1
|
Moumin NA, D'Vaz N, Kidd C, MacRae A, Zhou SJ, Richards T, Palmer DJ, Grzeskowiak LE, Sullivan TR, Green TJ. Urinary Ferritin as a Noninvasive Means of Assessing Iron Status in Young Children. J Nutr 2024; 154:2688-2695. [PMID: 38729575 DOI: 10.1016/j.tjnut.2024.04.040] [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: 01/26/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Iron deficiency (ID) is the most common nutritional deficiency affecting young children. Serum ferritin concentration is the preferred biomarker for measuring iron status because it reflects iron stores; however, blood collection can be distressing for young children and can be logistically difficult. A noninvasive means to measure iron status would be attractive to either diagnose or screen for ID in young children. OBJECTIVES This study aimed to determine the correlation between urinary and serum ferritin concentrations in young children; to determine whether correcting urinary ferritin for creatinine and specific gravity improves the correlation; and to determine a urine ferritin cut point to predict ID. METHODS Validation study was conducted using paired serum and urine collected from 3-y-old children (n = 142) participating in a longitudinal birth cohort study: the ORIGINS project in Perth, Western Australia. We calculated the sensitivity, specificity, positive, and negative predictive values of urinary ferritin amount in identifying those with ID at the clinical cut point used by the World Health Organization (serum ferritin concentration of <12 ng/mL). RESULTS Urine ferritin, corrected for creatinine, correlated moderately with serum ferritin [r = 0.53 (0.40-0.64)] and performed well in predicting those with ID (area under the curve: 0.85; 95% confidence interval: 0.75, 0.94). Urine ferritin <2.28 ng/mg creatinine was sensitive (86%) and specific (77%) in predicting ID and had a high negative predictive value of 97%; however, the positive predictive value was low (40%) owing to the low prevalence of ID in the sample (16%). CONCLUSIONS Urine ferritin shows good diagnostic performance for ID. This noninvasive biomarker maybe a useful screening tool to exclude ID in healthy young children; however, further research is needed in other populations.
Collapse
Affiliation(s)
- Najma A Moumin
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Discipline of Pediatrics, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Nina D'Vaz
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Courtney Kidd
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Andrea MacRae
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Shao J Zhou
- School of Agriculture, Food & Wine, The University of Adelaide, Urrbrae, South Australia, Australia; Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Toby Richards
- School of Medicine, Division of Surgery, University of Western Australia, Perth, Western Australia, Australia
| | - Debra J Palmer
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia; School of Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Luke E Grzeskowiak
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Thomas R Sullivan
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; Discipline of Public Health, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Tim J Green
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
2
|
Boehler JF, Brown KJ, Ricotti V, Morris CA. N-terminal titin fragment: a non-invasive, pharmacodynamic biomarker for microdystrophin efficacy. Skelet Muscle 2024; 14:2. [PMID: 38229112 PMCID: PMC10790446 DOI: 10.1186/s13395-023-00334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 12/29/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Multiple clinical trials to assess the efficacy of AAV-directed gene transfer in participants with Duchenne muscular dystrophy (DMD) are ongoing. The success of these trials currently relies on standard functional outcome measures that may exhibit variability within and between participants, rendering their use as sole measures of drug efficacy challenging. Given this, supportive objective biomarkers may be useful in enhancing observed clinical results. Creatine kinase (CK) is traditionally used as a diagnostic biomarker of DMD, but its potential as a robust pharmacodynamic (PD) biomarker is difficult due to the wide variability seen within the same participant over time. Thus, there is a need for the discovery and validation of novel PD biomarkers to further support and bolster traditional outcome measures of efficacy in DMD. METHOD Potential PD biomarkers in DMD participant urine were examined using a proteomic approach on the Somalogic platform. Findings were confirmed in both mdx mice and Golden Retriever muscular dystrophy (GRMD) dog plasma samples. RESULTS Changes in the N-terminal fragment of titin, a well-known, previously characterized biomarker of DMD, were correlated with the expression of microdystrophin protein in mice, dogs, and humans. Further, titin levels were sensitive to lower levels of expressed microdystrophin when compared to CK. CONCLUSION The measurement of objective PD biomarkers such as titin may provide additional confidence in the assessment of the mechanism of action and efficacy in gene therapy clinical trials of DMD. TRIAL REGISTRATION ClinicalTrials.gov NCT03368742.
Collapse
Affiliation(s)
- Jessica F Boehler
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA, 02129, USA.
| | - Kristy J Brown
- Rejuvenate Bio, 11425 Sorrento Valley Road, San Diego, CA, 92121, USA
| | - Valeria Ricotti
- National Institute for Health and Care Research Great Ormond Street Hospital Biomedical Research Centre/University College London Great Ormond Street Institute of Child Health, London, UK
| | - Carl A Morris
- PHDL Consulting LLC, 43 Sylvanus Wood Lane, Woburn, MA, 01801, USA
| |
Collapse
|
3
|
Dabaj I, Ducatez F, Marret S, Bekri S, Tebani A. Neuromuscular disorders in the omics era. Clin Chim Acta 2024; 553:117691. [PMID: 38081447 DOI: 10.1016/j.cca.2023.117691] [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: 09/21/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023]
Abstract
Neuromuscular disorders encompass a spectrum of conditions characterized by primary lesions within the peripheral nervous system, which include the anterior horn cell, peripheral nerve, neuromuscular junction, and muscle. In pediatrics, most of these disorders are linked to genetic causes. Despite the considerable progress, the diagnosis of these disorders remains a challenging due to wide clinical presentation, disease heterogeneity and rarity. It is noteworthy that certain neuromuscular disorders, once deemed untreatable, can now be effectively managed through novel therapies. Biomarkers emerge as indispensable tools, serving as objective measures that not only refine diagnostic accuracy but also provide guidance for therapeutic decision-making and the ongoing monitoring of long-term outcomes. Herein a comprehensive review of biomarkers in neuromuscular disorders is provided. We highlight the role of omics-based technologies that further characterize neuromuscular pathophysiology as well as identify potential therapeutic targets to guide treatment strategies.
Collapse
Affiliation(s)
- Ivana Dabaj
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France.
| | - Franklin Ducatez
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France
| | - Soumeya Bekri
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, F-76000 Rouen, France
| | - Abdellah Tebani
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, F-76000 Rouen, France
| |
Collapse
|
4
|
Fortunato F, Ferlini A. Biomarkers in Duchenne Muscular Dystrophy: Current Status and Future Directions. J Neuromuscul Dis 2023; 10:987-1002. [PMID: 37545256 PMCID: PMC10657716 DOI: 10.3233/jnd-221666] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2023] [Indexed: 08/08/2023]
Abstract
Duchenne muscular dystrophy is a severe, X-linked disease characterized by decreased muscle mass and function in children. Genetic and biochemical research over the years has led to the characterization of the cause and the pathophysiology of the disease. Moreover, the elucidation of genetic mechanisms underlining Duchenne muscular dystrophy has allowed for the design of innovative personalized therapies.The identification of specific, accurate, and sensitive biomarkers is becoming crucial for evaluating muscle disease progression and response to therapies, disease monitoring, and the acceleration of drug development and related regulatory processes.This review illustrated the up-to-date progress in the development of candidate biomarkers in DMD at the level of proteins, metabolites, micro-RNAs (miRNAs) and genetic modifiers also highlighting the complexity of translating research results to clinical practice.We highlighted the challenges encountered in translating biomarkers into the clinical context and the existing bottlenecks hampering the adoption of biomarkers as surrogate endpoints. These challenges could be overcome by national and international collaborative efforts, multicenter data sharing, definition of public biobanks and patients' registries, and creation of large cohorts of patients. Novel statistical tools/ models suitable to analyze small patient numbers are also required.Finally, collaborations with pharmaceutical companies would greatly benefit biomarker discovery and their translation in clinical trials.
Collapse
Affiliation(s)
- Fernanda Fortunato
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
5
|
Riddell DO, Hildyard JCW, Harron RCM, Wells DJ, Piercy RJ. Longitudinal assessment of blood-borne musculoskeletal disease biomarkers in the DE50-MD dog model of Duchenne muscular dystrophy. Wellcome Open Res 2022; 6:354. [PMID: 35600245 PMCID: PMC9111359 DOI: 10.12688/wellcomeopenres.17398.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 01/01/2024] Open
Abstract
Background: Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease caused by mutations in the dystrophin gene. Due to their phenotypic similarity to human patients, large animal models are invaluable tools for pre-clinical trials. The DE50-MD dog is a relatively new model of DMD, and carries a therapeutically-tractable mutation lying within the hotspot for human patients, making it especially valuable. Prior to conducting therapeutic trials using this novel animal model, it is essential to establish a panel of viable biomarkers. Methods: We evaluated a panel of blood-borne biomarkers of musculoskeletal disease in the DE50-MD dog. Venous blood samples were obtained monthly throughout an 18-month study period in DE50-MD (N=18) and wild-type (WT) control (N=14) dogs. A panel of potential plasma/serum biomarkers of DMD was measured and their theoretical utility in future clinical trials determined using sample size calculations. Results: Compared to WT dogs, DE50-MD dogs had substantially higher circulating creatine kinase (CK) activities, myomesin-3 (MYOM3), and the dystromiRs miR-1, miR-133a and miR-206, but significantly lower serum myostatin concentrations. An age-associated pattern, similar to that observed in DMD patients, was seen for CK and MYOM3. Sample size calculations suggested that low cohort sizes (N≤3) could be used to detect up to a 50% improvement in DE50-MD results towards WT levels for each biomarker or a combination thereof (via principal component analysis); as few as N=3 animals should enable detection of a 25% improvement using a combined biomarker approach (alpha 0.05, power 0.8). Conclusions: We have established a panel of blood-borne biomarkers that could be used to monitor musculoskeletal disease or response to a therapeutic intervention in the DE50-MD dog using low numbers of animals. The blood biomarker profile closely mimics that of DMD patients, supporting the hypothesis that this DMD model would be suitable for use in pre-clinical trials.
Collapse
Affiliation(s)
- Dominique O. Riddell
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - Rachel C. M. Harron
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW10TU, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| |
Collapse
|
6
|
Riddell DO, Hildyard JCW, Harron RCM, Wells DJ, Piercy RJ. Longitudinal assessment of blood-borne musculoskeletal disease biomarkers in the DE50-MD dog model of Duchenne muscular dystrophy. Wellcome Open Res 2022; 6:354. [PMID: 35600245 PMCID: PMC9111359 DOI: 10.12688/wellcomeopenres.17398.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease caused by mutations in the dystrophin gene. Due to their phenotypic similarity to human patients, large animal models are invaluable tools for pre-clinical trials. The DE50-MD dog is a relatively new model of DMD, and carries a therapeutically-tractable mutation lying within the hotspot for human patients, making it especially valuable. Prior to conducting therapeutic trials using this novel animal model, it is essential to establish a panel of viable biomarkers. Methods: We evaluated a panel of blood-borne biomarkers of musculoskeletal disease in the DE50-MD dog. Venous blood samples were obtained monthly throughout an 18-month study period in DE50-MD (N=18) and wild-type (WT) control (N=14) dogs. A panel of potential plasma/serum biomarkers of DMD was measured and their theoretical utility in future clinical trials determined using sample size calculations. Results: Compared to WT dogs, DE50-MD dogs had substantially higher circulating creatine kinase (CK) activities, myomesin-3 (MYOM3), and the dystromiRs miR-1, miR-133a and miR-206, but significantly lower serum myostatin concentrations. An age-associated pattern, similar to that observed in DMD patients, was seen for CK and MYOM3. Sample size calculations suggested that low cohort sizes (N≤3) could be used to detect up to a 50% improvement in DE50-MD results towards WT levels for each biomarker or a combination thereof (via principal component analysis); as few as N=3 animals should enable detection of a 25% improvement using a combined biomarker approach (alpha 0.05, power 0.8). Conclusions: We have established a panel of blood-borne biomarkers that could be used to monitor musculoskeletal disease or response to a therapeutic intervention in the DE50-MD dog using low numbers of animals. The blood biomarker profile closely mimics that of DMD patients, supporting the hypothesis that this DMD model would be suitable for use in pre-clinical trials.
Collapse
Affiliation(s)
- Dominique O. Riddell
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - Rachel C. M. Harron
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW10TU, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW This review highlights the key studies investigating various types of biomarkers in Duchenne muscular dystrophy (DMD). RECENT FINDINGS Several proteomic and metabolomic studies have been undertaken in both human DMD patients and animal models of DMD that have identified potential biomarkers in DMD. Although there have been a number of proteomic and metabolomic studies that have identified various potential biomarkers in DMD, more definitive studies still need to be undertaken in DMD patients to firmly correlate these biomarkers with diagnosis, disease progression, and monitoring the effects of novel treatment strategies being developed.
Collapse
Affiliation(s)
- Theo Lee-Gannon
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xuan Jiang
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tara C Tassin
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Pradeep P A Mammen
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Heart Failure, Ventricular Assist Device & Heart Transplant Program, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
8
|
Ohlendieck K, Swandulla D. Complexity of skeletal muscle degeneration: multi-systems pathophysiology and organ crosstalk in dystrophinopathy. Pflugers Arch 2021; 473:1813-1839. [PMID: 34553265 PMCID: PMC8599371 DOI: 10.1007/s00424-021-02623-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disorder due to primary abnormalities in one of the largest genes in the human genome, the DMD gene, which encodes various tissue-specific isoforms of the protein dystrophin. Although dystrophinopathies are classified as primary neuromuscular disorders, the body-wide abnormalities that are associated with this disorder and the occurrence of organ crosstalk suggest that a multi-systems pathophysiological view should be taken for a better overall understanding of the complex aetiology of X-linked muscular dystrophy. This article reviews the molecular and cellular effects of deficiency in dystrophin isoforms in relation to voluntary striated muscles, the cardio-respiratory system, the kidney, the liver, the gastrointestinal tract, the nervous system and the immune system. Based on the establishment of comprehensive biomarker signatures of X-linked muscular dystrophy using large-scale screening of both patient specimens and genetic animal models, this article also discusses the potential usefulness of novel disease markers for more inclusive approaches to differential diagnosis, prognosis and therapy monitoring that also take into account multi-systems aspects of dystrophinopathy. Current therapeutic approaches to combat muscular dystrophy are summarised.
Collapse
Affiliation(s)
- Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Co. Kildare, Maynooth, W23F2H6, Ireland.
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Co. Kildare, Maynooth, W23F2H6, Ireland.
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
9
|
Review of novel liquid-based biomarkers for prostate cancer: towards personalised and targeted medicine. JOURNAL OF RADIOTHERAPY IN PRACTICE 2021. [DOI: 10.1017/s1460396921000248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Abstract
Background:
Prostate cancer is the most commonly diagnosed cancer in men and it is responsible for about 10% of all cancer mortalities in both American and Canadian men. At present, serum prostate-specific antigen levels remain the most commonly used test to detect prostate cancer, and the standard and definitive diagnosis of the disease is via prostate biopsy. Conventional tissue biopsies are usually invasive, expensive, painful, time-consuming, and unsuitable for screening and need to be consistently evaluated by expert pathologists and have limited repeatability. Consequently, liquid biopsies are emerging as a favourable alternative to conventional tissue biopsies, providing a non-invasive and cost-effective approach for screening, diagnosis, treatment and monitoring of prostate cancer patients.
Materials and methods:
We searched several databases from August to December 2020 for relevant studies published in English between 2000 and 2020 and reporting on liquid-based biomarkers available in detectable quantities in patient bodily fluid samples. In this narrative review paper, we describe seven novel and promising liquid-based biomarkers that potentially account for individual patient variability as well as used in disease risk assessment, screening for early disease detection and diagnosis, identification of patients’ risk for metastatic disease and subsequent relapse, monitoring patient response to specific treatment and providing clinicians the potential to stratify patients likely to benefit from a particular treatment.
Conclusions:
The concept of precision medicine from prevention to treatment techniques that take individual patient variability into account will depend on the development of effective clinical biomarkers that interrogate key aberrant pathways potentially targetable with molecular targets or immunologic therapies. Liquid-based biomarkers with high sensitivity and specificity for prostate cancer are emerging as minimally invasive, lower risk, readily obtainable and easily repeatable technique for screening for early disease detection and diagnosis, patient stratification at diagnosis into different risk categories, identification of patients’ risk for metastatic disease and subsequent relapse, and real-time monitoring of patient response to specific treatment. Thus, effective liquid-based biomarkers will potentially shift the treatment paradigm of prostate cancer towards more personalised and targeted medicine.
Collapse
|
10
|
Biochemistry of mammalian ferritins in the regulation of cellular iron homeostasis and oxidative responses. SCIENCE CHINA. LIFE SCIENCES 2020; 64:352-362. [PMID: 32974854 DOI: 10.1007/s11427-020-1795-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/11/2020] [Indexed: 02/08/2023]
Abstract
Ferritin, an iron-storage protein, regulates cellular iron metabolism and oxidative stress. The ferritin structure is characterized as a spherical cage, inside which large amounts of iron are deposited in a safe, compact and bioavailable form. All ferritins readily catalyze Fe(II) oxidation by peroxides at the ferroxidase center to prevent free Fe(II) from participating in oxygen free radical formation via Fenton chemistry. Thus, ferritin is generally recognized as a cytoprotective stratagem against intracellular oxidative damage The expression of cytosolic ferritins is usually regulated by iron status and oxidative stress at both the transcriptional and post-transcriptional levels. The mechanism of ferritin-mediated iron recycling is far from clarified, though nuclear receptor co-activator 4 (NCOA4) was recently identified as a cargo receptor for ferritin-based lysosomal degradation. Cytosolic ferritins are heteropolymers assembled by H- and L-chains in different proportions. The mitochondrial ferritins are homopolymers and distributed in restricted tissues. They play protective roles in mitochondria where heme- and Fe/S-enzymes are synthesized and high levels of ROS are produced. Genetic ferritin disorders are mainly related to the L-chain mutations, which generally cause severe movement diseases. This review is focused on the biochemistry and function of mammalian intracellular ferritin as the major iron-storage and anti-oxidation protein.
Collapse
|
11
|
Grounds MD, Terrill JR, Al-Mshhdani BA, Duong MN, Radley-Crabb HG, Arthur PG. Biomarkers for Duchenne muscular dystrophy: myonecrosis, inflammation and oxidative stress. Dis Model Mech 2020; 13:13/2/dmm043638. [PMID: 32224496 PMCID: PMC7063669 DOI: 10.1242/dmm.043638] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked disease that causes severe loss of muscle mass and function in young children. Promising therapies for DMD are being developed, but the long lead times required when using clinical outcome measures are hindering progress. This progress would be facilitated by robust molecular biomarkers in biofluids, such as blood and urine, which could be used to monitor disease progression and severity, as well as to determine optimal drug dosing before a full clinical trial. Many candidate DMD biomarkers have been identified, but there have been few follow-up studies to validate them. This Review describes the promising biomarkers for dystrophic muscle that have been identified in muscle, mainly using animal models. We strongly focus on myonecrosis and the associated inflammation and oxidative stress in DMD muscle, as the lack of dystrophin causes repeated bouts of myonecrosis, which are the key events that initiate the resultant severe dystropathology. We discuss the early events of intrinsic myonecrosis, along with early regeneration in the context of histological and other measures that are used to quantify its incidence. Molecular biomarkers linked to the closely associated events of inflammation and oxidative damage are discussed, with a focus on research related to protein thiol oxidation and to neutrophils. We summarise data linked to myonecrosis in muscle, blood and urine of dystrophic animal species, and discuss the challenge of translating such biomarkers to the clinic for DMD patients, especially to enhance the success of clinical trials. Summary: This Review discusses biomarkers in blood and urine linked to myonecrosis, inflammation and oxidative stress, to enhance development of therapies for DMD, and the challenges to be overcome for clinical translation.
Collapse
Affiliation(s)
- Miranda D Grounds
- School of Human Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Jessica R Terrill
- School of Molecular Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Basma A Al-Mshhdani
- School of Molecular Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Marisa N Duong
- School of Molecular Sciences, the University of Western Australia, Perth, WA 6009, Australia
| | - Hannah G Radley-Crabb
- School of Pharmacy and Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
| | - Peter G Arthur
- School of Molecular Sciences, the University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
12
|
Thangarajh M, Zhang A, Gill K, Ressom HW, Li Z, Varghese RS, Hoffman EP, Nagaraju K, Hathout Y, Boca SM. Discovery of potential urine-accessible metabolite biomarkers associated with muscle disease and corticosteroid response in the mdx mouse model for Duchenne. PLoS One 2019; 14:e0219507. [PMID: 31310630 PMCID: PMC6634414 DOI: 10.1371/journal.pone.0219507] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Urine is increasingly being considered as a source of biomarker development in Duchenne Muscular Dystrophy (DMD), a severe, life-limiting disorder that affects approximately 1 in 4500 boys. In this study, we considered the mdx mice-a murine model of DMD-to discover biomarkers of disease, as well as pharmacodynamic biomarkers responsive to prednisolone, a corticosteroid commonly used to treat DMD. Longitudinal urine samples were analyzed from male age-matched mdx and wild-type mice randomized to prednisolone or vehicle control via liquid chromatography tandem mass spectrometry. A large number of metabolites (869 out of 6,334) were found to be significantly different between mdx and wild-type mice at baseline (Bonferroni-adjusted p-value < 0.05), thus being associated with disease status. These included a metabolite with m/z = 357 and creatine, which were also reported in a previous human study looking at serum. Novel observations in this study included peaks identified as biliverdin and hypusine. These four metabolites were significantly higher at baseline in the urine of mdx mice compared to wild-type, and significantly changed their levels over time after baseline. Creatine and biliverdin levels were also different between treated and control groups, but for creatine this may have been driven by an imbalance at baseline. In conclusion, our study reports a number of biomarkers, both known and novel, which may be related to either the mechanisms of muscle injury in DMD or prednisolone treatment.
Collapse
Affiliation(s)
- Mathula Thangarajh
- Department of Neurology, George Washington University School of Medicine and Children’s National Health Systems, Washington, D.C., United States of America
| | - Aiping Zhang
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Children’s National Health Systems, Washington, D.C., United States of America
| | - Kirandeep Gill
- Department of Oncology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Habtom W. Ressom
- Department of Oncology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Zhenzhi Li
- Department of Oncology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Rency S. Varghese
- Department of Oncology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Eric P. Hoffman
- School of Pharmacy & Pharmaceutical Sciences, Binghamton University, Binghamton, N.Y., United States of America
| | - Kanneboyina Nagaraju
- School of Pharmacy & Pharmaceutical Sciences, Binghamton University, Binghamton, N.Y., United States of America
| | - Yetrib Hathout
- School of Pharmacy & Pharmaceutical Sciences, Binghamton University, Binghamton, N.Y., United States of America
| | - Simina M. Boca
- Department of Oncology, Georgetown University Medical Center, Washington, D.C., United States of America
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, D.C., United States of America
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University Medical Center, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
13
|
Murphy S, Zweyer M, Henry M, Meleady P, Mundegar RR, Swandulla D, Ohlendieck K. Proteomic profiling of liver tissue from the mdx- 4cv mouse model of Duchenne muscular dystrophy. Clin Proteomics 2018; 15:34. [PMID: 30386187 PMCID: PMC6205794 DOI: 10.1186/s12014-018-9212-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/23/2018] [Indexed: 12/30/2022] Open
Abstract
Background Duchenne muscular dystrophy is a highly complex multi-system disease caused by primary abnormalities in the membrane cytoskeletal protein dystrophin. Besides progressive skeletal muscle degeneration, this neuromuscular disorder is also associated with pathophysiological perturbations in many other organs including the liver. To determine potential proteome-wide alterations in liver tissue, we have used a comparative and mass spectrometry-based approach to study the dystrophic mdx-4cv mouse model of dystrophinopathy. Methods The comparative proteomic profiling of mdx-4cv versus wild type liver extracts was carried out with an Orbitrap Fusion Tribrid mass spectrometer. The distribution of identified liver proteins within protein families and potential protein interaction patterns were analysed by systems bioinformatics. Key findings on fatty acid binding proteins were confirmed by immunoblot analysis and immunofluorescence microscopy. Results The proteomic analysis revealed changes in a variety of protein families, affecting especially fatty acid, carbohydrate and amino acid metabolism, biotransformation, the cellular stress response and ion handling in the mdx-4cv liver. Drastically increased protein species were identified as fatty acid binding protein FABP5, ferritin and calumenin. Decreased liver proteins included phosphoglycerate kinase, apolipoprotein and perilipin. The drastic change in FABP5 was independently verified by immunoblotting and immunofluorescence microscopy. Conclusions The proteomic results presented here indicate that the intricate and multifaceted pathogenesis of the mdx-4cv model of dystrophinopathy is associated with secondary alterations in the liver affecting especially fatty acid transportation. Since FABP5 levels were also shown to be elevated in serum from dystrophic mice, this protein might be a useful indicator for monitoring liver changes in X-linked muscular dystrophy.
Collapse
Affiliation(s)
- Sandra Murphy
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Margit Zweyer
- 2Institute of Physiology II, University of Bonn, 53115 Bonn, Germany
| | - Michael Henry
- 3National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Paula Meleady
- 3National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Rustam R Mundegar
- 2Institute of Physiology II, University of Bonn, 53115 Bonn, Germany
| | - Dieter Swandulla
- 2Institute of Physiology II, University of Bonn, 53115 Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland
| |
Collapse
|