1
|
Hesse F, Low J, Cao J, Bulat F, Kreis F, Wright AJ, Brindle KM. Deuterium MRI of serine metabolism in mouse models of glioblastoma. Magn Reson Med 2024; 92:1811-1821. [PMID: 38946234 DOI: 10.1002/mrm.30198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/10/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024]
Abstract
PURPOSE Serine is a major source of one-carbon units needed for the synthesis of nucleotides and the production of intramitochondrial nicotinamide adenine dinucleotide phosphate (NADPH), and it plays an important role in cancer cell proliferation. The aim of this study was to develop a deuterium (2H) MRS imaging method for imaging tumor serine metabolism. METHODS Sequential (2H) spectra and spectroscopic images were used to monitor the metabolism of [2,3,3-2H3]serine in patient-derived glioblastoma cells in vitro and in tumors obtained by their orthotopic implantation in mouse brain. RESULTS [14,14-2H2] 5,10-methylene-tetrahydrofolate, [2H]glycine, [2H]formate, and labeled water were detected in cell suspensions and water labeling in spectroscopic images of tumors. Studies in cells and tumors with variable mitochondrial content and inhibitor studies in cells demonstrated that most of the labeled serine was metabolized in the mitochondria. Water labeling in the cell suspensions was correlated with formate labeling; therefore, water labeling observed in tumors could be used to provide a surrogate measure of flux in the pathway of one-carbon metabolism in vivo. CONCLUSION The method has the potential to be used clinically to select patients for treatment with inhibitors of one-carbon metabolism and subsequently to detect their early responses to such treatment.
Collapse
Affiliation(s)
- Friederike Hesse
- Cancer Research UK Cambridge Institute, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Jacob Low
- Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Jianbo Cao
- Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Flaviu Bulat
- Cancer Research UK Cambridge Institute, Cambridge, UK
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Felix Kreis
- Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Alan J Wright
- Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
2
|
Khan AS, McLean MA, Kaggie JD, Horvat-Menih I, Matys T, Schulte RF, Locke MJ, Grimmer A, Wodtke P, Latimer E, Frary A, Graves MJ, Gallagher FA. Measuring cerebral enzymatic activity, brain pH and extracranial muscle metabolism with hyperpolarized 13C-pyruvate. NMR IN BIOMEDICINE 2024:e5271. [PMID: 39367692 DOI: 10.1002/nbm.5271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/06/2024] [Accepted: 09/20/2024] [Indexed: 10/06/2024]
Abstract
Hyperpolarized carbon-13 (13C) magnetic resonance imaging (MRI) has shown promise for non-invasive assessment of the cerebral metabolism of [1-13C]pyruvate in both healthy volunteers and patients. The exchange of pyruvate to lactate catalysed by lactate dehydrogenase (LDH) and that of pyruvate flux to bicarbonate through pyruvate dehydrogenase (PDH) are the most widely studied reactions in vivo. Here we show the potential of the technique to probe additional enzymatic activity within the brain. Approximately 50 s after intravenous injection of hyperpolarized pyruvate, high-flip-angle pulses were used to detect cerebral 13C-labelled carbon dioxide (13CO2), in addition to the 13C-bicarbonate (H13CO3 -) subsequently formed by carbonic anhydrase (CA). Brain pH measurements, which were weighted towards the extracellular compartment, were calculated from the ratio of H13CO3 - to 13CO2 in seven volunteers using the Henderson-Hasselbalch equation, demonstrating an average pH ± SD of 7.40 ± 0.02, with inter-observer reproducibility of 0.04. In addition, hyperpolarized [1-13C]aspartate was also detected, demonstrating irreversible pyruvate carboxylation to oxaloacetate by pyruvate carboxylase (PC) and subsequent transamination by aspartate aminotransferase (AST), with the average flux being on average 11% ± 3% of that through PDH. A hyperpolarized [1-13C]alanine signal was also detected, but this was localized to extracranial muscle tissue in keeping with skeletal alanine aminotransferase (ALT) activity. The results demonstrate the potential of hyperpolarized 13C-MRI to assess cerebral and extracerebral [1-13C]pyruvate metabolism in addition to LDH and PDH activity. Non-invasive measurements of brain pH could be particularly important in assessing cerebral pathology given the wide range of disease processes that alter acid-base balance.
Collapse
Affiliation(s)
- Alixander S Khan
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Mary A McLean
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joshua D Kaggie
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Ines Horvat-Menih
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Tomasz Matys
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | | | - Matthew J Locke
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Ashley Grimmer
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Pascal Wodtke
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Elizabeth Latimer
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Amy Frary
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Martin J Graves
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Ferdia A Gallagher
- Department of Radiology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Liu X, Bao Q, Liu Z, Wang J, Otikovs M, Zhang Z, Cheng X, Wang J, Frydman L, Zhou X, Liu M, Liu C. Exploring Metabolic Aberrations after Intracerebral Hemorrhage In Vivo with Deuterium Metabolic Spectroscopy Imaging. Anal Chem 2024; 96:15563-15571. [PMID: 39295127 DOI: 10.1021/acs.analchem.4c01999] [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: 09/21/2024]
Abstract
Aberrations in metabolism after intracerebral hemorrhage (ICH), particularly lactate metabolism, play a crucial role in the pathophysiology and patient outcome. To date, the evaluation of metabolism relies heavily on invasive methods such as microdialysis, restricting a comprehensive understanding of the metabolic mechanisms associated with ICH. This study proposes a noninvasive metabolic imaging method based on 2H magnetic resonance spectroscopy and imaging (2H-MRS/MRSI) to detect metabolic changes after ICH in vivo. To overcome the low-sensitivity limitation of 2H, we designed a new 1H-2H double-resonance coil with 2H-channel active detuning and proposed chemical shift imaging based on the balanced steady-state free precession method (CSI-bSSFP). Compared with the volume coil, the signal-to-noise ratio (SNR) of the new coil was increased by 4.5 times. In addition, the SNR of CSI-bSSFP was 1.5 times higher than that of conventional CSI. These two technologies were applied to measure lactate metabolic flux at different phases of ICH. The results show a higher lactate concentration in ICH rats than in control rats, which is in line with the increased expression of lactate dehydrogenase measured via immunohistochemistry staining (AUCLac_area/Glc_area: control, 0.08 ± 0.02 vs ICH-3d, 0.39 ± 0.05 vs ICH-7d, 0.18 ± 0.02, P < 0.01; H-score: control, 126.4 ± 5.03 vs ICH-3d, 168.4 ± 5.71 vs ICH-7d,133.6 ± 7.70, P < 0.05). A higher lactate signal also appeared near the ICH region than in normal brain tissue. In conclusion, 2H-MRS/MRSI shows potential as a useful method for in vivo metabolic imaging and noninvasive assessment of ICH.
Collapse
Affiliation(s)
- Xinjie Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
| | - Qingjia Bao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
| | - Zhuang Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
| | - Martins Otikovs
- Department of Chemical and Biological Physics, Weizmann Institute of Science, 234 Herzl Street, Rehovot 76100, Israel
| | - Zhi Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
| | - Xin Cheng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
| | - Jiazheng Wang
- Clinical & Technical Support, Philips Healthcare, Beijing 100600, China
| | - Lucio Frydman
- Department of Chemical and Biological Physics, Weizmann Institute of Science, 234 Herzl Street, Rehovot 76100, Israel
| | - Xin Zhou
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
- Optics Valley Laboratory, Hubei 430074, China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
- Optics Valley Laboratory, Hubei 430074, China
| | - Chaoyang Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100864, China
- Optics Valley Laboratory, Hubei 430074, China
| |
Collapse
|
4
|
Karkouri J, Rodgers CT. Sequence building block for magnetic resonance spectroscopy on Siemens VE-series scanners. NMR IN BIOMEDICINE 2024; 37:e5165. [PMID: 38807311 DOI: 10.1002/nbm.5165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 05/30/2024]
Abstract
We present a sequence building block (SBB) that embeds magnetic resonance spectroscopy (MRS) into another sequence on the Siemens VE platform without any custom hardware. This enables dynamic studies such as functional MRS (fMRS), dynamic shimming and frequency correction, and acquisition of navigator images for motion correction. The SBB supports nonlocalised spectroscopy (free induction decay), STimulated Echo Acquisition Mode single voxel spectroscopy, and 1D, 2D and 3D phase-encoded chemical shift imaging. It can embed 1H or X-nuclear MRS into a 1H sequence; and 1H-MRS into an X-nuclear sequence. We demonstrate integration into the vendor's gradient-recalled echo sequence. We acquire test data in phantoms with three coils (31P/1H, 13C/1H and 2H/1H) and in two volunteers on a 7-T Terra MRI scanner. Fifteen lines of code are required to insert the SBB into a sequence. Spectra and images are acquired successfully in all cases in phantoms, and in human abdomen and calf muscle. Phantom comparison of signal-to-noise ratio and linewidth showed that the SBB has negligible effects on image and spectral quality, except that it sometimes produces a nuclear Overhauser effect (NOE) signal enhancement for multinuclear applications in line with conventional 1H NOE pulses. Our new SBB embeds MRS into a host imaging or spectroscopy sequence in 15 lines of code. It allows homonuclear and heteronuclear interleaving. The package is available through the standard C2P procedure. We hope this will lower the barrier for entry to studies applying dynamic fMRS and for online motion correction and B0-shim updating.
Collapse
Affiliation(s)
- Jabrane Karkouri
- Wolfson Brain Imaging Center, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
5
|
van den Wildenberg L, Runderkamp BA, Seelen LWF, van Laarhoven HWM, Gosselink MWJM, van der Kemp WJM, Haj Mohammad N, Klomp DWJ, Prompers JJ. Measurement of metabolite levels and treatment-induced changes in hepatic metastases of gastro-esophageal cancer using 7-T phosphorus magnetic resonance spectroscopic imaging. NMR IN BIOMEDICINE 2024; 37:e5155. [PMID: 38616046 DOI: 10.1002/nbm.5155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/16/2024]
Abstract
Methods for early treatment response evaluation to systemic therapy of liver metastases are lacking. Tumor tissue often exhibits an increased ratio of phosphomonoesters to phosphodiesters (PME/PDE), which can be noninvasively measured by phosphorus magnetic resonance spectroscopy (31P MRS), and may be a marker for early therapy response assessment in liver metastases. However, with commonly used 31P surface coils for liver 31P MRS, the liver is not fully covered, and metastases may be missed. The objective of this study was to demonstrate the feasibility of 31P MRS imaging (31P MRSI) with full liver coverage to assess 31P metabolite levels and chemotherapy-induced changes in liver metastases of gastro-esophageal cancer, using a 31P whole-body birdcage transmit coil in combination with a 31P body receive array at 7 T. 3D 31P MRSI data were acquired in two patients with hepatic metastases of esophageal cancer, before the start of chemotherapy and after 2 (and 9 in patient 2) weeks of chemotherapy. 3D 31P MRSI acquisitions were performed using an integrated 31P whole-body transmit coil in combination with a 16-channel body receive array at 7 T, with a field of view covering the full abdomen and a nominal voxel size of 20-mm isotropic. From the 31P MRSI data, 12 31P metabolite signals were quantified. Prior to chemotherapy initiation, both PMEs, that is, phosphocholine (PC) and phosphoethanolamine (PE), were significantly higher in all metastases compared with the levels previously determined in the liver of healthy volunteers. After 2 weeks of chemotherapy, PC and PE levels remained high or even increased further, resulting in increased PME/PDE ratios compared with healthy liver tissue, in correspondence with the clinical assessment of progressive disease after 2 months of chemotherapy. The suggested approach may present a viable tool for early therapy (non)response assessment of tumor metabolism in patients with liver metastases.
Collapse
Affiliation(s)
| | - Bobby A Runderkamp
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Leonard W F Seelen
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Surgery, UMC Utrecht Cancer Center, Utrecht, The Netherlands
- Sint Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark W J M Gosselink
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wybe J M van der Kemp
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nadia Haj Mohammad
- Department of Medical Oncology, Utrecht Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis W J Klomp
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeanine J Prompers
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
6
|
Ahmadian N, Konig MM, Otto S, Tesselaar K, van Eijsden P, Gosselink M, Gursan A, Klomp DW, Prompers JJ, Wiegers EC. Human Brain Deuterium Metabolic Imaging at 7 T: Impact of Different [6,6'- 2H 2]Glucose Doses. J Magn Reson Imaging 2024. [PMID: 39058248 DOI: 10.1002/jmri.29532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Deuterium metabolic imaging (DMI) is an innovative, noninvasive metabolic MR imaging method conducted after administration of 2H-labeled substrates. DMI after [6,6'-2H2]glucose consumption has been used to investigate brain metabolic processes, but the impact of different [6,6'-2H2]glucose doses on DMI brain data is not well known. PURPOSE To investigate three different [6,6'-2H2]glucose doses for DMI in the human brain at 7 T. STUDY TYPE Prospective. POPULATION Six healthy participants (age: 28 ± 8 years, male/female: 3/3). FIELD STRENGTH/SEQUENCE 7 T, 3D 2H free-induction-decay (FID)-magnetic resonance spectroscopic imaging (MRSI) sequence. ASSESSMENT Three subjects received two different doses (0.25 g/kg, 0.50 g/kg or 0.75 g/kg body weight) of [6,6'-2H2]glucose on two occasions and underwent consecutive 2H-MRSI scans for 120 minutes. Blood was sampled every 10 minutes during the scan, to determine plasma glucose levels and plasma 2H-Glucose atom percent excess (APE) (part-1). Three subjects underwent the same protocol once after receiving 0.50 g/kg [6,6'-2H2]glucose (part-2). STATISTICAL TEST Mean plasma 2H-Glucose APE and glucose plasma concentrations were compared using one-way ANOVA. Brain 2H-Glc and brain 2H-Glx (part-1) were analyzed with a two-level Linear Mixed Model. In part-2, a General Linear Model was used to compare brain metabolite signals. Statistical significance was set at P < 0.05. RESULTS Between 60 and 100 minutes after ingesting [6,6'-2H2]glucose, plasma 2H-Glc APE did not differ between 0.50 g/kg and 0.75 g/kg doses (P = 0.961), but was significantly lower for 0.25 g/kg. Time and doses significantly affected brain 2H-Glucose levels (estimate ± standard error [SE]: 0.89 ± 0.01, 1.09 ± 0.01, and 1.27 ± 0.01, for 0.25 g/kg, 0.50 g/kg, and 0.75 g/kg, respectively) and brain 2H-Glutamate/Glutamine levels (estimate ± SE: 1.91 ± 0.03, 2.27 ± 0.03, and 2.46 ± 0.03, for 0.25 g/kg, 0.50 g/kg, and 0.75 g/kg, respectively). Plasma 2H-Glc APE, brain 2H-Glc, and brain 2H-Glx levels were comparable among subjects receiving 0.50 g/kg [6,6'-2H2]glucose. DATA CONCLUSION Brain 2H-Glucose and brain 2H-Glutamate/Glutamine showed to be [6,6'-2H2]glucose dose dependent. A dose of 0.50 g/kg demonstrated comparable, and well-detectable, 2H-Glucose and 2H-Glutamate/Glutamine signals in the brain. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Narjes Ahmadian
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maaike M Konig
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sigrid Otto
- CTI Lab Support, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kiki Tesselaar
- CTI Lab Support, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pieter van Eijsden
- Neurosurgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mark Gosselink
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ayhan Gursan
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis W Klomp
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeanine J Prompers
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
- Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Evita C Wiegers
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
7
|
Khan AS, Peterson KA, Vittay OI, McLean MA, Kaggie JD, O’Brien JT, Rowe JB, Gallagher FA, Matys T, Wolfe S. Deuterium Metabolic Imaging of Alzheimer Disease at 3-T Magnetic Field Strength: A Pilot Case-Control Study. Radiology 2024; 312:e232407. [PMID: 39012255 PMCID: PMC11294762 DOI: 10.1148/radiol.232407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 07/17/2024]
Abstract
Background Impaired glucose metabolism is characteristic of several types of dementia, preceding cognitive symptoms and structural brain changes. Reduced glucose uptake in specific brain regions, detected using fluorine 18 (18F) fluorodeoxyglucose (FDG) PET, is a valuable diagnostic marker in Alzheimer disease (AD). However, the use of 18F-FDG PET in clinical practice may be limited by equipment availability and high cost. Purpose To test the feasibility of using MRI-based deuterium (2H) metabolic imaging (DMI) at a clinical magnetic field strength (3 T) to detect and localize changes in the concentration of glucose and its metabolites in the brains of patients with a clinical diagnosis of AD. Materials and Methods Participants were recruited for this prospective case-control pilot study between March 2021 and February 2023. DMI was performed at 3 T using a custom birdcage head coil following oral administration of deuterium-labeled glucose (0.75 g/kg). Unlocalized whole-brain MR spectroscopy (MRS) and three-dimensional MR spectroscopic imaging (MRSI) (voxel size, 3.2 cm cubic) were performed. Ratios of 2H-glucose, 2H-glutamate and 2H-glutamine (2H-Glx), and 2H-lactate spectroscopic peak signals to 2H-water peak signal were calculated for the whole-brain MR spectra and for individual MRSI voxels. Results A total of 19 participants, including 10 participants with AD (mean age, 68 years ± 5 [SD]; eight males) and nine cognitively healthy control participants (mean age, 70 years ± 6; six males) were evaluated. Whole-brain spectra demonstrated a reduced ratio of 2H-Glx to 2H-glucose peak signals in participants with AD compared with control participants (0.41 ± 0.09 vs 0.58 ± 0.20, respectively; P = .04), suggesting an impairment of oxidative glucose metabolism in AD. However, there was no evidence of localization of these changes to the expected regions of metabolic impairment at MRSI, presumably due to insufficient spatial resolution. Conclusion DMI at 3 T demonstrated impairment of oxidative glucose metabolism in the brains of patients with AD but no evidence of regional signal differences. © RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Alixander S. Khan
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Katie A. Peterson
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Orsolya I. Vittay
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Mary A. McLean
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Joshua D. Kaggie
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - John T. O’Brien
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - James B. Rowe
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Ferdia A. Gallagher
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Tomasz Matys
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| | - Shannyn Wolfe
- From the Departments of Radiology (A.S.K., K.A.P., M.A.M., J.D.K.,
F.A.G., T.M.), Psychiatry (J.T.O.), and Clinical Neurosciences (J.B.R.),
University of Cambridge, Hills Road, Cambridge CB2 0QQ, England; and
Departments of Radiology (O.I.V., F.A.G., T.M.) and Neurology (J.B.R.),
Cambridge University Hospitals NHS Foundation Trust, Cambridge, England
| |
Collapse
|
8
|
Bozymski B, Emir U, Dydak U, Shen X, Thomas MA, Özen A, Chiew M, Clarke W, Sawiak S. 3D ultra-short echo time 31P-MRSI with rosette k-space pattern: Feasibility and comparison with conventional weighted CSI. RESEARCH SQUARE 2024:rs.3.rs-4223790. [PMID: 38659806 PMCID: PMC11042414 DOI: 10.21203/rs.3.rs-4223790/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Phosphorus-31 magnetic resonance spectroscopic imaging (31P-MRSI) provides valuable non-invasive in vivo information on tissue metabolism but is burdened by poor sensitivity and prolonged scan duration. Ultra-short echo time (UTE) acquisitions minimize signal loss when probing signals with relatively short spin-spin relaxation time (T2), while also preventing first-order dephasing. Here, a three-dimensional (3D) UTE sequence with a rosette k-space trajectory is applied to 31P-MRSI at 3T. Conventional chemical shift imaging (CSI) employs highly regular Cartesian k-space sampling, susceptible to substantial artifacts when accelerated via undersampling. In contrast, this novel sequence's "petal-like" pattern offers incoherent sampling more suitable for compressed sensing (CS). These results showcase the competitive performance of UTE rosette 31P-MRSI against conventional weighted CSI with simulation, phantom, and in vivo leg muscle comparisons.
Collapse
Affiliation(s)
| | - Uzay Emir
- School of Health Sciences, Purdue University
| | | | - Xin Shen
- Radiology and Biomedical Imaging, University of California San Francisco
| | | | - Ali Özen
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg
| | - Mark Chiew
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences
| | - William Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences
| | - Stephen Sawiak
- Department of Clinical Neuroscience, University of Cambridge
| |
Collapse
|
9
|
Bøgh N, Vaeggemose M, Schulte RF, Hansen ESS, Laustsen C. Repeatability of deuterium metabolic imaging of healthy volunteers at 3 T. Eur Radiol Exp 2024; 8:44. [PMID: 38472611 PMCID: PMC10933246 DOI: 10.1186/s41747-024-00426-4] [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: 10/26/2023] [Accepted: 01/02/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Magnetic resonance (MR) imaging of deuterated glucose, termed deuterium metabolic imaging (DMI), is emerging as a biomarker of pathway-specific glucose metabolism in tumors. DMI is being studied as a useful marker of treatment response in a scan-rescan scenario. This study aims to evaluate the repeatability of brain DMI. METHODS A repeatability study was performed in healthy volunteers from December 2022 to March 2023. The participants consumed 75 g of [6,6'-2H2]glucose. The delivery of 2H-glucose to the brain and its conversion to 2H-glutamine + glutamate, 2H-lactate, and 2H-water DMI was imaged at baseline and at 30, 70, and 120 min. DMI was performed using MR spectroscopic imaging on a 3-T system equipped with a 1H/2H-tuned head coil. Coefficients of variation (CoV) were computed for estimation of repeatability and between-subject variability. In a set of exploratory analyses, the variability effects of region, processing, and normalization were estimated. RESULTS Six male participants were recruited, aged 34 ± 6.5 years (mean ± standard deviation). There was 42 ± 2.7 days between sessions. Whole-brain levels of glutamine + glutamate, lactate, and glucose increased to 3.22 ± 0.4 mM, 1.55 ± 0.3 mM, and 3 ± 0.7 mM, respectively. The best signal-to-noise ratio and repeatability was obtained at the 120-min timepoint. Here, the within-subject whole-brain CoVs were -10% for all metabolites, while the between-subject CoVs were -20%. CONCLUSIONS DMI of glucose and its downstream metabolites is feasible and repeatable on a clinical 3 T system. TRIAL REGISTRATION ClinicalTrials.gov, NCT05402566 , registered the 25th of May 2022. RELEVANCE STATEMENT Brain deuterium metabolic imaging of healthy volunteers is repeatable and feasible at clinical field strengths, enabling the study of shifts in tumor metabolism associated with treatment response. KEY POINTS • Deuterium metabolic imaging is an emerging tumor biomarker with unknown repeatability. • The repeatability of deuterium metabolic imaging is on par with FDG-PET. • The study of deuterium metabolic imaging in clinical populations is feasible.
Collapse
Affiliation(s)
- Nikolaj Bøgh
- The MR Research Centre, Dept. Of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark.
- A&E, Gødstrup Hospital, Herning, Denmark.
| | - Michael Vaeggemose
- The MR Research Centre, Dept. Of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark
- GE HealthCare, Brondby, Denmark
| | | | - Esben S S Hansen
- The MR Research Centre, Dept. Of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark
| | - Christoffer Laustsen
- The MR Research Centre, Dept. Of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark
| |
Collapse
|
10
|
Craven AR, Bell TK, Ersland L, Harris AD, Hugdahl K, Oeltzschner G. Linewidth-related bias in modelled concentration estimates from GABA-edited 1H-MRS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582249. [PMID: 38464094 PMCID: PMC10925149 DOI: 10.1101/2024.02.27.582249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
J-difference-edited MRS is widely used to study GABA in the human brain. Editing for low-concentration target molecules (such as GABA) typically exhibits lower signal-to-noise ratio (SNR) than conventional non-edited MRS, varying with acquisition region, volume and duration. Moreover, spectral lineshape may be influenced by age-, pathology-, or brain-region-specific effects of metabolite T2, or by task-related blood-oxygen level dependent (BOLD) changes in functional MRS contexts. Differences in both SNR and lineshape may have systematic effects on concentration estimates derived from spectral modelling. The present study characterises the impact of lineshape and SNR on GABA+ estimates from different modelling algorithms: FSL-MRS, Gannet, LCModel, Osprey, spant and Tarquin. Publicly available multi-site GABA-edited data (222 healthy subjects from 20 sites; conventional MEGA-PRESS editing; TE = 68 ms) were pre-processed with a standardised pipeline, then filtered to apply controlled levels of Lorentzian and Gaussian linebroadening and SNR reduction. Increased Lorentzian linewidth was associated with a 2-5% decrease in GABA+ estimates per Hz, observed consistently (albeit to varying degrees) across datasets and most algorithms. Weaker, often opposing effects were observed for Gaussian linebroadening. Variations are likely caused by differing baseline parametrization and lineshape constraints between models. Effects of linewidth on other metabolites (e.g., Glx and tCr) varied, suggesting that a linewidth confound may persist after scaling to an internal reference. These findings indicate a potentially significant confound for studies where linewidth may differ systematically between groups or experimental conditions, e.g. due to T2 differences between brain regions, age, or pathology, or varying T2* due to BOLD-related changes. We conclude that linewidth effects need to be rigorously considered during experimental design and data processing, for example by incorporating linewidth into statistical analysis of modelling outcomes or development of appropriate lineshape matching algorithms.
Collapse
Affiliation(s)
- Alexander R. Craven
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
- Department of Clinical Engineering, Haukeland University Hospital, Bergen, Norway
| | - Tiffany K. Bell
- Department of Radiology, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Lars Ersland
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
- Department of Clinical Engineering, Haukeland University Hospital, Bergen, Norway
| | - Ashley D. Harris
- Department of Radiology, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Kenneth Hugdahl
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Carrell T, McDougall MP. Multi-channel magnetic resonance spectroscopy graphical user interface (McMRSGUI). PLoS One 2024; 19:e0299142. [PMID: 38416774 PMCID: PMC10901321 DOI: 10.1371/journal.pone.0299142] [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: 08/29/2023] [Accepted: 02/06/2024] [Indexed: 03/01/2024] Open
Abstract
This work introduces an open-sourced graphical user interface (GUI) software enabling the combination of multi-channel magnetic resonance spectroscopy data with different literature-based methods for the improvement of the quality and reliability of combined spectra. The multi-channel magnetic resonance spectroscopy graphical user interface (McMRSGUI) is a MATLAB-based spectroscopy processing GUI equipped to load multi-channel MRS data, pre-process, combine, and export combined data for evaluation with open-source quantification software (jMRUI). A literature-based, decision-tree process was incorporated into the combination type selection to serve as a guide to minimize spectral distortion in selecting between weighting methods. Multi-channel, simulated spectra were combined with the different combination techniques and evaluated for spectral distortion to validate the code. The incorporation of the combination methods into a single processing software enables multi-channel magnetic resonance spectroscopy (MRS) data to be combined and compared for improved spectral quality with little user knowledge of combination techniques. Through the spectral peak distortion simulation of the combination methods, combined signal-to-noise ratio (SNR) values from the literature were verified. The spectral peak distortion simulation provides a secondary tool for researchers to estimate the spectral SNR levels when spectral distortion could occur and use this knowledge to further guide the selection of their combination technique. The McMRSGUI provides a software toolkit for evaluating multi-channel MRS data and their combination. Simulations evaluating spectral distortion at different noise levels were performed for each combination method to validate the GUI and demonstrate a method for researchers to assess the combined SNR levels at which they could be introducing spectral distortion.
Collapse
Affiliation(s)
- Travis Carrell
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Mary P McDougall
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
12
|
Tyler A, Hundertmark MJ, Miller JJ, Rider O, Tyler DJ, Valkovič L. Compartment-based reconstruction of acquisition-weighted 31P cardiac MRSI reduces sensitivity to cardiac motion and scan planning. Front Physiol 2024; 14:1325458. [PMID: 38314138 PMCID: PMC10834798 DOI: 10.3389/fphys.2023.1325458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/21/2023] [Indexed: 02/06/2024] Open
Abstract
Motivation: 31P magnetic resonance spectroscopic imaging (31P MRSI) is a powerful technique for investigating the metabolic effects of treatments for heart failure in vivo, allowing a better understanding of their mechanism of action in patient cohorts. Unfortunately, cardiac 31P MRSI is fundamentally limited by low SNR, which leads to compromises in acquisition, such as no cardiac or respiratory gating or low spatial resolution, in order to achieve reasonable scan times. Spectroscopy with linear algebra modeling (SLAM) reconstruction may be able to address these challenges and therefore improve repeatability by incorporating a segmented localizer into the reconstruction. Methods: Six healthy volunteers were scanned twice in a test-retest procedure to allow quantification of repeatability. Each scan consisted of anatomical localizers and two acquisition-weighted (AW) 31P MRSI acquisitions, which were acquired with and without cardiac gating. Five patients with heart failure with a preserved ejection fraction were then scanned with the same 31P MRSI sequence without cardiac gating. All 31P MRSI datasets were reconstructed with both conventional Fourier transform (FT)-based reconstruction and SLAM reconstruction, which were compared statistically. The effect of shifting the 31P MRSI acquisition field of view was also investigated. Results: In the healthy volunteer cohort, the spectral fit of the SLAM reconstructions had significantly improved Cramer-Rao lower bounds (CRLBs) compared to the FT-based reconstruction of non-cardiac gated data, as well as improved coefficients of variability and repeatability. The SLAM reconstruction found a significant difference in the PCr/ATP ratio between the healthy volunteer and patient cohorts, which the FT-based reconstruction did not find. Furthermore, the SLAM reconstruction was less influenced by the placement of the field of view (FOV) of the 31P MRSI acquisition in post hoc analysis. Discussion: The experimental benefits of the SLAM reconstruction for AW data were demonstrated by the improvements in fit confidence and repeatability seen in the healthy volunteer cohort and post hoc FOV analysis. The benefit of SLAM reconstruction of AW data for clinical studies was then illustrated by the patient cohort, which suggested improved sensitivity to clinically significant changes in the PCr/ATP ratio.
Collapse
Affiliation(s)
- Andrew Tyler
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Physiology, University of Oxford, Oxford, United Kingdom
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, United Kingdom
| | - Moritz J. Hundertmark
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Jack J. Miller
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Physiology, University of Oxford, Oxford, United Kingdom
- The MR and PET Research Centres, Aarhus University, Aarhus, Denmark
| | - Oliver Rider
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Damian J. Tyler
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Physiology, University of Oxford, Oxford, United Kingdom
| | - Ladislav Valkovič
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
13
|
Wodtke P, Grashei M, Schilling F. Quo Vadis Hyperpolarized 13C MRI? Z Med Phys 2023:S0939-3889(23)00120-4. [PMID: 38160135 DOI: 10.1016/j.zemedi.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 01/03/2024]
Abstract
Over the last two decades, hyperpolarized 13C MRI has gained significance in both preclinical and clinical studies, hereby relying on technologies like PHIP-SAH (ParaHydrogen-Induced Polarization-Side Arm Hydrogenation), SABRE (Signal Amplification by Reversible Exchange), and dDNP (dissolution Dynamic Nuclear Polarization), with dDNP being applied in humans. A clinical dDNP polarizer has enabled studies across 24 sites, despite challenges like high cost and slow polarization. Parahydrogen-based techniques like SABRE and PHIP offer faster, more cost-efficient alternatives but require molecule-specific optimization. The focus has been on imaging metabolism of hyperpolarized probes, which requires long T1, high polarization and rapid contrast generation. Efforts to establish novel probes, improve acquisition techniques and enhance data analysis methods including artificial intelligence are ongoing. Potential clinical value of hyperpolarized 13C MRI was demonstrated primarily for treatment response assessment in oncology, but also in cardiology, nephrology, hepatology and CNS characterization. In this review on biomedical hyperpolarized 13C MRI, we summarize important and recent advances in polarization techniques, probe development, acquisition and analysis methods as well as clinical trials. Starting from those we try to sketch a trajectory where the field of biomedical hyperpolarized 13C MRI might go.
Collapse
Affiliation(s)
- Pascal Wodtke
- Department of Nuclear Medicine, TUM School of Medicine and Health, Klinikum rechts der Isar of Technical University of Munich, 81675 Munich, Germany; Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, United Kingdom; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge UK
| | - Martin Grashei
- Department of Nuclear Medicine, TUM School of Medicine and Health, Klinikum rechts der Isar of Technical University of Munich, 81675 Munich, Germany
| | - Franz Schilling
- Department of Nuclear Medicine, TUM School of Medicine and Health, Klinikum rechts der Isar of Technical University of Munich, 81675 Munich, Germany; Munich Institute of Biomedical Engineering, Technical University of Munich, 85748 Garching, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany.
| |
Collapse
|
14
|
Monga S, Valkovič L, Myerson SG, Neubauer S, Mahmod M, Rider OJ. Role of Cardiac Energetics in Aortic Stenosis Disease Progression: Identifying the High-risk Metabolic Phenotype. Circ Cardiovasc Imaging 2023; 16:e014863. [PMID: 37847766 PMCID: PMC10581424 DOI: 10.1161/circimaging.122.014863] [Citation(s) in RCA: 2] [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: 09/07/2022] [Accepted: 09/01/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Severe aortic stenosis (AS) is associated with left ventricular (LV) hypertrophy and cardiac metabolic alterations with evidence of steatosis and impaired myocardial energetics. Despite this common phenotype, there is an unexplained and wide individual heterogeneity in the degree of hypertrophy and progression to myocardial fibrosis and heart failure. We sought to determine whether the cardiac metabolic state may underpin this variability. METHODS We recruited 74 asymptomatic participants with AS and 13 healthy volunteers. Cardiac energetics were measured using phosphorus spectroscopy to define the myocardial phosphocreatine to adenosine triphosphate ratio. Myocardial lipid content was determined using proton spectroscopy. Cardiac function was assessed by cardiovascular magnetic resonance cine imaging. RESULTS Phosphocreatine/adenosine triphosphate was reduced early and significantly across the LV wall thickness quartiles (Q2, 1.50 [1.21-1.71] versus Q1, 1.64 [1.53-1.94]) with a progressive decline with increasing disease severity (Q4, 1.48 [1.18-1.70]; P=0.02). Myocardial triglyceride content levels were overall higher in all the quartiles with a significant increase seen across the AV pressure gradient quartiles (Q2, 1.36 [0.86-1.98] versus Q1, 1.03 [0.81-1.56]; P=0.034). While all AS groups had evidence of subclinical LV dysfunction with impaired strain parameters, impaired systolic longitudinal strain was related to the degree of energetic impairment (r=0.219; P=0.03). Phosphocreatine/adenosine triphosphate was not only an independent predictor of LV wall thickness (r=-0.20; P=0.04) but also strongly associated with myocardial fibrosis (r=-0.24; P=0.03), suggesting that metabolic changes play a role in disease progression. The metabolic and functional parameters showed comparable results when graded by clinical severity of AS. CONCLUSIONS A gradient of myocardial energetic deficit and steatosis exists across the spectrum of hypertrophied AS hearts, and these metabolic changes precede irreversible LV remodeling and subclinical dysfunction. As such, cardiac metabolism may play an important and potentially causal role in disease progression.
Collapse
Affiliation(s)
- Shveta Monga
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| | - Ladislav Valkovič
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia (L.V.)
| | - Saul G. Myerson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| | - Masliza Mahmod
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| | - Oliver J. Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (S.M., L.V., S.G.M., S.N., M.M., O.J.R.)
| |
Collapse
|
15
|
Hesse F, Wright A, Bulat F, Kreis F, Brindle KM. Assessment of the sensitivity of 2 H MR spectroscopy measurements of [2,3- 2 H 2 ]fumarate metabolism for detecting tumor cell death. NMR IN BIOMEDICINE 2023; 36:e4965. [PMID: 37148156 PMCID: PMC10909471 DOI: 10.1002/nbm.4965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Imaging the metabolism of [2,3-2 H2 ]fumarate to produce malate can be used to detect tumor cell death post-treatment. Here, we assess the sensitivity of the technique for detecting cell death by lowering the concentration of injected [2,3-2 H2 ]fumarate and by varying the extent of tumor cell death through changes in drug concentration. Mice were implanted subcutaneously with human triple negative breast cancer cells (MDA-MB-231) and injected with 0.1, 0.3, and 0.5 g/kg [2,3-2 H2 ]fumarate before and after treatment with a multivalent TRAlL-R2 agonist (MEDI3039) at 0.1, 0.4, and 0.8 mg/kg. Tumor conversion of [2,3-2 H2 ]fumarate to [2,3-2 H2 ]malate was assessed from a series of 13 spatially localized 2 H MR spectra acquired over 65 min using a pulse-acquire sequence with a 2-ms BIR4 adiabatic excitation pulse. Tumors were then excised and stained for histopathological markers of cell death: cleaved caspase 3 (CC3) and DNA damage (terminal deoxynucleotidyl transferase dUTP nick end labeling [TUNEL]). The rate of malate production and the malate/fumarate ratio plateaued at tumor fumarate concentrations of 2 mM, which were obtained with injected [2,3-2 H2 ]fumarate concentrations of 0.3 g/kg and above. Tumor malate concentration and the malate/fumarate ratio increased linearly with the extent of cell death determined histologically. At an injected [2,3-2 H2 ]fumarate concentration of 0.3 g/kg, 20% CC3 staining corresponded to a malate concentration of 0.62 mM and a malate/fumarate ratio of 0.21. Extrapolation indicated that there would be no detectable malate at 0% CC3 staining. The use of low and nontoxic fumarate concentrations and the production of [2,3-2 H2 ]malate at concentrations that are within the range that can be detected clinically suggest this technique could translate to the clinic.
Collapse
Affiliation(s)
- Friederike Hesse
- Cancer Research UK Cambridge InstituteCambridgeUK
- Department of RadiologyUniversity of CambridgeCambridgeUK
| | - Alan Wright
- Guy's and St Thomas's NHS Foundation TrustSt Thomas' HospitalLondonUK
| | - Flaviu Bulat
- Cancer Research UK Cambridge InstituteCambridgeUK
- Department of ChemistryUniversity of CambridgeCambridgeUK
| | - Felix Kreis
- Cancer Research UK Cambridge InstituteCambridgeUK
| | - Kevin M. Brindle
- Cancer Research UK Cambridge InstituteCambridgeUK
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|
16
|
Watson WD, Green PG, Lewis AJ, Arvidsson P, De Maria GL, Arheden H, Heiberg E, Clarke WT, Rodgers CT, Valkovič L, Neubauer S, Herring N, Rider OJ. Retained Metabolic Flexibility of the Failing Human Heart. Circulation 2023; 148:109-123. [PMID: 37199155 PMCID: PMC10417210 DOI: 10.1161/circulationaha.122.062166] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 05/01/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND The failing heart is traditionally described as metabolically inflexible and oxygen starved, causing energetic deficit and contractile dysfunction. Current metabolic modulator therapies aim to increase glucose oxidation to increase oxygen efficiency of adenosine triphosphate production, with mixed results. METHODS To investigate metabolic flexibility and oxygen delivery in the failing heart, 20 patients with nonischemic heart failure with reduced ejection fraction (left ventricular ejection fraction 34.9±9.1) underwent separate infusions of insulin+glucose infusion (I+G) or Intralipid infusion. We used cardiovascular magnetic resonance to assess cardiac function and measured energetics using phosphorus-31 magnetic resonance spectroscopy. To investigate the effects of these infusions on cardiac substrate use, function, and myocardial oxygen uptake (MVo2), invasive arteriovenous sampling and pressure-volume loops were performed (n=9). RESULTS At rest, we found that the heart had considerable metabolic flexibility. During I+G, cardiac glucose uptake and oxidation were predominant (70±14% total energy substrate for adenosine triphosphate production versus 17±16% for Intralipid; P=0.002); however, no change in cardiac function was seen relative to basal conditions. In contrast, during Intralipid infusion, cardiac long-chain fatty acid (LCFA) delivery, uptake, LCFA acylcarnitine production, and fatty acid oxidation were all increased (LCFA 73±17% of total substrate versus 19±26% total during I+G; P=0.009). Myocardial energetics were better with Intralipid compared with I+G (phosphocreatine/adenosine triphosphate 1.86±0.25 versus 2.01±0.33; P=0.02), and systolic and diastolic function were improved (LVEF 34.9±9.1 baseline, 33.7±8.2 I+G, 39.9±9.3 Intralipid; P<0.001). During increased cardiac workload, LCFA uptake and oxidation were again increased during both infusions. There was no evidence of systolic dysfunction or lactate efflux at 65% maximal heart rate, suggesting that a metabolic switch to fat did not cause clinically meaningful ischemic metabolism. CONCLUSIONS Our findings show that even in nonischemic heart failure with reduced ejection fraction with severely impaired systolic function, significant cardiac metabolic flexibility is retained, including the ability to alter substrate use to match both arterial supply and changes in workload. Increasing LCFA uptake and oxidation is associated with improved myocardial energetics and contractility. Together, these findings challenge aspects of the rationale underlying existing metabolic therapies for heart failure and suggest that strategies promoting fatty acid oxidation may form the basis for future therapies.
Collapse
Affiliation(s)
- William D. Watson
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
- Department of Cardiovascular Medicine (W.D.W.), University of Cambridge, UK
| | - Peregrine G. Green
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
- Department for Physiology, Anatomy and Genetics (P.G.G., N.H.), University of Oxford, UK
| | - Andrew J.M. Lewis
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
| | - Per Arvidsson
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden (P.A., H.A., E.H.)
| | | | - Håkan Arheden
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden (P.A., H.A., E.H.)
| | - Einar Heiberg
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden (P.A., H.A., E.H.)
| | - William T. Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences (W.T.C.), University of Oxford, UK
| | | | - Ladislav Valkovič
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
- Institute of Measurement Science, Slovak Academy of Sciences, Slovakia (L.V.)
| | - Stefan Neubauer
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
| | - Neil Herring
- Department for Physiology, Anatomy and Genetics (P.G.G., N.H.), University of Oxford, UK
| | - Oliver J. Rider
- Oxford Centre for Magnetic Resonance Research (W.D.W., P.G.G., A.J.M.L., P.A., L.V., S.N., O.J.R.), University of Oxford, UK
| |
Collapse
|
17
|
Hundertmark MJ, Adler A, Antoniades C, Coleman R, Griffin JL, Holman RR, Lamlum H, Lee J, Massey D, Miller JJ, Milton JE, Monga S, Mózes FE, Nazeer A, Raman B, Rider O, Rodgers CT, Valkovič L, Wicks E, Mahmod M, Neubauer S. Assessment of Cardiac Energy Metabolism, Function, and Physiology in Patients With Heart Failure Taking Empagliflozin: The Randomized, Controlled EMPA-VISION Trial. Circulation 2023; 147:1654-1669. [PMID: 37070436 PMCID: PMC10212585 DOI: 10.1161/circulationaha.122.062021] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/16/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Sodium-glucose co-transporter 2 inhibitors (SGLT2i) have emerged as a paramount treatment for patients with heart failure (HF), irrespective of underlying reduced or preserved ejection fraction. However, a definite cardiac mechanism of action remains elusive. Derangements in myocardial energy metabolism are detectable in all HF phenotypes, and it was proposed that SGLT2i may improve energy production. The authors aimed to investigate whether treatment with empagliflozin leads to changes in myocardial energetics, serum metabolomics, and cardiorespiratory fitness. METHODS EMPA-VISION (Assessment of Cardiac Energy Metabolism, Function and Physiology in Patients With Heart Failure Taking Empagliflozin) is a prospective, randomized, double-blind, placebo-controlled, mechanistic trial that enrolled 72 symptomatic patients with chronic HF with reduced ejection fraction (HFrEF; n=36; left ventricular ejection fraction ≤40%; New York Heart Association class ≥II; NT-proBNP [N-terminal pro-B-type natriuretic peptide] ≥125 pg/mL) and HF with preserved ejection fraction (HFpEF; n=36; left ventricular ejection fraction ≥50%; New York Heart Association class ≥II; NT-proBNP ≥125 pg/mL). Patients were stratified into respective cohorts (HFrEF versus HFpEF) and randomly assigned to empagliflozin (10 mg; n=35: 17 HFrEF and 18 HFpEF) or placebo (n=37: 19 HFrEF and 18 HFpEF) once daily for 12 weeks. The primary end point was a change in the cardiac phosphocreatine:ATP ratio (PCr/ATP) from baseline to week 12, determined by phosphorus magnetic resonance spectroscopy at rest and during peak dobutamine stress (65% of age-maximum heart rate). Mass spectrometry on a targeted set of 19 metabolites was performed at baseline and after treatment. Other exploratory end points were investigated. RESULTS Empagliflozin treatment did not change cardiac energetics (ie, PCr/ATP) at rest in HFrEF (adjusted mean treatment difference [empagliflozin - placebo], -0.25 [95% CI, -0.58 to 0.09]; P=0.14) or HFpEF (adjusted mean treatment difference, -0.16 [95% CI, -0.60 to 0.29]; P=0.47]. Likewise, there were no changes in PCr/ATP during dobutamine stress in HFrEF (adjusted mean treatment difference, -0.13 [95% CI, -0.35 to 0.09]; P=0.23) or HFpEF (adjusted mean treatment difference, -0.22 [95% CI, -0.66 to 0.23]; P=0.32). No changes in serum metabolomics or levels of circulating ketone bodies were observed. CONCLUSIONS In patients with either HFrEF or HFpEF, treatment with 10 mg of empagliflozin once daily for 12 weeks did not improve cardiac energetics or change circulating serum metabolites associated with energy metabolism when compared with placebo. Based on our results, it is unlikely that enhancing cardiac energy metabolism mediates the beneficial effects of SGLT2i in HF. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03332212.
Collapse
Affiliation(s)
- Moritz J. Hundertmark
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
- Department of Internal Medicine I, University Hospital Wuerzburg, Germany (M.J.H.)
| | - Amanda Adler
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (A.A., R.C., R.R.H., J.E.M.), University of Oxford, UK
| | - Charalambos Antoniades
- Acute Multidisciplinary Imaging and Interventional Centre (C.A., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Ruth Coleman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (A.A., R.C., R.R.H., J.E.M.), University of Oxford, UK
| | | | - Rury R. Holman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (A.A., R.C., R.R.H., J.E.M.), University of Oxford, UK
| | - Hanan Lamlum
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Jisoo Lee
- John Radcliffe Hospital, Oxford University Hospitals National Health Service Foundation Trust, UK (J.L., E.W.)
| | - Daniel Massey
- Elderbrook Solutions GmbH on behalf of Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany (D.M.)
| | - Jack J.J.J. Miller
- Department of Physics (J.M.), University of Oxford, UK
- Department of Clinical Medicine, Aarhus University, Denmark (J.J.M.)
| | - Joanne E. Milton
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (A.A., R.C., R.R.H., J.E.M.), University of Oxford, UK
| | - Shveta Monga
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Ferenc E. Mózes
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Areesha Nazeer
- Oxford National Institutes of Health and Care Research Biomedical Research Centre, Oxford University Hospitals, Oxford, UK (R.R.H.)
| | - Betty Raman
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Oliver Rider
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Christopher T. Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, Cambridge Biomedical Campus, UK (C.T.R.)
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava (L.V.)
| | - Eleanor Wicks
- John Radcliffe Hospital, Oxford University Hospitals National Health Service Foundation Trust, UK (J.L., E.W.)
| | - Masliza Mahmod
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research (M.J.H., H.L., S.M., F.E.M., B.R., O.R., C.T.R., L.V., M.M., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
- Acute Multidisciplinary Imaging and Interventional Centre (C.A., S.N.), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, UK
| |
Collapse
|
18
|
Nam KM, Gursan A, Bhogal AA, Wijnen JP, Klomp DWJ, Prompers JJ, Hendriks AD. Deuterium echo-planar spectroscopic imaging (EPSI) in the human liver in vivo at 7 T. Magn Reson Med 2023. [PMID: 37154391 DOI: 10.1002/mrm.29696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/04/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE To demonstrate the feasibility of deuterium echo-planar spectroscopic imaging (EPSI) to accelerate 3D deuterium metabolic imaging in the human liver at 7 T. METHODS A deuterium EPSI sequence, featuring a Hamming-weighted k-space acquisition pattern for the phase-encoding directions, was implemented. Three-dimensional deuterium EPSI and conventional MRSI were performed on a water/acetone phantom and in vivo in the human liver at natural abundance. Moreover, in vivo deuterium EPSI measurements were acquired after oral administration of deuterated glucose. The effect of acquisition time on SNR was evaluated by retrospectively reducing the number of averages. RESULTS The SNR of natural abundance deuterated water signal in deuterium EPSI was 6.5% and 5.9% lower than that of MRSI in the phantom and in vivo experiments, respectively. In return, the acquisition time of in vivo EPSI data could be reduced retrospectively to 2 min, beyond the minimal acquisition time of conventional MRSI (of 20 min in this case), while still leaving sufficient SNR. Three-dimensional deuterium EPSI, after administration of deuterated glucose, enabled monitoring of hepatic glucose dynamics with full liver coverage, a spatial resolution of 20 mm isotropic, and a temporal resolution of 9 min 50 s, which could retrospectively be shortened to 2 min. CONCLUSION In this work, we demonstrate the feasibility of accelerated 3D deuterium metabolic imaging of the human liver using deuterium EPSI. The acceleration obtained with EPSI can be used to increase temporal and/or spatial resolution, which will be valuable to study tissue metabolism of deuterated compounds over time.
Collapse
Affiliation(s)
- Kyung Min Nam
- Center for Image Sciences, Department of High Field MR Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ayhan Gursan
- Center for Image Sciences, Department of High Field MR Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Alex A Bhogal
- Center for Image Sciences, Department of High Field MR Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jannie P Wijnen
- Center for Image Sciences, Department of High Field MR Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Dennis W J Klomp
- Center for Image Sciences, Department of High Field MR Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jeanine J Prompers
- Center for Image Sciences, Department of High Field MR Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Arjan D Hendriks
- Center for Image Sciences, Department of High Field MR Research, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
19
|
Finnigan LEM, Cassar MP, Koziel MJ, Pradines J, Lamlum H, Azer K, Kirby D, Montgomery H, Neubauer S, Valkovič L, Raman B. Efficacy and tolerability of an endogenous metabolic modulator (AXA1125) in fatigue-predominant long COVID: a single-centre, double-blind, randomised controlled phase 2a pilot study. EClinicalMedicine 2023; 59:101946. [PMID: 37223439 PMCID: PMC10102537 DOI: 10.1016/j.eclinm.2023.101946] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 05/25/2023] Open
Abstract
Background 'Long COVID' describes persistent symptoms, commonly fatigue, lasting beyond 12 weeks following SARS-CoV-2 infection. Potential causes include reduced mitochondrial function and cellular bioenergetics. AXA1125 has previously increased β-oxidation and improved bioenergetics in preclinical models along with certain clinical conditions, and therefore may reduce fatigue associated with Long COVID. We aimed to assess the efficacy, safety and tolerability of AXA1125 in Long COVID. Methods Patients with fatigue-dominant Long COVID were recruited in this single-centre, double-blind, randomised controlled phase 2a pilot study completed in the UK. Patients were randomly assigned (1:1) using an Interactive Response Technology to receive either AXA1125 or matching placebo in a clinical-based setting. Each dose (33.9 g) of AXA1125 or placebo was administered orally in a liquid suspension twice daily for four weeks with a two-week follow-up period. The primary endpoint was the mean change from baseline to day 28 in the phosphocreatine (PCr) recovery rate following moderate exercise, assessed by 31P-magnetic resonance spectroscopy (MRS). All patients were included in the intention to treat analysis. This trial was registered at ClinicalTrials.gov, NCT05152849. Findings Between December 15th 2021, and May 23th 2022, 60 participants were screened, and 41 participants were randomised and included in the final analysis. Changes in skeletal muscle phosphocreatine recovery time constant (τPCr) and 6-min walk test (6MWT) did not significantly differ between treatment (n = 21) and placebo group (n = 20). However, treatment with AXA1125 was associated with significantly reduced day 28 Chalder Fatigue Questionnaire [CFQ-11] fatigue score when compared with placebo (least squares mean difference [LSMD] -4.30, 95% confidence interval (95% CI) -7.14, -1.47; P = 0.0039). Eleven (52.4%, AXA1125) and four (20.0%, placebo) patients reported treatment-emergent adverse events; none were serious or led to treatment discontinuation. Interpretation Although treatment with AXA1125 did not improve the primary endpoint (τPCr-measure of mitochondrial respiration), when compared to placebo, there were significant improvements in fatigue-based symptoms among patients living with Long COVID following a four-week treatment period. Further multicentre studies are needed to validate our findings in a larger cohort of patients with fatigue-dominant Long COVID. Funding Axcella Therapeutics.
Collapse
Affiliation(s)
- Lucy E M Finnigan
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Mark Philip Cassar
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | | | - Hanan Lamlum
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Karim Azer
- Axcella Therapeutics, Cambridge, MA, USA
| | - Dan Kirby
- Axcella Therapeutics, Cambridge, MA, USA
| | | | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Betty Raman
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford, UK
| |
Collapse
|
20
|
van den Wildenberg L, Gursan A, Seelen LWF, van der Velden TA, Gosselink MWJM, Froeling M, van der Kemp WJM, Klomp DWJ, Prompers JJ. In vivo phosphorus magnetic resonance spectroscopic imaging of the whole human liver at 7 T using a phosphorus whole-body transmit coil and 16-channel receive array: Repeatability and effects of principal component analysis-based denoising. NMR IN BIOMEDICINE 2023; 36:e4877. [PMID: 36400716 DOI: 10.1002/nbm.4877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 06/16/2023]
Abstract
Quantitative three-dimensional (3D) imaging of phosphorus (31 P) metabolites is potentially a promising technique with which to assess the progression of liver disease and monitor therapy response. However, 31 P magnetic resonance spectroscopy has a low sensitivity and commonly used 31 P surface coils do not provide full coverage of the liver. This study aimed to overcome these limitations by using a 31 P whole-body transmit coil in combination with a 16-channel 31 P receive array at 7 T. Using this setup, we determined the repeatability of whole-liver 31 P magnetic resonance spectroscopic imaging (31 P MRSI) in healthy subjects and assessed the effects of principal component analysis (PCA)-based denoising on the repeatability parameters. In addition, spatial variations of 31 P metabolites within the liver were analyzed. 3D 31 P MRSI data of the liver were acquired with a nominal voxel size of 20 mm isotropic in 10 healthy volunteers twice on the same day. Data were reconstructed without denoising, and with PCA-based denoising before or after channel combination. From the test-retest data, repeatability parameters for metabolite level quantification were determined for 12 31 P metabolite signals. On average, 31 P MR spectra from 100 ± 25 voxels in the liver were analyzed. Only voxels with contamination from skeletal muscle or the gall bladder were excluded and no voxels were discarded based on (low) signal-to-noise ratio (SNR). Repeatability for most quantified 31 P metabolite levels in the liver was good to excellent, with an intrasubject variability below 10%. PCA-based denoising increased the SNR ~ 3-fold, but did not improve the repeatability for mean liver 31 P metabolite quantification with the fitting constraints used. Significant spatial heterogeneity of various 31 P metabolite levels within the liver was observed, with marked differences for the phosphomonoester and phosphodiester metabolites between the left and right lobe. In conclusion, using a 31 P whole-body transmit coil in combination with a 16-channel 31 P receive array at 7 T allowed 31 P MRSI acquisitions with full liver coverage and good to excellent repeatability.
Collapse
Affiliation(s)
| | - Ayhan Gursan
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leonard W F Seelen
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tijl A van der Velden
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mark W J M Gosselink
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martijn Froeling
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wybe J M van der Kemp
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis W J Klomp
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeanine J Prompers
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
21
|
Tyler A, Ellis J, Lau JYC, Miller JJ, Bottomley PA, Rodgers CT, Tyler DJ, Valkovič L. Compartment-based reconstruction of 3D acquisition-weighted 31 P cardiac magnetic resonance spectroscopic imaging at 7 T: A reproducibility study. NMR IN BIOMEDICINE 2023; 36:e4950. [PMID: 37046414 PMCID: PMC10658645 DOI: 10.1002/nbm.4950] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/22/2022] [Accepted: 03/06/2023] [Indexed: 05/06/2023]
Abstract
Even at 7 T, cardiac 31 P magnetic resonance spectroscopic imaging (MRSI) is fundamentally limited by low signal-to-noise ratio (SNR), leading to long scan times and poor temporal and spatial resolutions. Compartment-based reconstruction algorithms such as magnetic resonance spectroscopy with linear algebraic modeling (SLAM) and spectral localization by imaging (SLIM) may improve SNR or reduce scan time without changes to acquisition. Here, we compare the repeatability and SNR performance of these compartment-based methods, applied to three different acquisition schemes at 7 T. Twelve healthy volunteers were scanned twice. Each scan session consisted of a 6.5-min 3D acquisition-weighted (AW) cardiac 31 P phase encode-based MRSI acquisition and two 6.5-min truncated k-space acquisitions with increased averaging (4 × 4 × 4 central k-space phase encodes and fractional SLAM [fSLAM] optimized k-space phase encodes). Spectra were reconstructed using (i) AW Fourier reconstruction; (ii) AW SLAM; (iii) AW SLIM; (iv) 4 × 4 × 4 SLAM; (v) 4 × 4 × 4 SLIM; and (vi) fSLAM acquisition-reconstruction combinations. The phosphocreatine-to-adenosine triphosphate (PCr/ATP) ratio, the PCr SNR, and spatial response functions were computed, in addition to coefficients of reproducibility and variability. Using the compartment-based reconstruction algorithms with the AW 31 P acquisition resulted in a significant increase in SNR compared with previously published Fourier-based MRSI reconstruction methods while maintaining the measured PCr/ATP ratio and improving interscan reproducibility. The alternative acquisition strategies with truncated k-space performed no better than the common AW approach. Compartment-based spectroscopy approaches provide an attractive reconstruction method for cardiac 31 P spectroscopy at 7 T, improving reproducibility and SNR without the need for a dedicated k-space sampling strategy.
Collapse
Affiliation(s)
- Andrew Tyler
- Department of Physiology, Anatomy & GeneticsUniversity of OxfordOxfordUK
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), RDM Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Jane Ellis
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), RDM Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Justin Y. C. Lau
- Department of Physiology, Anatomy & GeneticsUniversity of OxfordOxfordUK
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), RDM Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Jack J. Miller
- Department of Physiology, Anatomy & GeneticsUniversity of OxfordOxfordUK
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), RDM Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Physics, Clarendon LaboratoryUniversity of OxfordOxfordUK
- The MR Research Centre & The PET Research CentreAarhus UniversityAarhusDenmark
| | - Paul A. Bottomley
- The Division of MR ResearchJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Christopher T. Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), RDM Cardiovascular MedicineUniversity of OxfordOxfordUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Damian J. Tyler
- Department of Physiology, Anatomy & GeneticsUniversity of OxfordOxfordUK
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), RDM Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), RDM Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Imaging Methods, Institute of Measurement ScienceSlovak Academy of SciencesBratislavaSlovakia
| |
Collapse
|
22
|
Savic D, Mózes FE, Green PG, Burrage MK, Kjær MS, Hodson L, Neubauer S, Pavlides M, Valkovič L. Detection and alterations of acetylcarnitine (AC) in human liver by 1 H MRS at 3T after supplementation with l-carnitine. Magn Reson Med 2023; 89:1314-1322. [PMID: 36573435 PMCID: PMC11497247 DOI: 10.1002/mrm.29544] [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: 08/18/2022] [Revised: 10/24/2022] [Accepted: 11/14/2022] [Indexed: 12/28/2022]
Abstract
PURPOSE Acetylcarnitine can be assessed in vivo using proton MRS (1 H-MRS) with long TEs and this has been previously applied successfully in muscle. The aim of this study was to evaluate a 1 H-MRS technique for liver acetylcarnitine quantification in healthy humans before and after l-carnitine supplementation. METHOD Baseline acetylcarnitine levels were quantified using a STEAM sequence with prolonged TE in 15 healthy adults. Using STEAM with four different TEs was evaluated in phantoms. To assess reproducibility of the measurements, five of the participants had repeated 1 H-MRS without receiving l-carnitine supplementation. To determine if liver acetylcarnitine could be changed after l-carnitine supplementation, acetylcarnitine was quantified 2 h after intravenous l-carnitine supplementation (50 mg/kg body weight) in the other 10 participants. Hepatic lipids were also quantified from the 1 H-MRS spectra. RESULTS There was good separation between the acetylcarnitine and fat in the phantoms using TE = 100 ms. Hepatic acetylcarnitine levels were reproducible (coefficient of reproducibility = 0.049%) and there was a significant (p < 0.001) increase in the relative abundance after a single supplementation of l-carnitine. Hepatic allylic, methyl, and methylene peaks were not altered by l-carnitine supplementation in healthy volunteers. CONCLUSION Our results demonstrate that our 1 H-MRS technique could be used to measure acetylcarnitine in the liver and detect changes following intravenous supplementation in healthy adults despite the presence of lipids. Our techniques should be explored further in the study of fatty liver disease, where acetylcarnitine is suggested to be altered due to hepatic inflexibilities.
Collapse
Affiliation(s)
- Dragana Savic
- The Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of MedicineUniversity of Oxford
OxfordUK
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
| | - Ferenc E. Mózes
- The Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of MedicineUniversity of Oxford
OxfordUK
| | - Peregrine G. Green
- The Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of MedicineUniversity of Oxford
OxfordUK
| | - Matthew K. Burrage
- The Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of MedicineUniversity of Oxford
OxfordUK
- Faculty of MedicineUniversity of QueenslandSt LuciaQueenslandAustralia
| | | | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
- Oxford NIHR Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Stefan Neubauer
- The Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of MedicineUniversity of Oxford
OxfordUK
| | - Michael Pavlides
- The Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of MedicineUniversity of Oxford
OxfordUK
- Oxford NIHR Biomedical Research CentreUniversity of OxfordOxfordUK
- Translational Gastroenterology UnitUniversity of OxfordOxfordUK
| | - Ladislav Valkovič
- The Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of MedicineUniversity of Oxford
OxfordUK
- Department of Imaging MethodsInstitute of Measurement Science, Slovak Academy of SciencesBratislavaSlovakia
| |
Collapse
|
23
|
Gursan A, Hendriks AD, Welting D, de Jong PA, Klomp DWJ, Prompers JJ. Deuterium body array for the simultaneous measurement of hepatic and renal glucose metabolism and gastric emptying with dynamic 3D deuterium metabolic imaging at 7 T. NMR IN BIOMEDICINE 2023:e4926. [PMID: 36929629 DOI: 10.1002/nbm.4926] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Deuterium metabolic imaging (DMI) is a novel noninvasive method to assess tissue metabolism and organ (patho)physiology in vivo using deuterated substrates, such as [6,6'-2 H2 ]-glucose. The liver and kidneys play a central role in whole-body glucose homeostasis, and in type 2 diabetes, both hepatic and renal glucose metabolism are dysregulated. Diabetes is also associated with gastric emptying abnormalities. In this study, we developed a four-channel 2 H transmit/receive body array coil for DMI in the human abdomen at 7 T and assessed its performance. In addition, the feasibility of simultaneously measuring gastric emptying, and hepatic and renal glucose uptake and metabolism with dynamic 3D DMI upon administration of deuterated glucose, was investigated. Simulated and measured B1 + patterns were in good agreement. The intrasession variability of the natural abundance deuterated water signal in the liver and right kidney, measured in nine healthy volunteers, was 5.6% ± 0.9% and 4.9% ± 0.7%, respectively. Dynamic 3D DMI scans with oral administration of [6,6'-2 H2 ]-glucose showed similar kinetics of deuterated glucose appearance and disappearance in the liver and kidney. The measured gastric emptying half time was 80 ± 10 min, which is in good agreement with scintigraphy measurements. In conclusion, DMI with oral administration of [6,6'-2 H2 ]-glucose enables simultaneous assessment of gastric emptying and liver and kidney glucose uptake and metabolism. When applied in patients with diabetes, this approach may advance our understanding of the interplay between disturbances in liver and kidney glucose uptake and metabolism and gastric emptying, at a detail that cannot be achieved by any other method.
Collapse
Affiliation(s)
- Ayhan Gursan
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arjan D Hendriks
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dimitri Welting
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pim A de Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis W J Klomp
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeanine J Prompers
- Center for Image Sciences, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
24
|
Clarke WT, Hingerl L, Strasser B, Bogner W, Valkovič L, Rodgers CT. Three-dimensional, 2.5-minute, 7T phosphorus magnetic resonance spectroscopic imaging of the human heart using concentric rings. NMR IN BIOMEDICINE 2023; 36:e4813. [PMID: 35995750 PMCID: PMC7613900 DOI: 10.1002/nbm.4813] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/27/2022] [Accepted: 08/10/2022] [Indexed: 05/06/2023]
Abstract
A three-dimensional (3D), density-weighted, concentric rings trajectory (CRT) magnetic resonance spectroscopic imaging (MRSI) sequence is implemented for cardiac phosphorus (31 P)-MRS at 7 T. The point-by-point k-space sampling of traditional phase-encoded chemical shift imaging (CSI) sequences severely restricts the minimum scan time at higher spatial resolutions. Our proposed CRT sequence implements a stack of concentric rings, with a variable number of rings and planes spaced to optimise the density of k-space weighting. This creates flexibility in acquisition time, allowing acquisitions substantially faster than traditional phase-encoded CSI sequences, while retaining high signal-to-noise ratio (SNR). We first characterise the SNR and point-spread function of the CRT sequence in phantoms. We then evaluate it at five different acquisition times and spatial resolutions in the hearts of five healthy participants at 7 T. These different sequence durations are compared with existing published 3D acquisition-weighted CSI sequences with matched acquisition times and spatial resolutions. To minimise the effect of noise on the short acquisitions, low-rank denoising of the spatiotemporal data was also performed after acquisition. The proposed sequence measures 3D localised phosphocreatine to adenosine triphosphate (PCr/ATP) ratios of the human myocardium in 2.5 min, 2.6 times faster than the minimum scan time for acquisition-weighted phase-encoded CSI. Alternatively, in the same scan time, a 1.7-times smaller nominal voxel volume can be achieved. Low-rank denoising reduced the variance of measured PCr/ATP ratios by 11% across all protocols. The faster acquisitions permitted by 7-T CRT 31 P-MRSI could make cardiac stress protocols or creatine kinase rate measurements (which involve repeated scans) more tolerable for patients without sacrificing spatial resolution.
Collapse
Affiliation(s)
- William T. Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Lukas Hingerl
- High‐field MR Centre, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Bernhard Strasser
- High‐field MR Centre, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Wolfgang Bogner
- High‐field MR Centre, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
- Department of Imaging Methods, Institute of Measurement ScienceSlovak Academy of SciencesBratislavaSlovakia
| | - Christopher T. Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
- Wolfson Brain Imaging Centre, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| |
Collapse
|
25
|
Prasuhn J, Göttlich M, Ebeling B, Kourou S, Gerkan F, Bodemann C, Großer SS, Reuther K, Hanssen H, Brüggemann N. Assessment of Bioenergetic Deficits in Patients With Parkinson Disease and Progressive Supranuclear Palsy Using 31P-MRSI. Neurology 2022; 99:e2683-e2692. [PMID: 36195453 DOI: 10.1212/wnl.0000000000201288] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 08/10/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Bioenergetic disturbance, mainly caused by mitochondrial dysfunction, is an established pathophysiologic phenomenon in neurodegenerative movement disorders. The in vivo assessment of brain energy metabolism by 31phosphorus magnetic resonance spectroscopy imaging could provide pathophysiologic insights and serve in the differential diagnosis of parkinsonian disorders. In this study, we investigated such aspects of the underlying pathophysiology in patients with idiopathic Parkinson disease (PwPD) and progressive supranuclear palsy (PwPSP). METHODS In total, 30 PwPD, 16 PwPSP, and 25 healthy control subjects (HCs) underwent a clinical examination, structural magnetic resonance imaging, and 31phosphorus magnetic resonance spectroscopy imaging of the forebrain and basal ganglia in a cross-sectional study. RESULTS High-energy phosphate metabolites were remarkably decreased in PwPD, particularly in the basal ganglia (-42% compared with HCs and -43% compared with PwPSP, p < 0.0001). This result was not confounded by morphometric brain differences. By contrast, PwPSP had normal levels of high-energy energy metabolites. Thus, the combination of morphometric and metabolic neuroimaging was able to discriminate PwPD from PwPSP with an accuracy of up to 0.93 [95%-CI: 0.91-0.94]. DISCUSSION Our study shows that mitochondrial dysfunction and bioenergetic depletion contribute to idiopathic Parkinson disease pathophysiology but not to progressive supranuclear palsy. Combined morphometric and metabolic imaging could serve as an accompanying diagnostic biomarker in the neuroimaging-guided differential diagnosis of these parkinsonian disorders. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that 31phosphorus magnetic resonance spectroscopy imaging combined with morphometric MRI can differentiate PwPD from PwPSP.
Collapse
Affiliation(s)
- Jannik Prasuhn
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Martin Göttlich
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Britt Ebeling
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Sofia Kourou
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Friederike Gerkan
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Christina Bodemann
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Sinja S Großer
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Katharina Reuther
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Henrike Hanssen
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany
| | - Norbert Brüggemann
- From the Institute of Neurogenetics (J.P., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.) and Center for Brain, Behavior, and Metabolism (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University of Lübeck, Germany; and Department of Neurology (J.P., M.G., B.E., S.K., F.G., C.B., S.S.G., K.R., H.H., N.B.), University Medical Center Schleswig-Holstein, Germany.
| |
Collapse
|
26
|
Thirunavukarasu S, Ansari F, Cubbon R, Forbes K, Bucciarelli-Ducci C, Newby DE, Dweck MR, Rider OJ, Valkovič L, Rodgers CT, Tyler DJ, Chowdhary A, Jex N, Kotha S, Morley L, Xue H, Swoboda P, Kellman P, Greenwood JP, Plein S, Everett T, Scott E, Levelt E. Maternal Cardiac Changes in Women With Obesity and Gestational Diabetes Mellitus. Diabetes Care 2022; 45:3007-3015. [PMID: 36099225 PMCID: PMC9862457 DOI: 10.2337/dc22-0401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
Abstract
OBJECTIVE We investigated if women with gestational diabetes mellitus (GDM) in the third trimester of pregnancy exhibit adverse cardiac alterations in myocardial energetics, function, or tissue characteristics. RESEARCH DESIGN AND METHODS Thirty-eight healthy, pregnant women and 30 women with GDM were recruited. Participants underwent phosphorus MRS and cardiovascular magnetic resonance for assessment of myocardial energetics (phosphocreatine [PCr] to ATP ratio), tissue characteristics, biventricular volumes and ejection fractions, left ventricular (LV) mass, global longitudinal shortening (GLS), and mitral in-flow E-wave to A-wave ratio. RESULTS Participants were matched for age, gestational age, and ethnicity. The following data are reported as mean ± SD. The women with GDM had higher BMI (27 ± 4 vs. 33 ± 5 kg/m2; P = 0.0001) and systolic (115 ± 11 vs. 121 ± 13 mmHg; P = 0.04) and diastolic (72 ± 7 vs. 76 ± 9 mmHg; P = 0.04) blood pressures. There was no difference in N-terminal pro-brain natriuretic peptide concentrations between the groups. The women with GDM had lower myocardial PCr to ATP ratio (2.2 ± 0.3 vs. 1.9 ± 0.4; P < 0.0001), accompanied by lower LV end-diastolic volumes (76 ± 12 vs. 67 ± 11 mL/m2; P = 0.002) and higher LV mass (90 ± 13 vs. 103 ± 18 g; P = 0.001). Although ventricular ejection fractions were similar, the GLS was reduced in women with GDM (-20% ± 3% vs. -18% ± 3%; P = 0.008). CONCLUSIONS Despite no prior diagnosis of diabetes, women with obesity and GDM manifest impaired myocardial contractility and higher LV mass, associated with reductions in myocardial energetics in late pregnancy compared with lean women with healthy pregnancy. These findings may aid our understanding of the long-term cardiovascular risks associated with GDM.
Collapse
Affiliation(s)
| | - Faiza Ansari
- Department of Fetal Medicine, Leeds General Infirmary, The Leeds Teaching Hospitals National Health Service Trust, Leeds, U.K
| | - Richard Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Karen Forbes
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | | | - David E. Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, U.K
| | - Marc R. Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, U.K
| | - Oliver J. Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine Cardiovascular Medicine, University of Oxford, Oxford, U.K
| | - Ladislav Valkovič
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine Cardiovascular Medicine, University of Oxford, Oxford, U.K
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Damian J. Tyler
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine Cardiovascular Medicine, University of Oxford, Oxford, U.K
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Amrit Chowdhary
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Nicholas Jex
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Sindhoora Kotha
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Lara Morley
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Hui Xue
- National Heart, Lung, and Blood Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Peter Swoboda
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Peter Kellman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - John P. Greenwood
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Sven Plein
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Thomas Everett
- Department of Fetal Medicine, Leeds General Infirmary, The Leeds Teaching Hospitals National Health Service Trust, Leeds, U.K
| | - Eleanor Scott
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Eylem Levelt
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| |
Collapse
|
27
|
Hesse F, Wright AJ, Bulat F, Somai V, Kreis F, Brindle KM. Deuterium MRSI of tumor cell death in vivo following oral delivery of 2 H-labeled fumarate. Magn Reson Med 2022; 88:2014-2020. [PMID: 35816502 PMCID: PMC9545469 DOI: 10.1002/mrm.29379] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/25/2022] [Accepted: 06/16/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE There is an unmet clinical need for direct and sensitive methods to detect cell death in vivo, especially with regard to monitoring tumor treatment response. We have shown previously that tumor cell death can be detected in vivo from 2 H MRS and MRSI measurements of increased [2,3-2 H2 ]malate production following intravenous injection of [2,3-2 H2 ]fumarate. We show here that cell death can be detected with similar sensitivity following oral administration of the 2 H-labeled fumarate. METHODS Mice with subcutaneously implanted EL4 tumors were fasted for 1 h before administration (200 μl) of [2,3-2 H2 ]fumarate (2 g/kg bodyweight) via oral gavage without anesthesia. The animals were then anesthetized, and after 30 min, tumor conversion of [2,3-2 H2 ]fumarate to [2,3-2 H2 ]malate was assessed from a series of 13 2 H spectra acquired over a period of 65 min. The 2 H spectra and 2 H spectroscopic images were acquired using a surface coil before and at 48 h after treatment with a chemotherapeutic drug (etoposide, 67 mg/kg). RESULTS The malate/fumarate signal ratio increased from 0.022 ± 0.03 before drug treatment to 0.12 ± 0.04 following treatment (p = 0.023, n = 4). Labeled malate was undetectable in spectroscopic images acquired before treatment and increased in the tumor area following treatment. The increase in the malate/fumarate signal ratio was similar to that observed previously following intravenous administration of labeled fumarate. CONCLUSION Orally administered [2,3-2 H2 ]fumarate can be used to detect tumor cell death noninvasively following treatment with a sensitivity that is similar to that obtained with intravenous administration.
Collapse
Affiliation(s)
| | | | - Flaviu Bulat
- Cancer Research UK Cambridge Institute
CambridgeUK
- Department of ChemistryUniversity of CambridgeCambridgeUK
| | - Vencel Somai
- Cancer Research UK Cambridge Institute
CambridgeUK
- Department of RadiologyUniversity of CambridgeCambridgeUK
| | - Felix Kreis
- Cancer Research UK Cambridge Institute
CambridgeUK
| | - Kevin M. Brindle
- Cancer Research UK Cambridge Institute
CambridgeUK
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|
28
|
Hesse F, Wright AJ, Somai V, Bulat F, Kreis F, Brindle KM. Imaging Glioblastoma Response to Radiotherapy Using 2H Magnetic Resonance Spectroscopy Measurements of Fumarate Metabolism. Cancer Res 2022; 82:3622-3633. [PMID: 35972377 PMCID: PMC9530651 DOI: 10.1158/0008-5472.can-22-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/25/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022]
Abstract
Early detection of tumor cell death in glioblastoma following treatment with chemoradiation has the potential to distinguish between true disease progression and pseudoprogression. Tumor cell death can be detected noninvasively in vivo by imaging the production of [2,3-2H2]malate from [2,3-2H2]fumarate using 2H magnetic resonance (MR) spectroscopic imaging. We show here that 2H MR spectroscopy and spectroscopic imaging measurements of [2,3-2H2]fumarate metabolism can detect tumor cell death in orthotopically implanted glioblastoma models within 48 hours following the completion of chemoradiation. Following the injection of [2,3-2H2]fumarate into tumor-bearing mice, production of [2,3-2H2]malate was measured in a human cell line-derived model and in radiosensitive and radioresistant patient-derived models of glioblastoma that were treated with temozolomide followed by targeted fractionated irradiation. The increase in the [2,3-2H2]malate/[2,3-2H2]fumarate signal ratio posttreatment, which correlated with histologic assessment of cell death, was a more sensitive indicator of treatment response than diffusion-weighted and contrast agent-enhanced 1H MRI measurements, which have been used clinically to detect responses of glioblastoma to chemoradiation. Overall, early detection of glioblastoma cell death using 2H MRI of malate production from fumarate could help improve the clinical evaluation of response to chemoradiation. SIGNIFICANCE 2H magnetic resonance imaging of labeled fumarate metabolism can detect early evidence of tumor cell death following chemoradiation, meeting a clinical need to reliably detect treatment response in glioblastoma.
Collapse
Affiliation(s)
- Friederike Hesse
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Alan J. Wright
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Vencel Somai
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, School of Clinical Medicine, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Flaviu Bulat
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Felix Kreis
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kevin M. Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
29
|
Kaggie JD, Khan AS, Matys T, Schulte RF, Locke MJ, Grimmer A, Frary A, Menih IH, Latimer E, Graves MJ, McLean MA, Gallagher FA. Deuterium metabolic imaging and hyperpolarized 13C-MRI of the normal human brain at clinical field strength reveals differential cerebral metabolism. Neuroimage 2022; 257:119284. [PMID: 35533826 DOI: 10.1016/j.neuroimage.2022.119284] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 12/01/2022] Open
Abstract
Deuterium metabolic imaging (DMI) and hyperpolarized 13C-pyruvate MRI (13C-HPMRI) are two emerging methods for non-invasive and non-ionizing imaging of tissue metabolism. Imaging cerebral metabolism has potential applications in cancer, neurodegeneration, multiple sclerosis, traumatic brain injury, stroke, and inborn errors of metabolism. Here we directly compare these two non-invasive methods at 3 T for the first time in humans and show how they simultaneously probe both oxidative and non-oxidative metabolism. DMI was undertaken 1-2 h after oral administration of [6,6'-2H2]glucose, and 13C-MRI was performed immediately following intravenous injection of hyperpolarized [1-13C]pyruvate in ten and nine normal volunteers within each arm respectively. DMI was used to generate maps of deuterium-labelled water, glucose, lactate, and glutamate/glutamine (Glx) and the spectral separation demonstrated that DMI is feasible at 3 T. 13C-HPMRI generated maps of hyperpolarized carbon-13 labelled pyruvate, lactate, and bicarbonate. The ratio of 13C-lactate/13C-bicarbonate (mean 3.7 ± 1.2) acquired with 13C-HPMRI was higher than the equivalent 2H-lactate/2H-Glx ratio (mean 0.18 ± 0.09) acquired using DMI. These differences can be explained by the route of administering each probe, the timing of imaging after ingestion or injection, as well as the biological differences in cerebral uptake and cellular physiology between the two molecules. The results demonstrate these two metabolic imaging methods provide different yet complementary readouts of oxidative and reductive metabolism within a clinically feasible timescale. Furthermore, as DMI was undertaken at a clinical field strength within a ten-minute scan time, it demonstrates its potential as a routine clinical tool in the future.
Collapse
Affiliation(s)
- Joshua D Kaggie
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK.
| | - Alixander S Khan
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Tomasz Matys
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK
| | | | - Matthew J Locke
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Ashley Grimmer
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Amy Frary
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Ines Horvat Menih
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Elizabeth Latimer
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK
| | - Mary A McLean
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge UK
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Jex N, Chowdhary A, Thirunavukarasu S, Procter H, Sengupta A, Natarajan P, Kotha S, Poenar AM, Swoboda P, Xue H, Cubbon RM, Kellman P, Greenwood JP, Plein S, Page S, Levelt E. Coexistent Diabetes Is Associated With the Presence of Adverse Phenotypic Features in Patients With Hypertrophic Cardiomyopathy. Diabetes Care 2022; 45:1852-1862. [PMID: 35789379 PMCID: PMC9346996 DOI: 10.2337/dc22-0083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/03/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) is associated with worsened clinical outcomes in hypertrophic cardiomyopathy (HCM) patients. We sought to investigate whether HCM patients with T2DM comorbidity exhibit adverse cardiac alterations in myocardial energetics, function, perfusion, or tissue characteristics. RESEARCH DESIGN AND METHODS A total of 55 participants with concomitant HCM and T2DM (HCM-DM) (n = 20) or isolated HCM (n = 20) and healthy volunteers (HV) (n = 15) underwent 31P-MRS and cardiovascular MRI. The HCM groups were matched for HCM phenotype. RESULTS Mean ± SD European Society of Cardiology sudden cardiac death risk scores were comparable between the HCM groups (HCM 2.2 ± 1.5%, HCM-DM 1.9 ± 1.2%; P = not significant), and sarcomeric mutations were equally common. HCM-DM patients had the highest median NT-proBNP levels (HV 42 ng/L [interquartile range 35-66], HCM 298 ng/L [157-837], HCM-DM 726 ng/L [213-8,695]; P < 0.0001). Left ventricular (LV) ejection fraction, mass, and wall thickness were similar between the HCM groups. HCM-DM patients displayed a greater degree of fibrosis burden with higher scar percentage and lower global longitudinal strain compared with HCM patients. PCr/ATP (the relative concentrations of phosphocreatine and ATP) was significantly lower in the HCM-DM group than in both HCM and HV (HV 2.17 ± 0.49, HCM 1.93 ± 0.38, HCM-DM 1.54 ± 0.27; P = 0.002). In a similar pattern, stress myocardial blood flow was significantly lower in the HCM-DM group than in both HCM and HV (HV 2.06 ± 0.42 mL/min/g, HCM 1.74 ± 0.44 mL/min/g, HCM-DM 1.39 ± 0.42 mL/min/g; P = 0.002). CONCLUSIONS We show for the first time that HCM-DM patients display greater reductions in myocardial energetics, perfusion, and contractile function and higher myocardial scar burden and serum NT-proBNP levels compared with patients with isolated HCM despite similar LV mass and wall thickness and presence of sarcomeric mutations. These adverse phenotypic features may be important components of the adverse clinical manifestation attributable to a combined presence of HCM and T2DM.
Collapse
Affiliation(s)
- Nicholas Jex
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Amrit Chowdhary
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Sharmaine Thirunavukarasu
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Henry Procter
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, U.K
| | - Anshuman Sengupta
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, U.K
| | - Pavithra Natarajan
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Sindhoora Kotha
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Ana-Maria Poenar
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, U.K
| | - Peter Swoboda
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Hui Xue
- National Heart, Lung, and Blood Institute, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD
| | - Richard M Cubbon
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Peter Kellman
- National Heart, Lung, and Blood Institute, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD
| | - John P Greenwood
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Sven Plein
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| | - Stephen Page
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, U.K
| | - Eylem Levelt
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K
| |
Collapse
|
31
|
Craven AR, Bhattacharyya PK, Clarke WT, Dydak U, Edden RAE, Ersland L, Mandal PK, Mikkelsen M, Murdoch JB, Near J, Rideaux R, Shukla D, Wang M, Wilson M, Zöllner HJ, Hugdahl K, Oeltzschner G. Comparison of seven modelling algorithms for γ-aminobutyric acid-edited proton magnetic resonance spectroscopy. NMR IN BIOMEDICINE 2022; 35:e4702. [PMID: 35078266 PMCID: PMC9203918 DOI: 10.1002/nbm.4702] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 06/01/2023]
Abstract
Edited MRS sequences are widely used for studying γ-aminobutyric acid (GABA) in the human brain. Several algorithms are available for modelling these data, deriving metabolite concentration estimates through peak fitting or a linear combination of basis spectra. The present study compares seven such algorithms, using data obtained in a large multisite study. GABA-edited (GABA+, TE = 68 ms MEGA-PRESS) data from 222 subjects at 20 sites were processed via a standardised pipeline, before modelling with FSL-MRS, Gannet, AMARES, QUEST, LCModel, Osprey and Tarquin, using standardised vendor-specific basis sets (for GE, Philips and Siemens) where appropriate. After referencing metabolite estimates (to water or creatine), systematic differences in scale were observed between datasets acquired on different vendors' hardware, presenting across algorithms. Scale differences across algorithms were also observed. Using the correlation between metabolite estimates and voxel tissue fraction as a benchmark, most algorithms were found to be similarly effective in detecting differences in GABA+. An interclass correlation across all algorithms showed single-rater consistency for GABA+ estimates of around 0.38, indicating moderate agreement. Upon inclusion of a basis set component explicitly modelling the macromolecule signal underlying the observed 3.0 ppm GABA peaks, single-rater consistency improved to 0.44. Correlation between discrete pairs of algorithms varied, and was concerningly weak in some cases. Our findings highlight the need for consensus on appropriate modelling parameters across different algorithms, and for detailed reporting of the parameters adopted in individual studies to ensure reproducibility and meaningful comparison of outcomes between different studies.
Collapse
Affiliation(s)
- Alexander R. Craven
- Department of Biological and Medical PsychologyUniversity of BergenBergenNorway
- Department of Clinical EngineeringHaukeland University HospitalBergenNorway
- NORMENT Center of ExcellenceHaukeland University HospitalBergenNorway
| | | | - William T. Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- MRC Brain Network Dynamics UnitUniversity of OxfordOxfordUK
| | - Ulrike Dydak
- School of Health SciencesPurdue UniversityIndianaWest LafayetteUSA
| | - Richard A. E. Edden
- Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- F. M. Kirby Research Center for Functional Brain ImagingKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Lars Ersland
- Department of Biological and Medical PsychologyUniversity of BergenBergenNorway
- Department of Clinical EngineeringHaukeland University HospitalBergenNorway
| | - Pravat K. Mandal
- NeuroImaging and NeuroSpectroscopy (NINS) Laboratory, National Brain Research CentreGurgaonIndia
- Florey Institute of Neuroscience and Mental HealthParkvilleMelbourneVictoriaAustralia
| | - Mark Mikkelsen
- Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- F. M. Kirby Research Center for Functional Brain ImagingKennedy Krieger InstituteBaltimoreMarylandUSA
- Department of RadiologyWeill Cornell MedicineNew YorkNew YorkUSA
| | | | - Jamie Near
- Centre d'Imagerie CérébraleDouglas Mental Health University InstituteMontrealCanada
- Department of Biomedical EngineeringMcGill UniversityMontrealCanada
- Department of PsychiatryMcGill UniversityMontrealCanada
| | - Reuben Rideaux
- Queensland Brain InstituteThe University of QueenslandBrisbaneAustralia
| | - Deepika Shukla
- NeuroImaging and NeuroSpectroscopy (NINS) Laboratory, National Brain Research CentreGurgaonIndia
- Perinatal Trials Unit FoundationBengaluruIndia
- Centre for Perinatal NeuroscienceImperial College LondonLondonUK
| | - Min Wang
- College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Martin Wilson
- Centre for Human Brain Health and School of PsychologyUniversity of BirminghamBirminghamUK
| | - Helge J. Zöllner
- Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- F. M. Kirby Research Center for Functional Brain ImagingKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Kenneth Hugdahl
- Department of Biological and Medical PsychologyUniversity of BergenBergenNorway
- Division of PsychiatryHaukeland University HospitalBergenNorway
- Department of RadiologyHaukeland University HospitalBergenNorway
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- F. M. Kirby Research Center for Functional Brain ImagingKennedy Krieger InstituteBaltimoreMarylandUSA
| |
Collapse
|
32
|
Increased cardiac Pi/PCr in the diabetic heart observed using phosphorus magnetic resonance spectroscopy at 7T. PLoS One 2022; 17:e0269957. [PMID: 35709167 PMCID: PMC9202907 DOI: 10.1371/journal.pone.0269957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/01/2022] [Indexed: 12/04/2022] Open
Abstract
Phosphorus magnetic resonance spectroscopy (31P-MRS) has previously demonstrated decreased energy reserves in the form of phosphocreatine to adenosine-tri-phosphate ratio (PCr/ATP) in the hearts of patients with type 2 diabetes (T2DM). Recent 31P-MRS techniques using 7T systems, e.g. long mixing time stimulated echo acquisition mode (STEAM), allow deeper insight into cardiac metabolism through assessment of inorganic phosphate (Pi) content and myocardial pH, which play pivotal roles in energy production in the heart. Therefore, we aimed to further explore the cardiac metabolic phenotype in T2DM using STEAM at 7T. Seventeen patients with T2DM and twenty-three healthy controls were recruited and their cardiac PCr/ATP, Pi/PCr and pH were assessed at 7T. Diastolic function of all patients with T2DM was assessed using echocardiography to investigate the relationship between diastolic dysfunction and cardiac metabolism. Mirroring the decreased PCr/ATP (1.70±0.31 vs. 2.07±0.39; p<0.01), the cardiac Pi/PCr was increased (0.13±0.07 vs. 0.10±0.03; p = 0.02) in T2DM patients in comparison to healthy controls. Myocardial pH was not significantly different between the groups (7.14±0.12 vs. 7.10±0.12; p = 0.31). There was a negative correlation between PCr/ATP and diastolic function (R2 = 0.33; p = 0.02) in T2DM. No correlation was observed between diastolic function and Pi/PCr and (R2 = 0.16; p = 0.21). In addition, we did not observe any correlation between cardiac PCr/ATP and Pi/PCr (p = 0.19). Using STEAM 31P-MRS at 7T we have for the first time explored Pi/PCr in the diabetic human heart and found it increased when compared to healthy controls. The lack of correlation between measured PCr/ATP and Pi/PCr suggests that independent mechanisms might contribute to these perturbations.
Collapse
|
33
|
Hansen K, Hansen ESS, Jespersen NRV, Bøtker HE, Pedersen M, Wang T, Laustsen C. Hyperpolarized
13
C
MRI Reveals Large Changes in Pyruvate Metabolism During Digestion in Snakes. Magn Reson Med 2022; 88:890-900. [PMID: 35426467 PMCID: PMC9321735 DOI: 10.1002/mrm.29239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 01/31/2022] [Accepted: 02/25/2022] [Indexed: 11/05/2022]
Abstract
Purpose Methods Results Conclusion
Collapse
Affiliation(s)
- Kasper Hansen
- Comparative Medicine Lab, Department of Clinical Medicine Aarhus University Aarhus Denmark
- Zoophysiology, Department of Biology Aarhus University Aarhus Denmark
- Department of Forensic Medicine Aarhus University Aarhus Denmark
| | | | | | - Hans Erik Bøtker
- Cardiology, Department of Clinical Medicine Aarhus University Aarhus Denmark
| | - Michael Pedersen
- Comparative Medicine Lab, Department of Clinical Medicine Aarhus University Aarhus Denmark
| | - Tobias Wang
- Zoophysiology, Department of Biology Aarhus University Aarhus Denmark
| | - Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine Aarhus University Aarhus Denmark
| |
Collapse
|
34
|
Brakedal B, Dölle C, Riemer F, Ma Y, Nido GS, Skeie GO, Craven AR, Schwarzlmüller T, Brekke N, Diab J, Sverkeli L, Skjeie V, Varhaug K, Tysnes OB, Peng S, Haugarvoll K, Ziegler M, Grüner R, Eidelberg D, Tzoulis C. The NADPARK study: A randomized phase I trial of nicotinamide riboside supplementation in Parkinson's disease. Cell Metab 2022; 34:396-407.e6. [PMID: 35235774 DOI: 10.1016/j.cmet.2022.02.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/17/2021] [Accepted: 01/31/2022] [Indexed: 02/07/2023]
Abstract
We conducted a double-blinded phase I clinical trial to establish whether nicotinamide adenine dinucleotide (NAD) replenishment therapy, via oral intake of nicotinamide riboside (NR), is safe, augments cerebral NAD levels, and impacts cerebral metabolism in Parkinson's disease (PD). Thirty newly diagnosed, treatment-naive patients received 1,000 mg NR or placebo for 30 days. NR treatment was well tolerated and led to a significant, but variable, increase in cerebral NAD levels-measured by 31phosphorous magnetic resonance spectroscopy-and related metabolites in the cerebrospinal fluid. NR recipients showing increased brain NAD levels exhibited altered cerebral metabolism, measured by 18fluoro-deoxyglucose positron emission tomography, and this was associated with mild clinical improvement. NR augmented the NAD metabolome and induced transcriptional upregulation of processes related to mitochondrial, lysosomal, and proteasomal function in blood cells and/or skeletal muscle. Furthermore, NR decreased the levels of inflammatory cytokines in serum and cerebrospinal fluid. Our findings nominate NR as a potential neuroprotective therapy for PD, warranting further investigation in larger trials.
Collapse
Affiliation(s)
- Brage Brakedal
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Christian Dölle
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Frank Riemer
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Yilong Ma
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Gonzalo S Nido
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Geir Olve Skeie
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Alexander R Craven
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway; Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway; Department of Clinical Engineering, Haukeland University Hospital, Bergen, Norway
| | - Thomas Schwarzlmüller
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Njål Brekke
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Joseph Diab
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Lars Sverkeli
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Vivian Skjeie
- Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Kristin Varhaug
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Ole-Bjørn Tysnes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Shichun Peng
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Kristoffer Haugarvoll
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Mathias Ziegler
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Renate Grüner
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - David Eidelberg
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Charalampos Tzoulis
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
35
|
Cameron D, Soto-Mota A, Willis DR, Ellis J, Procter NEK, Greenwood R, Saunders N, Schulte RF, Vassiliou VS, Tyler DJ, Schmid AI, Rodgers CT, Malcolm PN, Clarke K, Frenneaux MP, Valkovič L. Evaluation of Acute Supplementation With the Ketone Ester (R)-3-Hydroxybutyl-(R)-3-Hydroxybutyrate (deltaG) in Healthy Volunteers by Cardiac and Skeletal Muscle 31P Magnetic Resonance Spectroscopy. Front Physiol 2022; 13:793987. [PMID: 35173629 PMCID: PMC8841822 DOI: 10.3389/fphys.2022.793987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/05/2022] [Indexed: 01/11/2023] Open
Abstract
In this acute intervention study, we investigated the potential benefit of ketone supplementation in humans by studying cardiac phosphocreatine to adenosine-triphosphate ratios (PCr/ATP) and skeletal muscle PCr recovery using phosphorus magnetic resonance spectroscopy (31P-MRS) before and after ingestion of a ketone ester drink. We recruited 28 healthy individuals: 12 aged 23–70 years for cardiac 31P-MRS, and 16 aged 60–75 years for skeletal muscle 31P-MRS. Baseline and post-intervention resting cardiac and dynamic skeletal muscle 31P-MRS scans were performed in one visit, where 25 g of the ketone monoester, deltaG®, was administered after the baseline scan. Administration was timed so that post-intervention 31P-MRS would take place 30 min after deltaG® ingestion. The deltaG® ketone drink was well-tolerated by all participants. In participants who provided blood samples, post-intervention blood glucose, lactate and non-esterified fatty acid concentrations decreased significantly (−28.8%, p ≪ 0.001; −28.2%, p = 0.02; and −49.1%, p ≪ 0.001, respectively), while levels of the ketone body D-beta-hydroxybutyrate significantly increased from mean (standard deviation) 0.7 (0.3) to 4.0 (1.1) mmol/L after 30 min (p ≪ 0.001). There were no significant changes in cardiac PCr/ATP or skeletal muscle metabolic parameters between baseline and post-intervention. Acute ketone supplementation caused mild ketosis in blood, with drops in glucose, lactate, and free fatty acids; however, such changes were not associated with changes in 31P-MRS measures in the heart or in skeletal muscle. Future work may focus on the effect of longer-term ketone supplementation on tissue energetics in groups with compromised mitochondrial function.
Collapse
Affiliation(s)
- Donnie Cameron
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
- Department of Radiology, C.J. Gorter Center for High-Field MRI, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Donnie Cameron,
| | - Adrian Soto-Mota
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - David R. Willis
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Jane Ellis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
| | | | - Richard Greenwood
- Radiology Department, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Neil Saunders
- Radiology Department, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | | | | | - Damian J. Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
| | - Albrecht Ingo Schmid
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
- High Field MR Center, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Christopher T. Rodgers
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
- Department of Clinical Neurosciences, Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Paul N. Malcolm
- Radiology Department, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Ladislav Valkovič
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
36
|
Watson WD, Green PG, Valkovič L, Herring N, Neubauer S, Rider OJ. Myocardial Energy Response to Glyceryl Trinitrate: Physiology Revisited. Front Physiol 2021; 12:790525. [PMID: 35035360 PMCID: PMC8758569 DOI: 10.3389/fphys.2021.790525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/25/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Although intravenous nitrates are commonly used in clinical medicine, they have been shown to increase myocardial oxygen consumption and inhibit complex IV of the electron transport chain. As such we sought to measure whether myocardial energetics were impaired during glyceryl trinitrate (GTN) infusion. Methods: 10 healthy volunteers underwent cardiac magnetic resonance imaging to assess cardiac function and 31phosphorus magnetic resonance spectroscopy to measure Phosphocreatine/ATP (PCr/ATP) ratio and creatine kinase forward rate constant (CK kf ) before and during an intravenous infusion of GTN. Results: During GTN infusion, mean arterial pressure (78 ± 7 vs. 65 ± 6 mmHg, p < 0.001), left ventricular (LV) stroke work (7,708 ± 2,782 vs. 6,071 ± 2,660 ml mmHg, p < 0.001), and rate pressure product (7,214 ± 1,051 vs. 6,929 ± 976 mmHg bpm, p = 0.06) all fell. LV ejection fraction increased (61 ± 3 vs. 66 ± 4%, p < 0.001), with cardiac output remaining constant (6.2 ± 1.5 vs. 6.5 ± 1.4 l/min, p = 0.37). Myocardial PCr/ATP fell during GTN infusion (2.17 ± 0.2 vs. 1.99 ± 0.22, p = 0.03) with an increase in both CK kf (0.16 ± 0.07 vs. 0.25 ± 0.1 s-1, p = 0.006) and CK flux (1.8 ± 0.8 vs. 2.6 ± 1.1 μmol/g/s, p = 0.03). Conclusion: During GTN infusion, despite reduced LV stroke work and maintained cardiac output, there was a 44% increase in myocardial ATP delivery through CK. As PCr/ATP fell, this increase in ATP demand coincided with GTN-induced impairment of mitochondrial oxidative phosphorylation. Overall, this suggests that while GTN reduces cardiac work, it does so at the expense of increasing ATP demand beyond the capacity to increase ATP production.
Collapse
Affiliation(s)
- William D. Watson
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
| | - Peregrine G. Green
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
- Department for Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Ladislav Valkovič
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Neil Herring
- Department for Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Stefan Neubauer
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
| | - Oliver J. Rider
- Oxford Centre for Magnetic Resonance Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
37
|
Tan JL, Djayakarsana D, Wang H, Chan RW, Bailey C, Lau AZ. Deuterium MRS of early treatment-induced changes in tumour lactate in vitro. NMR IN BIOMEDICINE 2021; 34:e4599. [PMID: 34405471 DOI: 10.1002/nbm.4599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 06/13/2023]
Abstract
Elevated production of lactate is a key characteristic of aberrant tumour cell metabolism and can be non-invasively measured as an early marker of tumour response using deuterium (2 H) MRS. Following treatment, changes in the 2 H-labelled lactate signal could identify tumour cell death or impaired metabolic function, which precede morphological changes conventionally used to assess tumour response. In this work, the association between apoptotic cell death, extracellular lactate concentration, and early treatment-induced changes in the 2 H-labelled lactate signal was established in an in vitro tumour model. Experiments were conducted at 7 T on acute myeloid leukaemia (AML) cells, which had been treated with 10 μg/mL of the chemotherapeutic agent cisplatin. At 24 and 48 h after cisplatin treatment the cells were supplied with 20 mM of [6,6'-2 H2 ]glucose and scanned over 2 h using a two-dimensional 2 H MR spectroscopic imaging sequence. The resulting signals from 2 H-labelled glucose, lactate, and water were quantified using a spectral fitting algorithm implemented on the Oxford Spectroscopy Analysis MATLAB toolbox. After scanning, the cells were processed for histological stains (terminal deoxynucleotidyl transferase UTP nick end labelling and haematoxylin and eosin) to assess apoptotic area fraction and cell morphology respectively, while a colorimetric assay was used to measure extracellular lactate concentrations in the supernatant. Significantly lower levels of 2 H-labelled lactate were observed in the 48 h treated cells compared with the untreated and 24 h treated cells, and these changes were significantly correlated with an increase in apoptotic fraction and a decrease in extracellular lactate. By establishing the biological processes associated with treatment-induced changes in the 2 H-labelled lactate signal, these findings suggest that 2 H MRS of lactate may be valuable in evaluating early tumour response.
Collapse
Affiliation(s)
- Josephine L Tan
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Daniel Djayakarsana
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Hanzhi Wang
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Rachel W Chan
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Colleen Bailey
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Physics, Ryerson University, Toronto, Canada
| | - Angus Z Lau
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| |
Collapse
|
38
|
Young LAJ, Ceresa CDL, Mózes FE, Ellis J, Valkovič L, Colling R, Coussios CC, Friend PJ, Rodgers CT. Noninvasive assessment of steatosis and viability of cold-stored human liver grafts by MRI. Magn Reson Med 2021; 86:3246-3258. [PMID: 34272767 PMCID: PMC7613197 DOI: 10.1002/mrm.28930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/02/2022]
Abstract
PURPOSE A shortage of suitable donor livers is driving increased use of higher risk livers for transplantation. However, current biomarkers are not sensitive and specific enough to predict posttransplant liver function. This is limiting the expansion of the donor pool. Therefore, better noninvasive tests are required to determine which livers will function following implantation and hence can be safely transplanted. This study assesses the temperature sensitivity of proton density fat fraction and relaxometry parameters and examines their potential for assessment of liver function ex vivo. METHODS Six ex vivo human livers were scanned during static cold storage following normothermic machine perfusion. Proton density fat fraction, T1 , T2 , and T 2 ∗ were measured repeatedly during cooling on ice. Temperature corrections were derived from these measurements for the parameters that showed significant variation with temperature. RESULTS Strong linear temperature sensitivities were observed for proton density fat fraction (R2 = 0.61, P < .001) and T1 (R2 = 0.78, P < .001). Temperature correction according to a linear model reduced the coefficient of repeatability in these measurements by 41% and 36%, respectively. No temperature dependence was observed in T2 or T 2 ∗ measurements. Comparing livers deemed functional and nonfunctional during normothermic machine perfusion by hemodynamic and biochemical criteria, T1 differed significantly: 516 ± 50 ms for functional versus 679 ± 60 ms for nonfunctional, P = .02. CONCLUSION Temperature correction is essential for robust measurement of proton density fat fraction and T1 in cold-stored human livers. These parameters may provide a noninvasive measure of viability for transplantation.
Collapse
Affiliation(s)
- Liam A. J. Young
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Carlo D. L. Ceresa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Ferenc E. Mózes
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jane Ellis
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Richard Colling
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | | | - Peter J. Friend
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Christopher T. Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
39
|
Burrage MK, Hundertmark M, Valkovič L, Watson WD, Rayner J, Sabharwal N, Ferreira VM, Neubauer S, Miller JJ, Rider OJ, Lewis AJ. Energetic Basis for Exercise-Induced Pulmonary Congestion in Heart Failure With Preserved Ejection Fraction. Circulation 2021; 144:1664-1678. [PMID: 34743560 PMCID: PMC8601674 DOI: 10.1161/circulationaha.121.054858] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/01/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Transient pulmonary congestion during exercise is emerging as an important determinant of reduced exercise capacity in heart failure with preserved ejection fraction (HFpEF). We sought to determine whether an abnormal cardiac energetic state underpins this process. METHODS We recruited patients across the spectrum of diastolic dysfunction and HFpEF (controls, n=11; type 2 diabetes, n=9; HFpEF, n=14; and severe diastolic dysfunction attributable to cardiac amyloidosis, n=9). Cardiac energetics were measured using phosphorus spectroscopy to define the myocardial phosphocreatine to ATP ratio. Cardiac function was assessed by cardiovascular magnetic resonance cine imaging and echocardiography and lung water using magnetic resonance proton density mapping. Studies were performed at rest and during submaximal exercise using a magnetic resonance imaging ergometer. RESULTS Paralleling the stepwise decline in diastolic function across the groups (E/e' ratio; P<0.001) was an increase in NT-proBNP (N-terminal pro-brain natriuretic peptide; P<0.001) and a reduction in phosphocreatine/ATP ratio (control, 2.15 [2.09, 2.29]; type 2 diabetes, 1.71 [1.61, 1.91]; HFpEF, 1.66 [1.44, 1.89]; cardiac amyloidosis, 1.30 [1.16, 1.53]; P<0.001). During 20-W exercise, lower left ventricular diastolic filling rates (r=0.58; P<0.001), lower left ventricular diastolic reserve (r=0.55; P<0.001), left atrial dilatation (r=-0.52; P<0.001), lower right ventricular contractile reserve (right ventricular ejection fraction change, r=0.57; P<0.001), and right atrial dilation (r=-0.71; P<0.001) were all linked to lower phosphocreatine/ATP ratio. Along with these changes, pulmonary proton density mapping revealed transient pulmonary congestion in patients with HFpEF (+4.4% [0.5, 6.4]; P=0.002) and cardiac amyloidosis (+6.4% [3.3, 10.0]; P=0.004), which was not seen in healthy controls (-0.1% [-1.9, 2.1]; P=0.89) or type 2 diabetes without HFpEF (+0.8% [-1.7, 1.9]; P=0.82). The development of exercise-induced pulmonary congestion was associated with lower phosphocreatine/ATP ratio (r=-0.43; P=0.004). CONCLUSIONS A gradient of myocardial energetic deficit exists across the spectrum of HFpEF. Even at low workload, this energetic deficit is related to markedly abnormal exercise responses in all 4 cardiac chambers, which is associated with detectable pulmonary congestion. The findings support an energetic basis for transient pulmonary congestion in HFpEF.
Collapse
Affiliation(s)
- Matthew K. Burrage
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
| | - Moritz Hundertmark
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
| | - Ladislav Valkovič
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia (L.V.)
| | - William D. Watson
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
| | - Jennifer Rayner
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
- Department of Cardiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, UK (J.R., N.S., S.N., O.J.R., A.J.M.L.)
| | - Nikant Sabharwal
- Department of Cardiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, UK (J.R., N.S., S.N., O.J.R., A.J.M.L.)
| | - Vanessa M. Ferreira
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
| | - Stefan Neubauer
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
- Department of Cardiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, UK (J.R., N.S., S.N., O.J.R., A.J.M.L.)
| | - Jack J. Miller
- Department of Physics, Clarendon Laboratory (J.J.M.), University of Oxford, UK
- The MR Research Centre and The PET Research Centre, Aarhus University, Denmark (J.J.M.)
| | - Oliver J. Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
- Department of Cardiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, UK (J.R., N.S., S.N., O.J.R., A.J.M.L.)
| | - Andrew J.M. Lewis
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine (M.K.B., M.H., L.V., W.D.W., J.R., V.M.F., S.N., O.J.R., A.J.M.L.), University of Oxford, UK
- Department of Cardiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, UK (J.R., N.S., S.N., O.J.R., A.J.M.L.)
| |
Collapse
|
40
|
Rayner JJ, Peterzan MA, Clarke WT, Rodgers CT, Neubauer S, Rider OJ. Obesity modifies the energetic phenotype of dilated cardiomyopathy. Eur Heart J 2021; 43:ehab663. [PMID: 34542592 PMCID: PMC8885325 DOI: 10.1093/eurheartj/ehab663] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/13/2021] [Accepted: 09/13/2021] [Indexed: 01/05/2023] Open
Abstract
AIMS We sought to determine if myocardial energetics could distinguish obesity cardiomyopathy as a distinct entity from dilated cardiomyopathy. METHODS AND RESULTS Sixteen normal weight participants with dilated cardiomyopathy (DCMNW), and 27 with DCM and obesity (DCMOB), were compared to 26 normal weight controls (CTLNW). All underwent cardiac magnetic resonance imaging and 31P spectroscopy to assess function and energetics. Nineteen DCMOB underwent repeat assessment after a dietary weight loss intervention. Adenosine triphosphate (ATP) delivery through creatine kinase (CK flux) was 55% lower in DCMNW than in CTLNW (P = 0.004), correlating with left ventricular ejection fraction (LVEF, r = 0.4, P = 0.015). In contrast, despite similar LVEF (DCMOB 41 ± 7%, DCMNW 38 ± 6%, P = 0.14), CK flux was two-fold higher in DCMOB (P < 0.001), due to higher rate through CK [median kf 0.21 (0.14) vs. 0.11 (0.12) s-1, P = 0.002]. During increased workload, the CTLNW heart increased CK flux by 97% (P < 0.001). In contrast, CK flux was unchanged in DCMNW and fell in DCMOB (by >50%, P < 0.001). Intentional weight loss was associated with positive left ventricular remodelling, with reduced left ventricular end-diastolic volume (by 8%, P < 0.001) and a change in LVEF (40 ± 9% vs. 45 ± 10%, P = 0.002). This occurred alongside a fall in ATP delivery rate with weight loss (by 7%, P = 0.049). CONCLUSIONS In normal weight, DCM is associated with reduced resting ATP delivery. In obese DCM, ATP demand through CK is greater, suggesting reduced efficiency of energy utilization. Dietary weight loss is associated with significant improvement in myocardial contractility, and a fall in ATP delivery, suggesting improved metabolic efficiency. This highlights distinct energetic pathways in obesity cardiomyopathy, which are both different from dilated cardiomyopathy, and may be reversible with weight loss.
Collapse
Affiliation(s)
- Jennifer J Rayner
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Level 0, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Mark A Peterzan
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Level 0, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - William T Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Christopher T Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Level 0, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Box 65, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Level 0, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Oliver J Rider
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Level 0, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
41
|
Kerr M, Dennis KMJH, Carr CA, Fuller W, Berridge G, Rohling S, Aitken CL, Lopez C, Fischer R, Miller JJ, Clarke K, Tyler DJ, Heather LC. Diabetic mitochondria are resistant to palmitoyl CoA inhibition of respiration, which is detrimental during ischemia. FASEB J 2021; 35:e21765. [PMID: 34318967 PMCID: PMC8662312 DOI: 10.1096/fj.202100394r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/24/2021] [Accepted: 06/14/2021] [Indexed: 01/07/2023]
Abstract
The bioactive lipid intermediate palmitoyl CoA (PCoA) can inhibit mitochondrial ADP/ATP transport, though the physiological relevance of this regulation remains unclear. We questioned whether myocardial ischemia provides a pathological setting in which PCoA regulation of ADP/ATP transport would be beneficial, and secondly, whether the chronically elevated lipid content within the diabetic heart could make mitochondria less sensitive to the effects of PCoA. PCoA acutely decreased ADP‐stimulated state 3 respiration and increased the apparent Km for ADP twofold. The half maximal inhibitory concentration (IC50) of PCoA in control mitochondria was 22 µM. This inhibitory effect of PCoA on respiration was blunted in diabetic mitochondria, with no significant difference in the Km for ADP in the presence of PCoA, and an increase in the IC50 to 32 µM PCoA. The competitive inhibition by PCoA was localised to the phosphorylation apparatus, particularly the ADP/ATP carrier (AAC). During ischemia, the AAC imports ATP into the mitochondria, where it is hydrolysed by reversal of the ATP synthase, regenerating the membrane potential. Addition of PCoA dose‐dependently prevented this wasteful ATP hydrolysis for membrane repolarisation during ischemia, however, this beneficial effect was blunted in diabetic mitochondria. Finally, using 31P‐magnetic resonance spectroscopy we demonstrated that diabetic hearts lose ATP more rapidly during ischemia, with a threefold higher ATP decay rate compared with control hearts. In conclusion, PCoA plays a role in protecting mitochondrial energetics during ischemia, by preventing wasteful ATP hydrolysis. However, this beneficial effect is blunted in diabetes, contributing to the impaired energy metabolism seen during myocardial ischemia in the diabetic heart.
Collapse
Affiliation(s)
- M Kerr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - K M J H Dennis
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - C A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - W Fuller
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - G Berridge
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - S Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - C L Aitken
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - C Lopez
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - R Fischer
- Target Discovery Institute, University of Oxford, Oxford, UK
| | - J J Miller
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Department of Physics, University of Oxford, Oxford, UK.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - K Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - D J Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - L C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
42
|
Ding B, Peterzan M, Mózes FE, Rider OJ, Valkovič L, Rodgers CT. Water-suppression cycling 3-T cardiac 1 H-MRS detects altered creatine and choline in patients with aortic or mitral stenosis. NMR IN BIOMEDICINE 2021; 34:e4513. [PMID: 33826181 PMCID: PMC8243349 DOI: 10.1002/nbm.4513] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/23/2021] [Accepted: 03/03/2021] [Indexed: 05/06/2023]
Abstract
Cardiac proton spectroscopy (1 H-MRS) is widely used to quantify lipids. Other metabolites (e.g. creatine and choline) are clinically relevant but more challenging to quantify because of their low concentrations (approximately 10 mmol/L) and because of cardiac motion. To quantify cardiac creatine and choline, we added water-suppression cycling (WSC) to two single-voxel spectroscopy sequences (STEAM and PRESS). WSC introduces controlled residual water signals that alternate between positive and negative phases from transient to transient, enabling robust phase and frequency correction. Moreover, a particular weighted sum of transients eliminates residual water signals without baseline distortion. We compared WSC and the vendor's standard 'WET' water suppression in phantoms. Next, we tested repeatability in 10 volunteers (seven males, three females; age 29.3 ± 4.0 years; body mass index [BMI] 23.7 ± 4.1 kg/m2 ). Fat fraction, creatine concentration and choline concentration when quantified by STEAM-WET were 0.30% ± 0.11%, 29.6 ± 7.0 μmol/g and 7.9 ± 6.7 μmol/g, respectively; and when quantified by PRESS-WSC they were 0.30% ± 0.15%, 31.5 ± 3.1 μmol/g and 8.3 ± 4.4 μmol/g, respectively. Compared with STEAM-WET, PRESS-WSC gave spectra whose fitting quality expressed by Cramér-Rao lower bounds improved by 26% for creatine and 32% for choline. Repeatability of metabolite concentration measurements improved by 72% for creatine and 40% for choline. We also compared STEAM-WET and PRESS-WSC in 13 patients with severe symptomatic aortic or mitral stenosis indicated for valve replacement surgery (10 males, three females; age 75.9 ± 6.3 years; BMI 27.4 ± 4.3 kg/m2 ). Spectra were of analysable quality in eight patients for STEAM-WET, and in nine for PRESS-WSC. We observed comparable lipid concentrations with those in healthy volunteers, significantly reduced creatine concentrations, and a trend towards decreased choline concentrations. We conclude that PRESS-WSC offers improved performance and reproducibility for the quantification of cardiac lipids, creatine and choline concentrations in healthy volunteers at 3 T. It also offers improved performance compared with STEAM-WET for detecting altered creatine and choline concentrations in patients with valve disease.
Collapse
Affiliation(s)
- Belinda Ding
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR)University of OxfordOxfordUK
| | - Mark Peterzan
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR)University of OxfordOxfordUK
| | - Ferenc E. Mózes
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR)University of OxfordOxfordUK
| | - Oliver J. Rider
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR)University of OxfordOxfordUK
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR)University of OxfordOxfordUK
- Department of Imaging Methods, Institute of Measurement ScienceSlovak Academy of SciencesBratislavaSlovakia
| | - Christopher T. Rodgers
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR)University of OxfordOxfordUK
| |
Collapse
|
43
|
Jayaswal ANA, Levick C, Collier J, Tunnicliffe EM, Kelly MD, Neubauer S, Barnes E, Pavlides M. Liver cT 1 decreases following direct-acting antiviral therapy in patients with chronic hepatitis C virus. Abdom Radiol (NY) 2021; 46:1947-1957. [PMID: 33247768 PMCID: PMC8131342 DOI: 10.1007/s00261-020-02860-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/04/2020] [Accepted: 11/07/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Direct-acting antiviral therapies (DAAs) for treatment of chronic hepatitis C virus (HCV) have excellent rates of viral eradication, but their effect on regression of liver fibrosis is unclear. The primary aim was to use magnetic resonance imaging (MRI) and spectroscopy (MRS) to evaluate changes in liver fibrosis, liver fat and liver iron content (LIC) in patients with chronic HCV following treatment with DAAs. METHODS In this prospective study, 15 patients with chronic HCV due to start treatment with DAAs and with transient elastography (TE) > 8 kPa were recruited consecutively. Patients underwent MRI and MRS at baseline (before treatment), and at 24 weeks and 48 weeks after the end of treatment (EoT) for the measurement of liver cT1 (fibroinflammation), liver fat and T2* (LIC). RESULTS All patients achieved a sustained virological response. Liver cT1 showed significant decreases from baseline to 24 weeks post EoT (876 vs 806 ms, p = 0.002, n = 15), baseline to 48 weeks post EoT (876 vs 788 ms, p = 0.0002, n = 13) and 24 weeks post EoT to 48 weeks post EoT (806 vs 788 ms, p = 0.016, n = 13). Between baseline and 48 weeks EoT significant reduction in liver fat (5.17% vs 2.65%, p = 0.027) and an increase in reported LIC (0.913 vs 0.950 mg/g, p = 0.021) was observed. CONCLUSION Liver cT1 decreases in patients with chronic HCV undergoing successful DAA treatment. The relatively fast reduction in cT1 suggests a reduction in inflammation rather than regression of fibrosis.
Collapse
Affiliation(s)
- Arjun N A Jayaswal
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Christina Levick
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Jane Collier
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Elizabeth M Tunnicliffe
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford, UK
| | | | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford, UK
| | - Eleanor Barnes
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford, UK
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Michael Pavlides
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
- Oxford NIHR Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
44
|
Wampl S, Körner T, Valkovič L, Trattnig S, Wolzt M, Meyerspeer M, Schmid AI. Investigating the effect of trigger delay on cardiac 31P MRS signals. Sci Rep 2021; 11:9268. [PMID: 33927234 PMCID: PMC8085231 DOI: 10.1038/s41598-021-87063-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/22/2021] [Indexed: 01/15/2023] Open
Abstract
The heart’s geometry and its metabolic activity vary over the cardiac cycle. The effect of these fluctuations on phosphorus (31P) magnetic resonance spectroscopy (MRS) data quality and metabolite ratios was investigated. 12 healthy volunteers were measured using a 7 T MR scanner and a cardiac 31P-1H loop coil. 31P chemical shift imaging data were acquired untriggered and at four different times during the cardiac cycle using acoustic triggering. Signals of adenosine-triphosphate (ATP), phosphocreatine (PCr), inorganic phosphate (Pi) and 2,3-diphosphoglycerate (2,3-DPG) and their fit quality as Cramér-Rao lower bounds (CRLB) were quantified including corrections for contamination by 31P signals from blood, flip angle, saturation and total acquisition time. The myocardial filling factor was estimated from cine short axis views. The corrected signals of PCr and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\gamma$$\end{document}γ-ATP were higher during end-systole and lower during diastasis than in untriggered acquisitions (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P<0.05$$\end{document}P<0.05). Signal intensities of untriggered scans were between those with triggering to end-systole and diastasis. Fit quality of PCr and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\gamma$$\end{document}γ-ATP peaks was best during end-systole when blood contamination of ATP and Pi signals was lowest. While metabolite ratios and pH remained stable over the cardiac cycle, signal amplitudes correlated strongly with myocardial voxel filling. Triggering of cardiac 31P MRS acquisitions improves signal amplitudes and fit quality if the trigger delay is set to end-systole. We conclude that triggering to end-systole is superior to triggering to diastasis.
Collapse
Affiliation(s)
- Stefan Wampl
- Medical University of Vienna, High Field MR Center, Center for Medical Physics and Biomedical Engineering, Vienna, 1090, Austria
| | - Tito Körner
- Medical University of Vienna, High Field MR Center, Center for Medical Physics and Biomedical Engineering, Vienna, 1090, Austria
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom.,Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, 814 04, Slovakia
| | - Siegfried Trattnig
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, High Field MR Center, Vienna, 1090, Austria
| | - Michael Wolzt
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, 1090, Austria
| | - Martin Meyerspeer
- Medical University of Vienna, High Field MR Center, Center for Medical Physics and Biomedical Engineering, Vienna, 1090, Austria
| | - Albrecht Ingo Schmid
- Medical University of Vienna, High Field MR Center, Center for Medical Physics and Biomedical Engineering, Vienna, 1090, Austria.
| |
Collapse
|
45
|
Vaeggemose M, F. Schulte R, Laustsen C. Comprehensive Literature Review of Hyperpolarized Carbon-13 MRI: The Road to Clinical Application. Metabolites 2021; 11:metabo11040219. [PMID: 33916803 PMCID: PMC8067176 DOI: 10.3390/metabo11040219] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 01/02/2023] Open
Abstract
This review provides a comprehensive assessment of the development of hyperpolarized (HP) carbon-13 metabolic MRI from the early days to the present with a focus on clinical applications. The status and upcoming challenges of translating HP carbon-13 into clinical application are reviewed, along with the complexity, technical advancements, and future directions. The road to clinical application is discussed regarding clinical needs and technological advancements, highlighting the most recent successes of metabolic imaging with hyperpolarized carbon-13 MRI. Given the current state of hyperpolarized carbon-13 MRI, the conclusion of this review is that the workflow for hyperpolarized carbon-13 MRI is the limiting factor.
Collapse
Affiliation(s)
- Michael Vaeggemose
- GE Healthcare, 2605 Brondby, Denmark;
- MR Research Centre, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | | | - Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
- Correspondence:
| |
Collapse
|
46
|
Abstract
Magnetic resonance spectroscopy (MRS), being able to identify and measure some brain components (metabolites) in pathologic lesions and in normal-appearing tissue, offers a valuable additional diagnostic tool to assess several pediatric neurological diseases. In this review we will illustrate the basic principles and clinical applications of brain proton (H1; hydrogen) MRS (H1MRS), by now the only MRS method widely available in clinical practice. Performing H1MRS in the brain is inherently less complicated than in other tissues (e.g., liver, muscle), in which spectra are heavily affected by magnetic field inhomogeneities, respiration artifacts, and dominating signals from the surrounding adipose tissues. H1MRS in pediatric neuroradiology has some advantages over acquisitions in adults (lack of motion due to children sedation and lack of brain iron deposition allow optimal results), but it requires a deep knowledge of pediatric pathologies and familiarity with the developmental changes in spectral patterns, particularly occurring in the first two years of life. Examples from our database, obtained mainly from a 1.5 Tesla clinical scanner in a time span of 15 years, will demonstrate the efficacy of H1MRS in the diagnosis of a wide range of selected pediatric pathologies, like brain tumors, infections, neonatal hypoxic-ischemic encephalopathy, metabolic and white matter disorders.
Collapse
Affiliation(s)
- Roberto Liserre
- Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy
| | - Lorenzo Pinelli
- Department of Radiology, Neuroradiology Unit, ASST Spedali Civili University Hospital, Brescia, Italy
| | - Roberto Gasparotti
- Neuroradiology Unit, Department of Medical-Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| |
Collapse
|
47
|
Spartera M, Pessoa-Amorim G, Stracquadanio A, Von Ende A, Fletcher A, Manley P, Neubauer S, Ferreira VM, Casadei B, Hess AT, Wijesurendra RS. Left atrial 4D flow cardiovascular magnetic resonance: a reproducibility study in sinus rhythm and atrial fibrillation. J Cardiovasc Magn Reson 2021; 23:29. [PMID: 33745457 PMCID: PMC7983287 DOI: 10.1186/s12968-021-00729-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 02/03/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Four-dimensional (4D) flow cardiovascular magnetic resonance (CMR) allows sophisticated quantification of left atrial (LA) blood flow, and could yield novel biomarkers of propensity for intra-cardiac thrombus formation and embolic stroke. As reproducibility is critically important to diagnostic performance, we systematically investigated technical and temporal variation of LA 4D flow in atrial fibrillation (AF) and sinus rhythm (SR). METHODS Eighty-six subjects (SR, n = 64; AF, n = 22) with wide-ranging stroke risk (CHA2DS2VASc 0-6) underwent LA 4D flow assessment of peak and mean velocity, vorticity, vortex volume, and stasis. Eighty-five (99%) underwent a second acquisition within the same session, and 74 (86%) also returned at 30 (27-35) days for an interval scan. We assessed variability attributable to manual contouring (intra- and inter-observer), and subject repositioning and reacquisition of data, both within the same session (same-day scan-rescan), and over time (interval scan). Within-subject coefficients of variation (CV) and bootstrapped 95% CIs were calculated and compared. RESULTS Same-day scan-rescan CVs were 6% for peak velocity, 5% for mean velocity, 7% for vorticity, 9% for vortex volume, and 10% for stasis, and were similar between SR and AF subjects (all p > 0.05). Interval-scan variability was similar to same-day scan-rescan variability for peak velocity, vorticity, and vortex volume (all p > 0.05), and higher for stasis and mean velocity (interval scan CVs of 14% and 8%, respectively, both p < 0.05). Longitudinal changes in heart rate and blood pressure at the interval scan in the same subjects were associated with significantly higher variability for LA stasis (p = 0.024), but not for the remaining flow parameters (all p > 0.05). SR subjects showed significantly greater interval-scan variability than AF patients for mean velocity, vortex volume, and stasis (all p < 0.05), but not peak velocity or vorticity (both p > 0.05). CONCLUSIONS LA peak velocity and vorticity are the most reproducible and temporally stable novel LA 4D flow biomarkers, and are robust to changes in heart rate, blood pressure, and differences in heart rhythm.
Collapse
Affiliation(s)
- Marco Spartera
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, West Wing, Headley Way, Oxford, UK.
- The University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Oxford, UK.
| | - Guilherme Pessoa-Amorim
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, West Wing, Headley Way, Oxford, UK
- The University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Oxford, UK
| | - Antonio Stracquadanio
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, West Wing, Headley Way, Oxford, UK
- The University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Oxford, UK
| | - Adam Von Ende
- Department of Population Health, CTSU Nuffield University of Oxford, Oxford, UK
| | - Alison Fletcher
- The University of Oxford Acute Vascular Imaging Centre (AVIC), Oxford, UK
| | - Peter Manley
- The University of Oxford Acute Vascular Imaging Centre (AVIC), Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, West Wing, Headley Way, Oxford, UK
- The University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Oxford, UK
| | - Vanessa M Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, West Wing, Headley Way, Oxford, UK
- The University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Oxford, UK
| | - Barbara Casadei
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, West Wing, Headley Way, Oxford, UK
| | - Aaron T Hess
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, West Wing, Headley Way, Oxford, UK
- The University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Oxford, UK
| | - Rohan S Wijesurendra
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, West Wing, Headley Way, Oxford, UK
- The University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Oxford, UK
| |
Collapse
|
48
|
Miller JJ, Valkovič L, Kerr M, Timm KN, Watson WD, Lau JYC, Tyler A, Rodgers C, Bottomley PA, Heather LC, Tyler DJ. Rapid, B 1 -insensitive, dual-band quasi-adiabatic saturation transfer with optimal control for complete quantification of myocardial ATP flux. Magn Reson Med 2021; 85:2978-2991. [PMID: 33538063 PMCID: PMC7986077 DOI: 10.1002/mrm.28647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Phosphorus saturation-transfer experiments can quantify metabolic fluxes noninvasively. Typically, the forward flux through the creatine kinase reaction is investigated by observing the decrease in phosphocreatine (PCr) after saturation of γ-ATP. The quantification of total ATP utilization is currently underexplored, as it requires simultaneous saturation of inorganic phosphate ( P i ) and PCr. This is challenging, as currently available saturation pulses reduce the already-low γ-ATP signal present. METHODS Using a hybrid optimal-control and Shinnar-Le Roux method, a quasi-adiabatic RF pulse was designed for the dual saturation of PCr and P i to enable determination of total ATP utilization. The pulses were evaluated in Bloch equation simulations, compared with a conventional hard-cosine DANTE saturation sequence, before being applied to perfused rat hearts at 11.7 T. RESULTS The quasi-adiabatic pulse was insensitive to a >2.5-fold variation in B 1 , producing equivalent saturation with a 53% reduction in delivered pulse power and a 33-fold reduction in spillover at the minimum effective B 1 . This enabled the complete quantification of the synthesis and degradation fluxes for ATP in 30-45 minutes in the perfused rat heart. While the net synthesis flux (4.24 ± 0.8 mM/s, SEM) was not significantly different from degradation flux (6.88 ± 2 mM/s, P = .06) and both measures are consistent with prior work, nonlinear error analysis highlights uncertainties in the Pi -to-ATP measurement that may explain a trend suggesting a possible imbalance. CONCLUSIONS This work demonstrates a novel quasi-adiabatic dual-saturation RF pulse with significantly improved performance that can be used to measure ATP turnover in the heart in vivo.
Collapse
Affiliation(s)
- Jack J Miller
- Department of Physics, University of Oxford, Oxford, UK.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Headington, Oxford, UK.,Health, Aarhus University, Aarhus, Denmark
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Headington, Oxford, UK.,Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Matthew Kerr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Kerstin N Timm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - William D Watson
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Headington, Oxford, UK
| | - Justin Y C Lau
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Headington, Oxford, UK
| | - Andrew Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Headington, Oxford, UK
| | - Christopher Rodgers
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Headington, Oxford, UK.,Wolfson Brain Imaging Centre, University of Cambridge, Oxford, UK
| | - Paul A Bottomley
- Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Headington, Oxford, UK.,Division of MR Research, Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Damian J Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.,Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Headington, Oxford, UK
| |
Collapse
|
49
|
Watson WD, Timm KN, Lewis AJ, Miller JJJ, Emmanuel Y, Clarke K, Neubauer S, Tyler DJ, Rider OJ. Nicotinic acid receptor agonists impair myocardial contractility by energy starvation. FASEB J 2020; 34:14878-14891. [PMID: 32954525 DOI: 10.1096/fj.202000084rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 08/08/2020] [Accepted: 08/25/2020] [Indexed: 11/11/2022]
Abstract
Nicotinic acid receptor agonists have previously been shown to cause acute reductions in cardiac contractility. We sought to uncover the changes in cardiac metabolism underlying these alterations in function. In nine humans, we recorded cardiac energetics and function before and after a single oral dose of nicotinic acid using cardiac MRI to demonstrate contractile function and Phosphorus-31 (31 P) magnetic resonance spectroscopy to demonstrate myocardial energetics. Nicotinic Acid 400 mg lowered ejection fraction by 4% (64 ± 8% to 60 ± 7%, P = .03), and was accompanied by a fall in phosphocreatine/ATP ratio by 0.4 (2.2 ± 0.4 to 1.8 ± 0.1, P = .04). In four groups of eight Wistar rats, we used pyruvate dehydrogenase (PDH) flux studies to demonstrate changes in carbohydrate metabolism induced by the nicotinic acid receptor agonist, Acipimox, using hyperpolarized Carbon-13 (13 C) magnetic resonance spectroscopy. In rats which had been starved overnight, Acipimox caused a fall in ejection fraction by 7.8% (67.5 ± 8.9 to 60 ± 3.1, P = .03) and a nearly threefold rise in flux through PDH (from 0.182 ± 0.114 to 0.486 ± 0.139, P = .002), though this rise did not match pyruvate dehydrogenase flux observed in rats fed carbohydrate rich chow (0.726 ± 0.201). In fed rats, Acipimox decreased pyruvate dehydrogenase flux (to 0.512 ± 0.13, P = .04). Concentration of plasma insulin fell by two-thirds in fed rats administered Acipimox (from 1695 ± 891 ng/L to 550 ± 222 ng/L, P = .005) in spite of glucose concentrations remaining the same. In conclusion, we demonstrate that nicotinic acid receptor agonists impair cardiac contractility associated with a decline in cardiac energetics and show that the mechanism is likely a combination of reduced fatty acid availability and a failure to upregulate carbohydrate metabolism, essentially starving the heart of fuel.
Collapse
Affiliation(s)
- William D Watson
- Department of Cardiovascular Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Kerstin N Timm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Andrew J Lewis
- Department of Cardiovascular Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Jack J J Miller
- Department of Cardiovascular Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Physics, University of Oxford, Oxford, UK
| | - Yaso Emmanuel
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Department of Cardiovascular Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Damian J Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Department of Cardiovascular Medicine, Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| |
Collapse
|
50
|
Valkovič L, Lau JYC, Abdesselam I, Rider OJ, Frollo I, Tyler DJ, Rodgers CT, Miller JJJ. Effects of contrast agents on relaxation properties of 31P metabolites. Magn Reson Med 2020; 85:1805-1813. [PMID: 33090502 DOI: 10.1002/mrm.28541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/22/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022]
Abstract
PURPOSE Phosphorous MR spectroscopy (31P-MRS) forms a powerful, non-invasive research tool to quantify the energetics of the heart in diverse patient populations. 31P-MRS is frequently applied alongside other radiological examinations, many of which use various contrast agents that shorten relaxation times of water in conventional proton MR, for a better characterisation of cardiac function, or following prior computed tomography (CT). It is, however, unknown whether these agents confound 31P-MRS signals, for example, 2,3-diphosphoglycerate (2,3-DPG). METHODS In this work, we quantitatively assess the impact of non-ionic, low osmolar iodinated CT contrast agent (iopamidol/Niopam), gadolinium chelates (linear gadopentetic acid dimeglumine/Magnevist and macrocyclic gadoterate meglumine/Dotarem) and superparamagnetic iron oxide nanoparticles (ferumoxytol/Feraheme) on the nuclear T1 and T2 of 31P metabolites (ie, 2,3-DPG), and 1H in water in live human blood and saline phantoms at 11.7 T. RESULTS Addition of all contrast agents led to significant shortening of all relaxation times in both 1H and 31P saline phantoms. On the contrary, the T1 relaxation time of 2,3-DPG in blood was significantly shortened only by Magnevist (P = .03). Similarly, the only contrast agent that influenced the T2 relaxation times of 2,3-DPG in blood samples was ferumoxytol (P = .02). CONCLUSION Our results show that, unlike conventional proton MR, phosphorus MRS is unconfounded in patients who have had prior CT with contrast, not all gadolinium-based contrast agents influence 31P-MRS data in vivo, and that ferumoxytol is a promising contrast agent for the reduction in 31P-MRS blood-pool signal.
Collapse
Affiliation(s)
- Ladislav Valkovič
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Justin Y C Lau
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Ines Abdesselam
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Ivan Frollo
- Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Damian J Tyler
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Christopher T Rodgers
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- The Wolfson Brain Imaging Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Jack J J Miller
- Oxford Centre for Clinical MR Research (OCMR), RDM Cardiovascular Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford, UK
| |
Collapse
|