis. In addition to clinical and serological associations, biochemical studies and in-vivo models have attempted to explain theories behind its pathogenesis, including prolonged state of inflammation, mechanical stress, hypoxia, and dysregulation in bone and phosphate metabolism. Long-term use of proton pump inhibitors may increase the risk for calcinosis in SSc. Few single center observational studies have shown mild benefit with minocycline and topical sodium thiosulfate. SUMMARY Calcinosis cutis is the deposition of insoluble calcium in the skin and subcutaneous tissues. It affects up to 40% of SSc patients and causes significant morbidity. Long disease duration, features of vascular dysfunction, and osteoporosis have been associated with calcinosis. Altered levels of inorganic pyrophosphate and fibroblast growth factor-23 have been implicated in dysregulated phosphate metabolism that may lead to calcinosis in SSc. Plain radiography can help with diagnosis and quantifying the calcinosis burden. Surgical treatment remains the most effective therapy when feasible. At present, no medical therapies have proven efficacy in large randomized controlled trials.
Collapse
Affiliation(s)
- Srijana Davuluri
- Stanford School of Medicine, Division of Immunology &Rheumatology, Palo Alto, California
| | - Christian Lood
- University of Washington, Division of Rheumatology, Seattle, Washington
| | - Lorinda Chung
- Stanford School of Medicine & Palo Alto VA Healthcare System, Division of Immunology & Rheumatology, Palo Alto, California, USA
| |
Collapse
|
2
|
Duvvuri B, Pachman LM, Hermanson P, Wang T, Moore R, Ding-Hwa Wang D, Long A, Morgan GA, Doty S, Tian R, Sancak Y, Lood C. Role of mitochondria in the myopathy of juvenile dermatomyositis and implications for skeletal muscle calcinosis. J Autoimmun 2023; 138:103061. [PMID: 37244073 PMCID: PMC10330803 DOI: 10.1016/j.jaut.2023.103061] [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] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/21/2023] [Accepted: 05/08/2023] [Indexed: 05/29/2023]
Abstract
OBJECTIVES To elucidate mechanisms contributing to skeletal muscle calcinosis in patients with juvenile dermatomyositis. METHODS A well-characterized cohorts of JDM (n = 68), disease controls (polymyositis, n = 7; juvenile SLE, n = 10, and RNP + overlap syndrome, n = 12), and age-matched health controls (n = 17) were analyzed for circulating levels of mitochondrial (mt) markers including mtDNA, mt-nd6, and anti-mitochondrial antibodies (AMAs) using standard qPCR, ELISA, and novel-in-house assays, respectively. Mitochondrial calcification of affected tissue biopsies was confirmed using electron microscopy and energy dispersive X-ray analysis. A human skeletal muscle cell line, RH30, was used to generate an in vitro calcification model. Intracellular calcification is measured by flow cytometry and microscopy. Mitochondria were assessed for mtROS production and membrane potential by flow cytometry and real-time oxygen consumption rate by Seahorse bioanalyzer. Inflammation (interferon-stimulated genes) was measured by qPCR. RESULTS In the current study, patients with JDM exhibited elevated levels of mitochondrial markers associated with muscle damage and calcinosis. Of particular interest are AMAs predictive of calcinosis. Human skeletal muscle cells undergo time- and dose-dependent accumulation of calcium phosphate salts with preferential localization to mitochondria. Calcification renders skeletal muscle cells mitochondria stressed, dysfunctional, destabilized, and interferogenic. Further, we report that inflammation induced by interferon-alpha amplifies mitochondrial calcification of human skeletal muscle cells via the generation of mitochondrial reactive oxygen species (mtROS). CONCLUSIONS Overall, our study demonstrates the mitochondrial involvement in the skeletal muscle pathology and calcinosis of JDM and mtROS as a central player in the calcification of human skeletal muscle cells. Therapeutic targeting of mtROS and/or upstream inducers, such as inflammation, may alleviate mitochondrial dysfunction, leading to calcinosis. AMAs can potentially identify patients with JDM at risk for developing calcinosis.
Collapse
Affiliation(s)
- Bhargavi Duvvuri
- Division of Rheumatology, University of Washington, Seattle, WA, USA.
| | - Lauren M Pachman
- Division of Pediatric Rheumatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; CureJM Center of Excellence, Ann & Robert H. Lurie Children's Hospital of Chicago and the Stanley Manne Simpson-Quarrey Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Payton Hermanson
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Ting Wang
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Richard Moore
- Cedars Sinai Med Ctr, Division of Rheumatology, Los Angeles, CA, USA
| | | | - Aaron Long
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Gabrielle A Morgan
- Division of Pediatric Rheumatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; CureJM Center of Excellence, Ann & Robert H. Lurie Children's Hospital of Chicago and the Stanley Manne Simpson-Quarrey Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington School of Medicine, Seattle, WA, USA
| | - Yasemin Sancak
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
3
|
Wang Y, Lu J, Liu Y. Skeletal Muscle Regeneration in Cardiotoxin-Induced Muscle Injury Models. Int J Mol Sci 2022; 23:ijms232113380. [PMID: 36362166 PMCID: PMC9657523 DOI: 10.3390/ijms232113380] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle injuries occur frequently in daily life and exercise. Understanding the mechanisms of regeneration is critical for accelerating the repair and regeneration of muscle. Therefore, this article reviews knowledge on the mechanisms of skeletal muscle regeneration after cardiotoxin-induced injury. The process of regeneration is similar in different mouse strains and is inhibited by aging, obesity, and diabetes. Exercise, microcurrent electrical neuromuscular stimulation, and mechanical loading improve regeneration. The mechanisms of regeneration are complex and strain-dependent, and changes in functional proteins involved in the processes of necrotic fiber debris clearance, M1 to M2 macrophage conversion, SC activation, myoblast proliferation, differentiation and fusion, and fibrosis and calcification influence the final outcome of the regenerative activity.
Collapse
|
4
|
Abstract
PURPOSE OF REVIEW The aim of this study was to provide updated information on the prevalence, pathogenesis, diagnostics and therapeutics of calcinosis cutis associated with systemic sclerosis (SSc). RECENT FINDINGS Observational studies show ethnic and geographical differences in the prevalence of calcinosis. In addition to clinical and serological associations, biochemical studies and in-vivo models have attempted to explain theories behind its pathogenesis, including prolonged state of inflammation, mechanical stress, hypoxia and dysregulation in bone and phosphate metabolism. Long-term use of proton pump inhibitors may increase the risk for calcinosis in SSc. Few single-centre observational studies have shown mild benefit with minocycline and topical sodium thiosulfate. SUMMARY Calcinosis cutis is the deposition of insoluble calcium in the skin and subcutaneous tissues. It affects up to 40% of SSc patients and causes significant morbidity. Long disease duration, features of vascular dysfunction and osteoporosis have been associated with calcinosis. Altered levels of inorganic pyrophosphate and fibroblast growth factor-23 have been implicated in dysregulated phosphate metabolism that may lead to calcinosis in SSc. Plain radiography can help with diagnosis and quantifying the calcinosis burden. Surgical treatment remains the most effective therapy when feasible. At present, no medical therapies have proven efficacy in large randomized controlled trials.
Collapse
Affiliation(s)
- Srijana Davuluri
- Stanford School of Medicine, Division of Immunology & Rheumatology, Palo Alto, California
| | - Christian Lood
- University of Washington, Division of Rheumatology, Seattle, Washington
| | - Lorinda Chung
- Stanford School of Medicine & Palo Alto VA Healthcare System, Division of Immunology & Rheumatology, Palo Alto, California, USA
| |
Collapse
|
5
|
Davuluri S, Duvvuri B, Lood C, Faghihi-Kashani S, Chung L. Calcinosis in dermatomyositis: Origins and possible therapeutic avenues. Best Pract Res Clin Rheumatol 2022; 36:101768. [PMID: 35803868 DOI: 10.1016/j.berh.2022.101768] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Calcinosis, insoluble calcium compounds deposited in skin and other tissues, is a crippling sequela of dermatomyositis. Prolonged disease associated with ongoing inflammation, ischemia, repetitive trauma, and certain autoantibodies are associated with calcinosis. Herein, we describe potential pathogenic mechanisms including the role of mitochondrial calcification. There are no widely effective treatments for calcinosis. We review available pharmacologic therapies for calcinosis including those targeting calcium and phosphorus metabolism; immunosuppressive/anti-inflammatory therapies; and vasodilators. Mounting evidence supports the use of various formulations of sodium thiosulfate in the treatment of calcinosis. Although the early institution of aggressive immunosuppression may prevent calcinosis in juvenile dermatomyositis, only limited data support improvement once it has developed. Minocycline can be useful particularly for lesions associated with surrounding inflammation. Powerful vasodilators, such as prostacyclin analogs, may have promise in the treatment of calcinosis, but further studies are necessary. Surgical removal of lesions when amenable is our treatment of choice.
Collapse
Affiliation(s)
- Srijana Davuluri
- Stanford School of Medicine, Division of Immunology &Rheumatology, 1000 Welch Road, Suite 204, Palo Alto, 94304, California, USA.
| | - Bhargavi Duvvuri
- University of Washington, Department of Medicine, Division of Rheumatology, 750 Republican Street, Seattle, WA, 98109, USA.
| | - Christian Lood
- University of Washington, Division of Rheumatology, 750 Republican Street, Room E-545, Seattle, WA, 98109, USA.
| | - Sara Faghihi-Kashani
- Stanford School of Medicine, Division of Immunology &Rheumatology, 1000 Welch Road, Suite 204, Palo Alto, 94304, California, USA.
| | - Lorinda Chung
- Stanford School of Medicine & Palo Alto VA Health Care System, Division of Immunology &Rheumatology, 1000 Welch Road, Suite 203, Palo Alto, 94304, California, USA.
| |
Collapse
|
6
|
Bohner M, Maazouz Y, Ginebra MP, Habibovic P, Schoenecker JG, Seeherman H, van den Beucken JJ, Witte F. Sustained local ionic homeostatic imbalance caused by calcification modulates inflammation to trigger heterotopic ossification. Acta Biomater 2022; 145:1-24. [PMID: 35398267 DOI: 10.1016/j.actbio.2022.03.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022]
Abstract
Heterotopic ossification (HO) is a condition triggered by an injury leading to the formation of mature lamellar bone in extraskeletal soft tissues. Despite being a frequent complication of orthopedic and trauma surgery, brain and spinal injury, the etiology of HO is poorly understood. The aim of this study is to evaluate the hypothesis that a sustained local ionic homeostatic imbalance (SLIHI) created by mineral formation during tissue calcification modulates inflammation to trigger HO. This evaluation also considers the role SLIHI could play for the design of cell-free, drug-free osteoinductive bone graft substitutes. The evaluation contains five main sections. The first section defines relevant concepts in the context of HO and provides a summary of proposed causes of HO. The second section starts with a detailed analysis of the occurrence and involvement of calcification in HO. It is followed by an explanation of the causes of calcification and its consequences. This allows to speculate on the potential chemical modulators of inflammation and triggers of HO. The end of this second section is devoted to in vitro mineralization tests used to predict the ectopic potential of materials. The third section reviews the biological cascade of events occurring during pathological and material-induced HO, and attempts to propose a quantitative timeline of HO formation. The fourth section looks at potential ways to control HO formation, either acting on SLIHI or on inflammation. Chemical, physical, and drug-based approaches are considered. Finally, the evaluation finishes with a critical assessment of the definition of osteoinduction. STATEMENT OF SIGNIFICANCE: The ability to regenerate bone in a spatially controlled and reproducible manner is an essential prerequisite for the treatment of large bone defects. As such, understanding the mechanism leading to heterotopic ossification (HO), a condition triggered by an injury leading to the formation of mature lamellar bone in extraskeletal soft tissues, would be very useful. Unfortunately, the mechanism(s) behind HO is(are) poorly understood. The present study reviews the literature on HO and based on it, proposes that HO can be caused by a combination of inflammation and calcification. This mechanism helps to better understand current strategies to prevent and treat HO. It also shows new opportunities to improve the treatment of bone defects in orthopedic and dental procedures.
Collapse
|
7
|
Radvar E, Griffanti G, Tsolaki E, Bertazzo S, Nazhat SN, Addison O, Mata A, Shanahan CM, Elsharkawy S. Engineered In vitro Models for Pathological Calcification: Routes Toward Mechanistic Understanding. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Elham Radvar
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| | - Gabriele Griffanti
- Department of Mining and Materials Engineering Faculty of Engineering McGill University Montreal QC H3A 0C5 Canada
| | - Elena Tsolaki
- Department of Medical Physics and Biomedical Engineering University College London London WC1E 6BT UK
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering University College London London WC1E 6BT UK
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering Faculty of Engineering McGill University Montreal QC H3A 0C5 Canada
| | - Owen Addison
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| | - Alvaro Mata
- School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
| | - Catherine M. Shanahan
- BHF Centre of Research Excellence Cardiovascular Division James Black Centre King's College London London SE1 1UL UK
| | - Sherif Elsharkawy
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| |
Collapse
|
8
|
Averin AS, Utkin YN. Cardiovascular Effects of Snake Toxins: Cardiotoxicity and Cardioprotection. Acta Naturae 2021; 13:4-14. [PMID: 34707893 PMCID: PMC8526186 DOI: 10.32607/actanaturae.11375] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Snake venoms, as complex mixtures of peptides and proteins, affect various vital systems of the organism. One of the main targets of the toxic components from snake venoms is the cardiovascular system. Venom proteins and peptides can act in different ways, exhibiting either cardiotoxic or cardioprotective effects. The principal classes of these compounds are cobra cardiotoxins, phospholipases A2, and natriuretic, as well as bradykinin-potentiating peptides. There is another group of proteins capable of enhancing angiogenesis, which include, e.g., vascular endothelial growth factors possessing hypotensive and cardioprotective activities. Venom proteins and peptides exhibiting cardiotropic and vasoactive effects are promising candidates for the design of new drugs capable of preventing or constricting the development of pathological processes in cardiovascular diseases, which are currently the leading cause of death worldwide. For example, a bradykinin-potentiating peptide from Bothrops jararaca snake venom was the first snake venom compound used to create the widely used antihypertensive drugs captopril and enalapril. In this paper, we review the current state of research on snake venom components affecting the cardiovascular system and analyse the mechanisms of physiological action of these toxins and the prospects for their medical application.
Collapse
Affiliation(s)
- A. S. Averin
- Institute of Cell Biophysics of the Russian Academy of Sciences PSCBR RAS, Pushchino, Moscow region, 142290 Russia
| | - Yu. N. Utkin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997 Russia
| |
Collapse
|
9
|
Phadwal K, Vrahnas C, Ganley IG, MacRae VE. Mitochondrial Dysfunction: Cause or Consequence of Vascular Calcification? Front Cell Dev Biol 2021; 9:611922. [PMID: 33816463 PMCID: PMC8010668 DOI: 10.3389/fcell.2021.611922] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are crucial bioenergetics powerhouses and biosynthetic hubs within cells, which can generate and sequester toxic reactive oxygen species (ROS) in response to oxidative stress. Oxidative stress-stimulated ROS production results in ATP depletion and the opening of mitochondrial permeability transition pores, leading to mitochondria dysfunction and cellular apoptosis. Mitochondrial loss of function is also a key driver in the acquisition of a senescence-associated secretory phenotype that drives senescent cells into a pro-inflammatory state. Maintaining mitochondrial homeostasis is crucial for retaining the contractile phenotype of the vascular smooth muscle cells (VSMCs), the most prominent cells of the vasculature. Loss of this contractile phenotype is associated with the loss of mitochondrial function and a metabolic shift to glycolysis. Emerging evidence suggests that mitochondrial dysfunction may play a direct role in vascular calcification and the underlying pathologies including (1) impairment of mitochondrial function by mineral dysregulation i.e., calcium and phosphate overload in patients with end-stage renal disease and (2) presence of increased ROS in patients with calcific aortic valve disease, atherosclerosis, type-II diabetes and chronic kidney disease. In this review, we discuss the cause and consequence of mitochondrial dysfunction in vascular calcification and underlying pathologies; the role of autophagy and mitophagy pathways in preventing mitochondrial dysfunction during vascular calcification and finally we discuss mitochondrial ROS, DRP1, and HIF-1 as potential novel markers and therapeutic targets for maintaining mitochondrial homeostasis in vascular calcification.
Collapse
Affiliation(s)
- Kanchan Phadwal
- Functional Genetics and Development Division, The Roslin Institute and The Royal (Dick) School of Veterinary Studies (R(D)SVS), University of Edinburgh, Midlothian, United Kingdom
| | - Christina Vrahnas
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, United Kingdom
| | - Ian G. Ganley
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, United Kingdom
| | - Vicky E. MacRae
- Functional Genetics and Development Division, The Roslin Institute and The Royal (Dick) School of Veterinary Studies (R(D)SVS), University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
10
|
Abstract
One of the most fascinating aspects of mitochondria is their remarkable ability to accumulate and store large amounts of calcium in the presence of phosphate leading to mitochondrial calcification. In this paper, we briefly address the mechanisms that regulate mitochondrial calcium homeostasis followed by the extensive review on the formation and characterization of intramitochondrial calcium phosphate granules leading to mitochondrial calcification and its relevance to physiological and pathological calcifications of body tissues.
Collapse
Affiliation(s)
- Bhargavi Duvvuri
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA 98195, USA
| | - Christian Lood
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
11
|
Alexander KA, Tseng HW, Salga M, Genêt F, Levesque JP. When the Nervous System Turns Skeletal Muscles into Bones: How to Solve the Conundrum of Neurogenic Heterotopic Ossification. Curr Osteoporos Rep 2020; 18:666-676. [PMID: 33085000 DOI: 10.1007/s11914-020-00636-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Neurogenic heterotopic ossification (NHO) is the abnormal formation of extra-skeletal bones in periarticular muscles after damage to the central nervous system (CNS) such as spinal cord injury (SCI), traumatic brain injury (TBI), stroke, or cerebral anoxia. The purpose of this review is to summarize recent developments in the understanding of NHO pathophysiology and pathogenesis. Recent animal models of NHO and recent findings investigating the communication between CNS injury, tissue inflammation, and upcoming NHO therapeutics are discussed. RECENT FINDINGS Animal models of NHO following TBI or SCI have shown that NHO requires the combined effects of a severe CNS injury and soft tissue damage, in particular muscular inflammation and the infiltration of macrophages into damaged muscles plays a key role. In the context of a CNS injury, the inflammatory response to soft tissue damage is exaggerated and persistent with excessive signaling via substance P-, oncostatin M-, and TGF-β1-mediated pathways. This review provides an overview of the known animal models and mechanisms of NHO and current therapeutic interventions for NHO patients. While some of the inflammatory mechanisms leading to NHO are common with other forms of traumatic and genetic heterotopic ossifications (HO), NHOs uniquely involve systemic changes in response to CNS injury. Future research into these CNS-mediated mechanisms is likely to reveal new targetable pathways to prevent NHO development in patients.
Collapse
Affiliation(s)
- Kylie A Alexander
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Hsu-Wen Tseng
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia
| | - Marjorie Salga
- Department of Physical Medicine and Rehabilitation, CIC 1429, Raymond Poincaré Hospital, APHP, Garches, France
- END:ICAP U1179 INSERM, University of Versailles Saint Quentin en Yvelines, UFR Simone Veil-Santé, Montigny le Bretonneux, France
| | - François Genêt
- Department of Physical Medicine and Rehabilitation, CIC 1429, Raymond Poincaré Hospital, APHP, Garches, France
- END:ICAP U1179 INSERM, University of Versailles Saint Quentin en Yvelines, UFR Simone Veil-Santé, Montigny le Bretonneux, France
| | - Jean-Pierre Levesque
- Mater Research Institute, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, Queensland, 4102, Australia.
| |
Collapse
|
12
|
Ibarra M, Rigsby C, Morgan GA, Sammet CL, Huang CC, Xu D, Targoff IN, Pachman LM. Monitoring change in volume of calcifications in juvenile idiopathic inflammatory myopathy: a pilot study using low dose computed tomography. Pediatr Rheumatol Online J 2016; 14:64. [PMID: 27894310 PMCID: PMC5127038 DOI: 10.1186/s12969-016-0123-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/16/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Dystrophic calcifications may occur in patients with J uvenile Idiopathic Inflammatory Myopathy (JIIM) as well as other connective tissue and metabolic diseases, but a reliable method of measuring the volume of these calcifications has not been established. The purpose of this study is to determine the feasibility of low dose, limited slice, Computed Tomography (CT) to measure objectively in-situ calcification volumes in patients with JIIM over time. METHODS Ten JIIM patients (eight JDM, two Overlap) with calcifications were prospectively recruited over a 2-year period to undergo two limited, low dose, four-slice CT scans. Calculation of the volume of calcifications used a CT post processing workstation. Additional patient data included: Disease Activity Scores (DAS), Childhood Myositis Assessment Scale (CMAS), myositis specific antibodies (MSA), and the TNFα-308 promoter region A/G polymorphism. Statistical analysis utilized the Pearson correlation coefficient, the paired t-test and descriptive statistics. RESULTS Ten JIIM, mean age 14.54 ± 4.54 years, had a duration of untreated disease of 8.68 ± 5.65 months MSA status: U1RNP (1), PM-Scl (1), Ro (1, 4 indeterminate), p155/140 (2), MJ (3), Mi-2 indeterminate (1), negative (3). 4/8 JDM (50%) were TNF-α-308 A+. Overall, the calcification volumes tended to decrease from the first to the second CT study by 0.5 cm3 (from 2.79 ± 1.98 cm3 to 2.29 ± 2.25 cm3). The average effective radiation dose was 0.007 ± 0.002, 0.010 ± 0.005, and 0.245 mSv for the upper extremity, lower extremity and chest, respectively (compared to a standard chest x-ray-- 0.02mSV effective dosage). CONCLUSION We conclude: 1) the limited low dose CT technique provides objective data about volume of the calcifications in JIIM; 2) measuring the volume of calcifications in an extremity is associated with minimal radiation exposure; 3) This method may be useful to evaluate the efficacy of therapies for JIIM dystrophic calcification.
Collapse
Affiliation(s)
- Maria Ibarra
- Division of Pediatric Rheumatology, Children’s Mercy Hospital , 2401 Gillham Road, Kansas City, Missouri 64108-4619 USA
| | - Cynthia Rigsby
- Department of Medical Imaging, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL USA ,Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Gabrielle A. Morgan
- Cure JM Center of Excellence, Stanley Manne Research Institute affiliated with Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 East Chicago Avenue, Box 212, Chicago, IL 60611 USA
| | - Christina L. Sammet
- Department of Medical Imaging, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL USA ,Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Chiang-Ching Huang
- Joseph J. Zilber School of Public Health, University of Wisconsin, Milwaukee, WI USA
| | - Dong Xu
- Cure JM Center of Excellence, Stanley Manne Research Institute affiliated with Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 East Chicago Avenue, Box 212, Chicago, IL 60611 USA
| | - Ira N. Targoff
- The Department of Internal Medicine, The University of Oklahoma, Norman, OK USA
| | - Lauren M. Pachman
- Cure JM Center of Excellence, Stanley Manne Research Institute affiliated with Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 East Chicago Avenue, Box 212, Chicago, IL 60611 USA
| |
Collapse
|
13
|
Garza-Rodea ASDL, Boersma H, Dambrot C, Vries AAFD, Bekkum DWV, Knaän-Shanzer S. Barriers in contribution of human mesenchymal stem cells to murine muscle regeneration. World J Exp Med 2015; 5:140-153. [PMID: 25992329 PMCID: PMC4436938 DOI: 10.5493/wjem.v5.i2.140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/31/2014] [Accepted: 02/09/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To study regeneration of damaged human and murine muscle implants and the contribution of added xenogeneic mesenchymal stem cells (MSCs).
METHODS: Minced human or mouse skeletal muscle tissues were implanted together with human or mouse MSCs subcutaneously on the back of non-obese diabetic/severe combined immunodeficient mice. The muscle tissues (both human and murine) were minced with scalpels into small pieces (< 1 mm3) and aliquoted in portions of 200 mm3. These portions were either cryopreserved in 10% dimethylsulfoxide or freshly implanted. Syngeneic or xenogeneic MSCs were added to the minced muscles directly before implantation. Implants were collected at 7, 14, 30 or 45 d after transplantation and processed for (immuno)histological analysis. The progression of muscle regeneration was assessed using a standard histological staining (hematoxylin-phloxin-saffron). Antibodies recognizing Pax7 and von Willebrand factor were used to detect the presence of satellite cells and blood vessels, respectively. To enable detection of the bone marrow-derived MSCs or their derivatives we used MSCs previously transduced with lentiviral vectors expressing a cytoplasmic LacZ gene. X-gal staining of the fixed tissues was used to detect β-galactosidase-positive cells and myofibers.
RESULTS: Myoregeneration in implants of fresh murine muscle was evident as early as day 7, and progressed with time to occupy 50% to 70% of the implants. Regeneration of fresh human muscle was slower. These observations of fresh muscle implants were in contrast to the regeneration of cryopreserved murine muscle that proceeded similarly to that of fresh tissue except for day 45 (P < 0.05). Cryopreserved human muscle showed minimal regeneration, suggesting that the freezing procedure was detrimental to human satellite cells. In fresh and cryopreserved mouse muscle supplemented with LacZ-tagged mouse MSCs, β-galactosidase-positive myofibers were identified early after grafting at the well-vascularized periphery of the implants. The contribution of human MSCs to murine myofiber formation was, however, restricted to the cryopreserved mouse muscle implants. This suggests that fresh murine muscle tissue provides a suboptimal environment for maintenance of human MSCs. A detailed analysis of the histological sections of the various muscle implants revealed the presence of cellular structures with a deviating morphology. Additional stainings with alizarin red and alcian blue showed myofiber calcification in 50 of 66 human muscle implants, and encapsulated cartilage in 10 of 81 of murine muscle implants, respectively.
CONCLUSION: In mouse models the engagement of human MSCs in myoregeneration might be underestimated. Furthermore, our model permits the dissection of species-specific factors in the microenvironment.
Collapse
|
14
|
Chen J, Peacock JR, Branch J, David Merryman W. Biophysical analysis of dystrophic and osteogenic models of valvular calcification. J Biomech Eng 2015; 137:020903. [PMID: 25405546 DOI: 10.1115/1.4029115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Indexed: 12/27/2022]
Abstract
Calcific aortic valve disease (CAVD) is a significant cardiovascular disorder characterized by the formation of calcific nodules (CN) on the valve. In vitro assays studying the formation of these nodules were developed and have led to many significant mechanistic findings; however, the biophysical properties of CNs have not been clearly defined. A thorough analysis of dystrophic and osteogenic nodules utilizing scanning electron microscopy (SEM), energy dispersive spectrometry (EDS), and atomic force microscopy (AFM) was conducted to describe calcific nodule properties and provide a link between calcific nodule morphogenesis in vitro and in vivo. Unique nodule properties were observed for dystrophic and osteogenic nodules, highlighting the distinct mechanisms occurring in valvular calcification.
Collapse
|
15
|
Exacerbated skeletal muscle inflammation and calcification in the acute phase of infection by Mexican Trypanosoma cruzi DTUI strain. BIOMED RESEARCH INTERNATIONAL 2014; 2014:450389. [PMID: 24991553 PMCID: PMC4060783 DOI: 10.1155/2014/450389] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/09/2014] [Indexed: 12/14/2022]
Abstract
A murine model was used to study the histopathological aspects and cytokine expression levels in skeletal muscle provoked by the infection with Mexican TcI strains. BALB/c mice were inoculated with the virulent Querétaro strain and the nonvirulent Ninoa strain. Parasite numbers were counted in blood and skeletal muscle at different times post-infection, and real time-PCR expression levels of the cytokines IL-12, IL-4, IL-10, IFN- γ , and TNF- α were evaluated. In the acute phase of infection, a high parasitic load, both in blood and skeletal muscle, was detected. The histopathological analyses showed an exacerbated inflammation and granulomatous-like infiltrate with the Querétaro strain. Interestingly, extensive calcification areas were observed in the skeletal muscle surrounded by inflammatory infiltrates. TNF- α and IL-10 expression exhibited a significant increase at the peak of infection. In summary, Querétaro strain, a Mexican TcI strain, is virulent enough to induce high inflammation and calcification in skeletal muscle of the hind limbs, which could be related to high expression levels of TNF- α .
Collapse
|
16
|
Sowa AK, Kaiser FJ, Eckhold J, Kessler T, Aherrahrou R, Wrobel S, Kaczmarek PM, Doehring L, Schunkert H, Erdmann J, Aherrahrou Z. Functional interaction of osteogenic transcription factors Runx2 and Vdr in transcriptional regulation of Opn during soft tissue calcification. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:60-8. [PMID: 23644099 DOI: 10.1016/j.ajpath.2013.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 03/11/2013] [Accepted: 03/12/2013] [Indexed: 01/31/2023]
Abstract
Loss of Abcc6 gene expression was identified to be responsible for dystrophic calcification of the heart (DCC) or vessels after acute injury in several strains of laboratory mice. This calcification shares features with osteogenesis and may involve osteogenic factors. Tissue expression of osteopontin (Opn) and 11 osteogenic transcription factors were studied in vivo in mouse models for DCC and in vitro using luciferase reporter gene assays. Compared with DCC-resistant C57BL/6 mice, a significant increase in Opn transcription was demonstrated in necrotic lesions of both DCC-susceptible C3H/He and B6.C3H(Dyscalc1) congenic mice at day 3 after injury. Significant increases in gene expression were also demonstrated for the transcription factors runt domain-containing transcription factor 2 (Runx2), vitamin D receptor (Vdr), SRY (sex-determining region Y)-box 9 protein, and Nfkb1 in C3H/He mice versus C57BL/6 controls. However, only Runx2 remained significantly increased in the B6.C3H(Dyscalc1) congenic mice, which carry only the Dyscalc1 locus with functional Abcc6 deletion on a C57BL/6 genetic background. Luciferase assay use increased Opn promoter activity, which was demonstrated after overexpression of Runx2. A poly-T stretch insertion was identified to stabilize the binding of Runx2, thus significantly enhancing Opn promoter activity. This Runx2-mediated activation was further enhanced by cotransfection with Vdr. Our data suggest a key role of Runx2 in the regulation of Opn in a model of cardiovascular calcification and demonstrate a synergistic cooperation of Runx2 and Vdr.
Collapse
Affiliation(s)
- Ann-Kathrin Sowa
- Institute for Integrative and Experimental Genomics and DZHK-German Centre for Cardiovascular Research, partner site Lübeck/Hamburg/Kiel, University of Lübeck, Lübeck, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Glass AM, Coombs W, Taffet SM. Spontaneous cardiac calcinosis in BALB/cByJ mice. Comp Med 2013; 63:29-37. [PMID: 23561935 PMCID: PMC3567374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 06/19/2012] [Accepted: 07/16/2012] [Indexed: 06/02/2023]
Abstract
BALB/c mice are predisposed to dystrophic cardiac calcinosis-the mineralization of cardiac tissues, especially the right ventricular epicardium. In previous reports, the disease appeared in aged animals and had an unknown etiology. In the current study, we report a substrain of BALB/c mice (BALB/cByJ) that develops disease early and with high frequency. Here we analyzed hearts grossly to identify the presence and measure the severity of disease and to compare BALB/c substrains. Histologic analysis and fluorescent and immunofluorescent microscopy were used to characterize the calcinotic lesions. BALB/cByJ mice exhibited more frequent and severe calcium deposition than did BALB/c mice of other substrains (90% compared with 3% at 5 wk). At this age, lesions covered an average of 30% of the total ventricular surface area in BALB/cByJ mice, compared with less than 1% in other strains. In bone-marrow-chimeric mice, green fluorescent protein was used as a marker to show that the lesions contain an infiltration of cells of bone marrow origin. Lesion histology showed that calcium deposits were surrounded by fibrosis with interspersed immune cells. Lymphocytes, macrophages, and granulocytes were all present. Internalization of the gap-junction protein connexin 43 was observed in myocytes adjacent to lesions. In conclusion, BALB/cByJ mice exhibit more frequent and severe dystrophic cardiac calcinosis than do other BALB/c substrains. Our findings suggest that immune cells are actively recruited to lesions and that myocyte gap junctions are altered near lesions.
Collapse
|
18
|
Abstract
PURPOSE Gentamicin is a widely employed antibiotic, but may reduce calcium uptake by eukaryotic cells. This study was conducted to determine whether gentamicin reduces calcification by porcine aortic valvular interstitial cells (pAVICs) grown in 2D culture, which is a common model for calcific aortic valve disease (CAVD). METHODS AND RESULTS The presence of gentamicin (up to 0.2 mM) in the medium of pAVICs cultured for 8 days significantly lowered calcification and alkaline phosphatase content in a dose-dependent manner compared to pAVICs cultured without gentamicin. Gentamicin also significantly increased cell proliferation and apoptosis at concentrations of 0.1-0.2 mM. Next, gentamicin was applied to previously calcified pAVIC cultures (grown for 8 days) to determine whether it could stop or reverse the calcification process. Daily application of gentamicin for 8 additional days significantly reduced calcification to below the pre-calcification levels. CONCLUSIONS These results confirm that gentamicin should be used cautiously with in vitro studies of calcification, and suggest that gentamicin may have the ability to reverse calcification by pAVICs. Given the nephrotoxicity and ototoxicity of this antibiotic, its clinical potential for the treatment of calcification in heart valves is limited. However, further investigation of the pathways through which gentamicin alters calcium uptake by valvular cells may provide insight into novel therapies for CAVD.
Collapse
|
19
|
Evans ND, Swain RJ, Gentleman E, Gentleman MM, Stevens MM. Gene-expression analysis reveals that embryonic stem cells cultured under osteogenic conditions produce mineral non-specifically compared to marrow stromal cells or osteoblasts. Eur Cell Mater 2012; 24:211-23. [PMID: 23007907 PMCID: PMC5833941 DOI: 10.22203/ecm.v024a15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Pluripotent cells, such as embryonic stem cells (ESCs), divide indefinitely and can differentiate to form mineralised nodules in response to osteogenic supplements. This suggests that they may be used as a cell source for bone replacement strategies. Here, we related the expression of osteogenic and chondrogenic genes in cultures of murine ESCs, marrow stromal cells (MSCs) and calvarial osteoblasts (OBs) cultured under osteogenic conditions to the biochemical composition and quantity of mineral formed. Mineralisation, measured by calcium sequestration, was >2-fold greater in ESC cultures than in either MSCs or OBs. Micro-Raman spectroscopy and spectral mapping revealed a lower mineral-to-matrix ratio and confirmed a more diffuse pattern of mineralisation in ESCs compared to MSCs and OBs. Baseline expression of chondrogenic and osteogenic genes was between 1 and 4 orders of magnitude greater in MSCs and OBs than in ESCs. Osteogenic culture of MSCs and OBs was accompanied by increases in osteogenic gene expression by factors of ~100 compared to only ~10 in ESCs. Consequentially, peak expression of osteogenic and chondrogenic genes was greater in MSCs and OBs than ESCs by factors of 100-1000, despite the fact that mineralisation was more extensive in ESCs than either MSCs or OBs. We also observed significant cell death in ESC nodules. We conclude that the mineralised material observed in cultures of murine ESCs during osteogenic differentiation may accumulate non-specifically, perhaps in necrotic cell layers, and that thorough characterisation of the tissue formed by ESCs must be achieved before these cells can be considered as a cell source for clinical applications.
Collapse
Affiliation(s)
- Nicholas D. Evans
- Department of Materials, Imperial College London, South Kensington, London SW7 2AZ, UK,Institute of Biomedical Engineering, Imperial College London, South Kensington, London SW7 2AZ, UK
| | - Robin J. Swain
- Department of Materials, Imperial College London, South Kensington, London SW7 2AZ, UK,Institute of Biomedical Engineering, Imperial College London, South Kensington, London SW7 2AZ, UK
| | - Eileen Gentleman
- Department of Materials, Imperial College London, South Kensington, London SW7 2AZ, UK,Institute of Biomedical Engineering, Imperial College London, South Kensington, London SW7 2AZ, UK
| | - Molly M. Gentleman
- Mechanical Engineering Department, Texas A&M University, College Station TX 77843, USA
| | - Molly M. Stevens
- Department of Materials, Imperial College London, South Kensington, London SW7 2AZ, UK,Institute of Biomedical Engineering, Imperial College London, South Kensington, London SW7 2AZ, UK,To whom correspondence should be addressed.
Tel: +44 (0)20 7594 6804; Fax: +44 (0)20 7594 6757.
| |
Collapse
|
20
|
Harris NL, Huffer WE, von Stade E, Larson AI, Phinney S, Purnell ML. The effect of platelet-rich plasma on normal soft tissues in the rabbit. J Bone Joint Surg Am 2012; 94:786-93. [PMID: 22552667 DOI: 10.2106/jbjs.j.00984] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Platelet-rich plasma is reported to contain multiple growth factors, and has been utilized in orthopaedic surgery to aid healing in multiple tissues. To date, the use of autologous platelet-rich plasma has not been studied for its effects on normal soft tissue. METHODS Eighteen adult New Zealand White rabbits were injected with 0.5 mL of autologous platelet-rich plasma in the right or left quadriceps muscle, Achilles tendon, medial collateral ligament, subcutaneous tissue, tibial periosteum, and ankle joint. Saline solution was injected on the contralateral side as a control. The soft tissues were examined histologically at two weeks (six rabbits) and six weeks (six rabbits), and soft tissues from six rabbits that had been reinjected at six weeks were examined at twelve weeks. RESULTS Inflammatory skin lesions were visible at forty-eight hours at superficial platelet-rich plasma sites. All lesions resolved by six days. Compared with findings in control specimens, histological analysis of platelet-rich plasma injection sites at two weeks showed a marked inflammatory infiltrate with lymphocytic and monocytic predominance. Intra-articular injection showed villous synovial hyperplasia and chronic synovitis. Tendon and ligament sites showed new collagen deposition. Intramuscular injection sites showed thrombosis, necrosis, and calcium deposition. Subcutaneous sites also showed calcium deposition without necrosis as well as collagen nodules representing early scar tissue. Histological examination of platelet-rich plasma injection sites at six and twelve weeks demonstrated a persistent but diminished inflammatory infiltrate. Focal areas of scar tissue were seen with fibroblasts, collagen formation, and neovascularity. All saline solution sites at all times were nonreactive. CONCLUSIONS Platelet-rich plasma can initiate an inflammatory response in the absence of an inciting injury in normal soft tissue in rabbits.
Collapse
Affiliation(s)
- N Lindsay Harris
- Aspen Orthopaedic Associates, 100 East Main Street, Suite 101, Aspen, CO 81611, USA
| | | | | | | | | | | |
Collapse
|
21
|
Maetzler W, Berg D, Funke C, Sandmann F, Stünitz H, Maetzler C, Nitsch C. Progressive secondary neurodegeneration and microcalcification co-occur in osteopontin-deficient mice. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:829-39. [PMID: 20522649 DOI: 10.2353/ajpath.2010.090798] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the brain, osteopontin (OPN) may function in a variety of pathological conditions, including neurodegeneration, microcalcification, and inflammation. In this study, we addressed the role of OPN in primary and secondary neurodegeneration, microcalcification, and inflammation after an excitotoxic lesion by examining OPN knock-out (KO) mice. Two, four, and ten weeks after injection of the glutamate analogue ibotenate into the corticostriatal boundary, the brains of 12 mice per survival time and strain were evaluated. OPN was detectable in neuron-shaped cells, in microglia, and at the surface of dense calcium deposits. At this primary lesion site, although the glial reaction was attenuated in OPN-KO mice, lesion size and presence of microcalcification were comparable between OPN-KO and wild-type mice. In contrast, secondary neurodegeneration at the thalamus was more prominent in OPN-KO mice, and this difference increased over time. This was paralleled by a dramatic rise in the regional extent of dense microcalcification. Despite these differences, the numbers of glial cells did not significantly differ between the two strains. This study demonstrates for the first time a genetic model with co-occurrence of neurodegeneration and microcalcification, mediated by the lack of OPN, and suggests a basic involvement of OPN action in these conditions. In the case of secondary retrograde or transneuronal degeneration, OPN may have a protective role as intracellular actor.
Collapse
Affiliation(s)
- Walter Maetzler
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, German Center for Neurodegenerative Diseases, Tuebingen, Germany.
| | | | | | | | | | | | | |
Collapse
|