Phosphorus magnetic resonance spectroscopy and imaging (31P-MRS/MRSI) as a window to brain and muscle metabolism: A review of the methods

https://doi.org/10.1016/j.bspc.2020.101967Get rights and content

Abstract

Phosphorus magnetic resonance spectroscopy and spectroscopic imaging (31P-MRS/MRSI) non-invasively provide very important information regarding energy metabolism as they can detect high-energy metabolites and membrane phospholipids in vivo. They have repeatedly proven their utility in the study of healthy and disease conditions in skeletal muscle and brain tissue, as many disorders are related to imbalances in bioenergetics processes. However, they are not often used in a clinic setting, as there are technical challenges imposed by the low sensitivity and low concentration of metabolites leading to low signal to noise ratio (SNR), coarse spatial resolution and very long acquisition times. This paper presents an overview of the main techniques used for the acquisition, data reconstruction and processing of 31P-MRS/MRSI experiments with emphasis in methodological aspects, as well as some of their main applications in the study of skeletal muscle and brain tissue. Also, recent advances in the development of accelerated methods for acquisition of 31P-MR data are discussed.

Introduction

Phosphorus magnetic resonance spectroscopy (31P-MRS) and phosphorus magnetic resonance spectroscopic imaging (31P-MRSI) offer a very unique window to non-invasively look at tissue metabolism in vivo. These approaches are capable of tracking high energy metabolites and membrane phospholipids, all of which are involved in cellular energetic processes [1]. Thus phosphorus spectroscopic methods have been attractive across the scientific community in order to study energy metabolism in healthy and pathophysiological conditions. Furthermore, its noninvasive nature makes it an excellent candidate to perform repeated measurements and track disease progression or treatment response. In addition to the information contained in static spectra, these methods also allow evaluation of oxidative energy production by the mitochondria through an exercise challenge and subsequent recovery in skeletal muscle [2].

Although these methods have been around since the beginnings of MRI in the 1970’s, the current technological developments have been spearheaded by the ultra high magnetic field community. This includes improved radio-frequency (RF) coil designs, new data sampling methods and better reconstruction approaches all of which provide significant improvements to the technology making them more attractive to clinicians and researchers. This work provides an overview of the current status of 31P-MRS/31P-MRSI with focus on the technological developments and the main contributions in the study of skeletal muscle and brain metabolism.

The phosphorus nucleus is visible to nuclear magnetic resonance (NMR) due to its natural spin of ½. This isotope has a 100% natural abundance and albeit its relatively low sensitivity of 6.7% when compared to proton (1H), it has encountered relevance when used for in vivo applications due to its presence in important high energy and phospholipid metabolites [1,3].

The chemical shift range where in vivo phosphorus compounds resonate spans around 30 ppm in contrast to the narrow 5 ppm window for 1H MR spectra, additionally the amount of relevant metabolites is considerably less. Within the 31P-MRS spectrum, the fundamental high-energy metabolite phosphocreatine (PCr) is the dominant signal (in brain and skeletal muscle), thus it is often used as a reference and assigned the value of 0 ppm. PCr serves as an immediately available energy reserve ready to restore the expenditure of adenosine tri-phosphate (ATP) in energy demanding processes. ATP contains three phosphate groups; α, β and γ, that give rise to three independent signals in the spectrum at −7.56 ppm, −16.15 ppm and −2.53 ppm respectively (noting the intracellular magnesium (Mg2+) concentration will vary the resonant frequency of the β group). The interaction of the phosphorus nuclei with other nearby spins results in a homonuclear J-coupling effect, causing a line splitting ATP signals into doublets for γ-ATP and α-ATP, and a triplet for β-ATP. The ratios of peak heights are based on binomial expansion, hence doublets are 1:1 and the triplet 1:2:1.

Moving to the positive side of the spectrum, a relatively small singlet signal at approximately 4.85 ppm corresponds to inorganic phosphate (Pi), a degradation product of energy metabolism. This peak shifts in frequency according to intracellular pH (discussed below). Next to Pi, the cell membrane precursors phosphomonoesters (PMEs) phosphocholine (PC) and phosphoetanolamine (PE) are observable at around 6.2 ppm and 6.7 ppm, often times measured as a single PME signal at magnetic field strength B0 ≤3 T. Similarly, phosphodiester (PDE) degradation products of phospholipid metabolism, glycerophosphocholine (GPC) and glycerophosphoethanolamine (GPE) are observed at 2.95 ppm and 3.50 ppm respectively. Depending on the signal to noise (SNR), other important phosphorus containing metabolites such as nicotinamide adenine dinucleotide (NAD) in its oxidized and reduced form (NAD+ and NADH, respectively) and uridine diphosphate glucose (UDPG) are also visible, specially at ultra-high field (i.e. 7 T).

Aside from these metabolic signals, other important information can be deduced from the analysis of the 31P spectrum. First, the chemical environment where phosphorus-containing metabolites reside may change with different physiological or pathological conditions. Of main relevance, the chemical shift of Pi is dependent on the intracellular pH [6], while the resonance of PCr remains constant, the resonance of Pi shifts with pH. Thus it is possible to calculate the pH using the difference in chemical shifts from PCr and Pi (δ) through the so-called modified Henderson-Hasselbach equation as follows:pH(i)=pKA+log(δδHA)δAδHere, pKA=6.75 is the dissociation constant of Pi, δHA=3.27 and δA=5.63 are the chemical shifts of the protonated and non-protonated forms of Pi, respectively. As the main signal from Pi comes from cytoplasm (sarcoplasm in muscle tissue), it is the intracellular pH (pH(i)) that is measured. Second, the concentration of adenosine di-phosphate (ADP) under physiological conditions is too low to be detected using 31P-MRS, however it can be determined from the concentrations of creatine (Cr), ATP and PCr due to the chemical equilibrium in the creatine-kinase (CK) reaction [7], thus:CADP=[Cr]×[ATP][PCr]×[H+]×[KCK]Where KCK=1.66×109 is the equilibrium constant of the CK reaction, assuming that PCr accounts for 85% of total Cr [4,5]. Finally, the cellular concentration of free Mg2+, a physiologically important divalent cation, influences the chemical shift of phosphorus compounds as the Mg complexes of ADP and ATP act as substrates for ATPases and kinases. Thus, the concentration of free Mg2+ can be deducted from the chemical shifts of β and α-ATP or PCr and β-ATP [8,9]. Fig. 1 shows examples of 31P-MRS spectra acquired at 3 T whereas Table 1 summarizes the metabolites, chemical shifts and their in vivo concentrations in skeletal muscle and brain tissue [4,5].

Prior to metabolite quantification, free induction decay (FID) curves (i.e. time domain signals) acquired from a 31P-MRS experiment are preprocessed. This step typically includes zero and first order phase corrections, zero filling to increase spectral resolution and spectral apodization, the latter applied mainly for filtering, especially when SNR is poor.

SNR is perhaps the most important characteristic in obtaining reliable metabolite quantification. If adequate, a common procedure for performing spectral fitting is to use Advanced Method for Accurate, Robust and Efficient Spectral Fitting (AMARES) [10], a time domain algorithm that incorporates prior knowledge and offers flexibility of the fitting parameters. The AMARES routine is available in the jMRUI software package [11] or as part of the MATLAB based OXSA toolbox [12]. Also, the popular MRS fitting package LCModel has been successfully used in the quantification of 31P spectra [13].

Accurate quantification of 31P metabolites in vivo is a challenging task as factors such as field inhomogeneity, relaxation times and coil sensitivities may influence the measurements. Thus, metabolite ratios are often used when reporting results from MRS experiments, especially in clinical literature. However, ratios are incapable of providing information about metabolic changes in pathological conditions, as they may be ambiguous. In order to properly assess them, metabolite concentrations need to be reported. Such quantification requires a calibration or reference compound (Cref) of known concentration and generally, one of two methods is used to obtain the absolute values: in the first one, an external solution with a known concentration is placed outside of the object of study but within the coil sensitivity region. This requires the chemical standard to resonate outside of the spectral metabolite regions to avoid overlap and subsequent difficulty in differentiation. Furthermore, that approach is made more complicated by variation in B1+ field and B0 homogeneity, with respect to the sampled tissue volume. One example of such a compound is hexachlorocyclo-triphosphagene (HCCTP), which resonates at 22.9 ppm, well outside the range of physiological metabolites [14,15]. In the second approach, a metabolite naturally found in the tissue with established concentration is selected as the reference. From a quantitative review of experiments conducted in skeletal muscle [5], the concentration of ATP ([ATP]8.2mmol/L cell water) is appropriate as an internal reference for quantification of healthy muscle metabolites. In the case of brain tissue, a reference value of 3  mmol/L for γ-ATP is often used as internal reference [4,16]. Thus, the concentration of the metabolite (Cmet) of interest is determined as follows:Cmet=[Cref]SmetSrefCMRWhere Sref and Smet are the signals detected from the reference and metabolite, respectively, and CMR is a correction factor accounting for differences in relaxation time, position relative to the coil, magnetic susceptibility or in general any other difference between the reference and the metabolite of interest.

A straightforward way to understand the main requirements for 31P-MRS is by comparing it with 1H as all clinical and research MR systems clearly use the latter as their primary focus. Some disadvantages can be deduced from this comparison such as reduced sensitivity, lower concentration in tissue, longer T1 and shorter T2 relaxation times of 31P compounds. On the other hand, some advantages are also present. First, the significantly larger spectral dispersion of phosphorus metabolites leads to a better separation of signals. This is especially important when measuring PE, PC and GPC as they all are choline-containing metabolites that cannot be separated by 1H spectroscopy. Also, there are no fat or water dominating signals in phosphorus spectra, meaning no frequency selective suppression techniques are required to null these dominating signals. A further difference lies in the relaxation mechanisms that govern each nucleus, whereas for 1H the T1 relaxation is dominated by the magnetic dipole-dipole interaction, the relaxation of 31P metabolites is strongly influenced by the chemical shift anisotropy mechanism, which gets stronger as the magnetic field increases [17,18].

The lower gyromagnetic ratio and consequently lower Larmor frequency of phosphorus compounds is the main difference resulting in differing technical aspects, when comparing to proton scans, as all components in clinical scanners are tuned/optimized to the later. Thus, in order to use those same hardware systems, additional components such as a broadband transmitter/receiver and RF-coils are required. Most organ specific 31P-MRS studies are performed using single loop surface coils to take advantage of their high sensitivity, however their strong variability in B1+ flip angle due to the inhomogeneous excitation profile can compromise the results for spectroscopic imaging applications. As a result, complex phased arrays and whole volume coverage coils with high sensitivity and homogeneous B1+ excitation have been developed [[19], [20], [21], [22], [23]].

A technological development that has played a key role in improving the spectral quality of 31P-MRS experiments is the use of ultra-high field MR systems (i.e. B0 ≥ 7 T) [18,24]. The main features include an SNR boost of roughly double as well as better spectral resolution and the possibility to gain more SNR per unit of time due to the shortening of T1 relaxation times as a consequence of the above mentioned chemical shift anisotropy mechanism [18]. This gain in SNR can be traded for a higher resolution or faster acquisition times. Unfortunately, the availability of such systems for human scanning is still very limited.

Further improvement of signal in phosphorus MRS experiments is possible through the following mechanisms. First, analogous to the homonuclear coupling, there is another coupling interaction between the 31P and 1H nuclei that causes line broadening of the in vivo spectra, especially at the PME and PDE resonances. Applying RF irradiation at the proton frequency during the 31P signal acquisition reduces this heteronuclear coupling. This process effectively decouples the interaction and results in narrower spectral lines, making possible the separation of signals such as PC and PE as well as GPC and GPE at low field strength (i.e. B03T) [25]. Older decoupling approaches used continuous RF transmission at the 1H frequency which escalated the specific absorption rate (SAR). Later, however, clever combinations of RF cycled pulses, such as WALTZ-4 and WALTZ-16, and multiplets of these called supercycles, reduced SAR whole maintaining efficient proton decoupling [26]. Second, the utilization of the nuclear Overhauser effect (NOE) can also enhance the signal of interest. By applying proton RF irradiation during the 31P inter-pulse delay, the sensitivity for the detection of the later has been reported to improve up to 80% in skeletal muscle at low field [27] and 44% in brain at ultra high field [28] In theory the maximal possible NOE is 0.5(1Hγ/31Pγ) = 0.5(42.576 MHz/T / 17.23 MHz/T) = 124%. Here, it is important to mention that a drawback of the above-mentioned techniques is the significant increment in the specific absorption rate (SAR) due to the 1H-RF irradiation. Also, it is difficult to predict the maximal NOE one will get in practice, which adds to complexity in absolute quantitation. Hence, other polarization transfer methods have been explored to enhance the sensitivity of some 31P-MRS signals [[29], [30], [31], [32]].

Section snippets

Spatial localization of the 31P-MRS signal

Due to the low intrinsic SNR of 31P-MRS, it is common to use high sensitivity surface coils, especially when studying body parts of easy access such as skeletal muscle (i.e. lower leg muscles). Nevertheless, limiting the localization to the sensitive volume of the coil does not allow one to distinguish between signals coming from different compartments (i.e. different muscle groups) or deeper structures, thus at least a simple localization strategy is needed. Furthermore, two additional

Assessment of skeletal muscle metabolism

The acquisition of a phosphorus MR spectrum from the lower leg muscles is probably the most common experiment due to the relatively easy access and high 31P metabolite concentrations. Additionally, their physiological importance and high metabolic activity made them a popular target to be studied with a non-invasive technique such as 31P-MRS. The analysis of resting spectra gathers information about muscle fiber composition that is relevant in the assessment of fitness/training status,

Assessment of brain metabolism

The powerful capability of 31P-MRS to non-invasively measure fundamental compounds in energy metabolism has been exploited to better characterize and understand the energetic processes happening in the brain, whether healthy or in diseased condition. The first step towards understanding energy metabolism processes in the brain is done by characterizing the properties of phosphorus metabolites in a healthy condition. First, it is necessary to account for tissue differences in the metabolite

Beyond brain and muscle

In addition to skeletal muscle and brain tissue, the assessment of energy metabolism using 31P-MRS/MRSI has proven useful in other important organs such as the heart and liver.

Abnormalities in myocardial energy metabolism has been suggested as a contributing factor in the development of cardiac disease [133]. Also, energy production in the heart is of particular interest due to its high ATP turnover rate [134]. In order to fulfill the energy requirements of cardiac work load variations, the

Conclusion

Phosphorus MR spectroscopy and spectroscopic imaging provide powerful tools for the non-invasive assessment of energy metabolism in skeletal muscle and brain tissue. The ongoing technological advancements such as multi-channel RF coil arrays, efficient data sampling and reconstruction methods and foremost ultra-high field MR systems, play a fundamental role in order to better understand the physiology of bioenergetic processes as well as in the adoption of these techniques in clinical

Acknowledgements

Funding was provided through a CONACYT (Mexico) scholarship granted to ASD (CVU: 304930) and a NSERC Discovery Grant (RGPIN-2017-06318) to MDN.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (153)

  • T. Wilhelm et al.

    In Vivo 31P echo-planar spectroscopic imaging of human calf muscle

    J. Magn. Reson.

    (2001)
  • R. Pohmann et al.

    Theoretical evaluation and comparison of fast chemical shift imaging methods

    J. Magn. Reson.

    (1997)
  • S. Hu et al.

    Compressed sensing for resolution enhancement of hyperpolarized 13C flyback 3D-MRSI

    J. Magn. Reson.

    (2008)
  • A. Santos-Díaz et al.

    Comparison of compressed sensing reconstruction algorithms for 31P magnetic resonance spectroscopic imaging

    Magn. Reson. Imaging

    (2019)
  • R.L. Greenman et al.

    The feasibility of measuring phosphocreatine recovery kinetics in muscle using a single-shot 31P RARE MRI sequence

    Acad. Radiol.

    (2011)
  • R.L. Greenman et al.

    Simultaneous acquisition of phosphocreatine and inorganic phosphate images for Pi:PCr ratio mapping using a RARE sequence with chemically selective interleaving

    Magn. Reson. Imaging

    (2011)
  • J. Satrústegui et al.

    An in vivo phosphorus nuclear magnetic resonance study of the variations with age in the phosphodiers’ content of human muscle

    Mech. Ageing Dev.

    (1988)
  • D.J. Taylor et al.

    Bioenergetics of skeletal muscle in mitochondrial myopathy

    J. Neurol. Sci.

    (1994)
  • A. Greiner et al.

    High-energy phosphate metabolism during incremental calf exercise in patients with unilaterally symptomatic peripheral arterial disease measured by phosphor 31 magnetic resonance spectroscopy

    J. Vasc. Surg.

    (2006)
  • D.C. Isbell et al.

    Delayed calf muscle phosphocreatine recovery after exercise identifies peripheral arterial disease

    J. Am. Coll. Cardiol.

    (2006)
  • L. Valkovič et al.

    In-vivo 31P-MRS of skeletal muscle and liver: a way for non-invasive assessment of their metabolism

    Anal. Biochem.

    (2017)
  • J.A. Jeneson et al.

    The signal transduction function for oxidative phosphorylation is at least second order in ADP

    J. Biol. Chem.

    (1996)
  • K. Tschiesche et al.

    MR-compatible pedal ergometer for reproducible exercising of the human calf muscle

    Med. Eng. Phys.

    (2014)
  • R.A. de Graaf

    In Vivo NMR Spectroscopy: Principles and Techniques

    (2013)
  • G.J. Kemp et al.

    Quantification of skeletal muscle mitochondrial function by 31P magnetic resonance spectroscopy techniques: a quantitative review

    Acta Physiol. (Oxf)

    (2015)
  • J. Ren et al.

    31 P-MRS of healthy human brain: ATP synthesis, metabolite concentrations, pH, and T1 relaxation times

    NMR Biomed.

    (2015)
  • G.J. Kemp et al.

    Absolute quantification of phosphorus metabolite concentrations in human muscle in vivo by 31P MRS: a quantitative review

    NMR Biomed.

    (2007)
  • G.J. Kemp et al.

    Control of phosphocreatine resynthesis during recovery from exercise in human skeletal muscle

    NMR Biomed.

    (1993)
  • R. Gupta

    NMR studies of intracellular metal ions in intact cells and tissues

    Annu. Rev. Biophys. Biomol. Struct.

    (2002)
  • S. Iotti et al.

    In vivo assessment of free magnesium concentration in human brain by 31P MRS. A new calibration curve based on a mathematical algorithm

    NMR Biomed.

    (2002)
  • D. Stefan et al.

    Quantitation of magnetic resonance spectroscopy signals: the jMRUI software package

    Meas. Sci. Technol.

    (2009)
  • L.A.B. Purvis et al.

    OXSA: an open-source magnetic resonance spectroscopy analysis toolbox in MATLAB

    PLoS One

    (2017)
  • D.K. Deelchand et al.

    Quantification of in vivo 31P NMR brain spectra using LCModel

    NMR Biomed.

    (2015)
  • F. Du et al.

    Efficient in vivo 31P magnetization transfer approach for noninvasively determining multiple kinetic parameters and metabolic fluxes of ATP metabolism in the human brain

    Magn. Reson. Med.

    (2007)
  • W. Bogner et al.

    Assessment of 31 P relaxation times in the human calf muscle: a comparison between 3 T and 7 T in vivo

    Magn. Reson. Med.

    (2009)
  • B.L. van de Bank et al.

    Optimized 31 P MRS in the human brain at 7 T with a dedicated RF coil setup

    NMR Biomed.

    (2015)
  • N.I. Avdievich

    Transceiver-phased arrays for human brain studies at 7 T

    Appl. Magn. Reson.

    (2011)
  • S. Goluch et al.

    A form-fitted three channel 31 P, two channel 1 H transceiver coil array for calf muscle studies at 7 T

    Magn. Reson. Med.

    (2014)
  • J. Löring et al.

    Whole-body radiofrequency coil for 31P MRSI at 7T

    NMR Biomed.

    (2016)
  • W. Bogner et al.

    In vivo 31P spectroscopy by fully adiabatic extended image selected in vivo spectroscopy: a comparison between 3 T and 7 T

    Magn. Reson. Med.

    (2011)
  • P.R. Luyten et al.

    Broadband proton decoupling in human 31P NMR spectroscopy

    NMR Biomed.

    (1989)
  • R. Freeman

    Chapter 6: separating the wheat from the chaff

    Spin Choreography: Basic Steps in High Resolution NMR

    (1997)
  • T.R. Brown et al.

    NOE enhancements and T1 relaxation times of phosphorylated metabolites in human calf muscle at 1.5 Tesla

    Magn. Reson. Med.

    (1995)
  • H. Lei et al.

    In vivo 31P magnetic resonance spectroscopy of human brain at 7 T: an initial experience

    Magn. Reson. Med.

    (2003)
  • O. Gonen et al.

    In vivo phosphorus polarization transfer and decoupling from protons in three-dimensional localized nuclear magnetic resonance spectroscopy of human brain

    Magn. Reson. Med.

    (1997)
  • D.W.J. Klomp et al.

    Efficient 1H to 31P polarization transfer on a clinical 3T MR system

    Magn. Reson. Med.

    (2008)
  • W.J.M. Van der Kemp et al.

    Adiabatic multi-echo 31P spectroscopic imaging (AMESING) at 7T for the measurement of transverse relaxation times and regaining of sensitivity in tissues with short T2* values

    NMR Biomed.

    (2013)
  • W.J.M. van der Kemp et al.

    Increased sensitivity of 31P MRSI using direct detection integrated with multi-echo polarization transfer (DIMEPT)

    NMR Biomed.

    (2014)
  • M. Chmelík et al.

    Fully adiabatic 31P 2D-CSI with reduced chemical shift displacement error at 7 T - GOIA-1D-ISIS/2D-CSI

    Magn. Reson. Med.

    (2013)
  • C.T.W. Moonen et al.

    Comparison of single-shot localization methods (steam and press) for in vivo proton NMR spectroscopy

    NMR Biomed.

    (2007)
  • Cited by (15)

    • Magnetic Resonance Spectroscopy in Bipolar Disorder

      2022, Biomarkers in Bipolar Disorders
    • NMR spectroscopy of wastewater: A review, case study, and future potential

      2021, Progress in Nuclear Magnetic Resonance Spectroscopy
      Citation Excerpt :

      31P is a spin ½ nucleus whose chemical shift range spans ~800 ppm (though to be fair, most environmentally relevant compounds range from ~−20 to 20 ppm), and combined with sharp lineshapes, gives reasonable resolution. 31P has been heavily employed in NMR studies of energy metabolites [353,420–423]. In contrast to 19F, which is typically not present in naturally occurring metabolites, one of the major strengths of 31P is its prevalence in energy mediating molecules such as adenosine triphosphate, diphosphate, monophosphate (ATP, ADP, AMP) and phosphocreatine, as well as inorganic phosphate.

    View all citing articles on Scopus
    View full text