Muscle Biopsy for Diagnosis of Neuromuscular and Metabolic Diseases


Introduction

In the 1960s and early 1970s, histochemical stains were for the pathological examination of the routine muscle biopsy to define myofiber types and certain metabolic disturbances, and they continue to be the standard of diagnostic examination of the muscle biopsy. Transmission electron microscopy (EM) of the muscle biopsy began in earnest a decade earlier, but the findings and their interpretation were greatly expanded during the same period of histochemistry. The further introduction of immunocytochemistry to specify the expression of specific sarcolemmal regional proteins and distinguish types of inflammatory cells in the late 1980s and 1990s served to further enhance the value of the muscle biopsy as the primary diagnostic technique in neuromuscular diseases. Only a few specific disorders, notably disorders of neuromuscular transmission and a few myopathies such as myotonia congenita, remain better defined by electrophysiological testing than by muscle biopsy. Though muscle biopsy remains a primary diagnostic tool for myopathies and neuropathies, genetic testing for etiological mutations, deletions, and expansions in specific genes, performed from leukocytes in blood samples, has superseded muscle biopsy in recent years in the diagnosis of spinal muscular atrophy (SMA), many muscular dystrophies, and other myopathies because genetic testing is less invasive and more specific at the molecular genetic level. For many disorders in which the genetic basis remains unknown or testing is inaccessible because of cost, and also in disorders not of genetic origin, muscle biopsy remains a valuable and primary diagnostic method.

This chapter summarizes the most frequently employed neuropathological techniques in examining the muscle biopsy and the principal findings in the various disorders in which it contributes to a definitive diagnosis.

Tissue Selection and Preparation

Muscle selection for biopsy should avoid both end-stage muscles that are likely to show only nonspecific fibrosis and loss of myofibers, and clinically uninvolved muscles that may not yet be pathologically diagnostic. The vastus lateralis of the quadriceps femoris muscle is most often selected in children because it is a large muscle from which adequate tissue can be taken without causing weakness, it is involved in nearly all myopathic disorders, it is easily accessible away from major nerves and blood vessels, it shows relatively equal numbers of types I and II myofibers, and it is the best-documented muscle in terms of normal controls. In young children, the risk of general anesthesia can sometimes be avoided by a skilled femoral nerve block and mild oral or intravenous sedation.

The specimen should be dealt with promptly after removal to minimize autolytic loss of important enzymatic activities and of ultrastructural details by delayed freezing or improper fixation. Four portions of the muscle biopsy tissue are usually taken:

  • (1)

    A small portion frozen in isopentane (2-methylbutane) cooled to the temperature of liquid nitrogen (–160°C) for cryostat sections oriented transversely for light microscopy and histochemistry.

  • (2)

    A larger portion frozen in a freezer at −80°C for biochemical assay. Preservation of histological detail is not an important consideration for biochemical studies; therefore, isopentane is not needed and would interfere with some assays.

  • (3)

    A small portion is immersed in 2% buffered glutaraldehyde for electron microscopy. Use of an isometric clamp has advantages: the muscle is relatively relaxed, so features in the I-band are clearly seen. Orientation is made easy. Dicing makes orientation difficult. Gluteraldehyde penetrates between the muscle fibers sufficiently to allow good fixation as long as the piece of tissue removed is not too thick. After sufficient time in fixative, the specimen can remain refrigerated in buffer for several days if desired, to optimally schedule blocking and embedding. The semithin plastic sections should be carefully examined because they give the best light-microscopic morphology. The very best resolution comes from paraphenylenediamine-stained sections viewed with a phase microscope, but toluidine-blue stained sections are more widely used because phase microscopes are not readily available in all laboratories. Both transverse and longitudinal sections should be cut, and at least 2 transverse and 1 longitudinal examined. From the semithin sections, it can be decided whether or not electron microscopy is needed. The unused tissue can be kept in reserve in buffer for a few weeks or can be embedded in epoxy resin for future study, and preserved indefinitely.

  • (4)

    If enough tissue is available, a small portion can be fixed in 10% buffered formalin for paraffin embedding. Paraffin sections are not essential. The histology can be better studied from the frozen sections, which, when competently done, have fewer artifacts than paraffin sections, though nuclear detail is not as well preserved.

An open biopsy is generally preferable to needle biopsies of muscle because of the volume of tissue required for the various studies. Unlike needle biopsies of the liver or kidney, muscle needle biopsies still require a small skin incision and a large-bore needle, and they are not less traumatic than open biopsies. Needle biopsies are more likely to lead to the complication of focal hematoma formation after the biopsy.

Histopathological Aspects of Muscle Biopsy Preparation and Examination

Histological Examination

Hematoxylin and eosin (HE) stain has been the most universal and traditional method for examination of formalin-fixed, paraffin-embedded tissue sections of all tissues for more than a century, and its application to cryostat sections of the muscle biopsy is no exception. The amount of diagnostic information yielded by histological examination alone is limited, but it can detect the presence of inflammation; foreign tissues including Trichinella and other parasites; neoplasia, both primary and metastatic; myofiber necrosis; perimysial and endothelial connective tissue proliferation; and patterns of distribution of atrophic myofibers. Subcellular structural abnormalities, such as ragged-red fibers, are subtler and harder to see than with histochemical stains and some, such as nemaline rods, are almost impossible to detect because of lack of color contrast.

Histochemical Examination

The purpose of histochemistry is to distinguish myofiber types and subtypes, certain metabolic products such as glycogen and neutral lipids, and many subcellular abnormalities not well seen with HE. Histochemical studies are traditionally performed on freshly frozen sections only, though some enzymatic activities and fiber typing using myosin and actin antibodies can also now be demonstrated in paraffin sections by immunocytochemistry. The most useful and frequently applied histochemical stains are:

  • (1)

    Modified Gomori trichrome. The chromotrope-2 R component of this stain has a high affinity for phospholipids and is perceived as a red color; mitochondrial and endoplasmic reticulum membranes have high phospholipid content in contrast with most plasma membranes that have a higher ratio of glycolipids. In normal myofibers in transverse section, a thin subsarcolemmal rim of red staining may be seen because of mitochondrial concentration, particularly evident at motor endplates. The intermyofibrillar sarcoplasm appears as a red reticular network because of mitochondria and endoplasmic reticulum against the green staining of myofibrils from the fast green FCF component. The third component of this trichrome stain is hematoxylin, which stains nuclear chromatin (DNA) purple. This stain is ideal for demonstrating ragged-red fibers of some mitochondrial cytopathies. Chromotrope-2 R also stains tropomyosin, so nemaline rods arising from the Z-band also are well demonstrated. Myelin sheaths of normal intramuscular nerve twigs are also intensely red stained.

  • (2)

    Myofibrillar calcium-mediated adenosine triphosphatase (ATPase) stains. These stains are highly reliable for distinguishing fiber types and subtypes, depending on the pH of preincubation. At alkaline pH (usually 9.4 to 10.2), type II fibers are dark and type I fibers are light. At acid pH (usually 4.6–4.3), the reverse is true, and subtypes of II can be distinguished. Types of intrafusal myofibers of muscle spindles differ from those of extrafusal fibers. These stains are very helpful for those myopathies showing type-specific atrophies. In chronic neurogenic atrophy with many cycles of denervation and reinnervation, a phenomenon of histochemical type-grouping is seen, in which the normal intermixed checkerboard distribution of types I and II myofibers is replaced by small and large groups of myofibers, all of the same type, adjacent to other groups of the opposite type. Some groups may be so large as to occupy an entire fascicle.

  • (3)

    Respiratory chain complexes, or “oxidative mitochondrial enzymes.” The three most relevant stains include nicotinamide adenine dinucleotide tetrazolium reductase (NADH-TR; respiratory complex I), succinate dehydrogenase (SDH; complex II) and cytochrome c oxidase (COX; complex IV). These activities are greatly increased in ragged-red zones. In many mitochondrial diseases, scattered myofibers that are otherwise well preserved and not degenerating show loss of COX activity, even if complex IV is not selectively impaired. One caution is that the intensity of histochemical activity of COX is generally lower than either NADH-TR or SDH, particularly in type II myofibers, hence normally low activity should not be misinterpreted as pathologically absent. Modification of the SDH stain by the addition of phenazine methosulfate is useful because this compound suppresses activity in normal mitochondria but does not affect SDH expression in abnormal mitochondria. Because complex II has only four subunits, all encoded by nDNA, it remains reactive in all mitochondrial diseases regardless of any mtDNA point mutations. The other four complexes include a combination of mtDNA and nDNA encoded subunits. These mitochondrial enzymes also are markers of fiber types, showing more intense activity in type I fibers than those of type II, but caution must be exercised in interpretation because severely atrophic myofibers of either type show intense activity due to loss of myofibrils and concentration of sarcoplasmic mitochondria. NADH-TR marks not only mitochondria but sarcoplasmic (i.e. endoplasmic) reticulum as well, unlike SDH and COX, which are pure mitochondrial stains.

  • (4)

    Acid phosphatase. This stain is useful for showing abnormal lysosomal activity in diseases such as infantile (Pompe) and adult acid maltase deficiencies. It appears as granular material within the myofiber. Nonspecific esterase can used as an alternative. Alkaline phosphatase is not a suitable alternative.

  • (5)

    Oil Red O and Sudan Black B. These stains are used to demonstrate neutral lipids within myofibers and in the connective tissue, but complex phospho- and glycolipids, such as sphingomyelin and galactocerebroside, are not stained.

  • (6)

    Periodic acid-Schiff (PAS) reaction. Increased intramuscular glycogen occurs in some glycogenoses, but may also represent a physiological condition of a patient who is receiving good nutrition, even parenterally for an extended time, and who has little physical activity to utilize the glycogen that accumulates in myofiber, as in children in intensive care units. PAS also is good for demonstrating peripheral nerve myelin in both cryostat and paraffin sections.

Immunocytochemical Reactivities

These techniques are usually applied to frozen cryostat sections, but also may be applied to paraffin-embedded sections, except for dystrophin. They can demonstrate features not shown by the routine histochemical battery of stains, including sarcolemmal-region proteins both of the sarcolemma itself and those that attach the sarcolemma to the basal lamina (basement membrane). They are highly specific for many muscular dystrophies, such as the dystrophinopathies, merosin deficiency, and dystroglycanopathies. Other immunoreactivities can substitute for histochemistry in distinguishing fiber types if only formalin-fixed, paraffin-embedded sections are available, for example heavy- and light-chain (“slow” and “fast”) myosin immunoreactivities that provide similar information as myofibrillar ATPase stains. Immunocytochemistry can also distinguish types of inflammatory cells, such as T-lymphocytes (CD-43), B-lymphocytes (CD-20), and macrophages (CD-68). Endothelial cells of capillaries and other small intramuscular blood vessels are well demonstrated by vimentin and nestin immunoreactivities, or by the more specific CD-31 and CD-34 antibodies.

Silver Impregnations

This group is useful for showing axons in peripheral nerve, including intramuscular nerves, and includes variations of technique such as those of Bielschowsky, Bodian, Sevier-Munger, and others. Reticulin stains demonstrate reticulin fibers around intramuscular capillaries.

Transmission Electron Microscopy (EM)

Since the 1950s, EM has provided ultrastructural insight into the subcellular anatomy of myofibers ( Figure 4.1 ) and of aberrations in many myopathies, to both confirm and verify the interpretation of histochemical stains and to offer additional detail at a resolution not possible by light microscopy. In recent years, it has been used more sparsely and selectively because of its labor intensity for both technologists and pathologists and because of cost, including the maintenance of the equipment. Special tissue preparation is required, with fixation in glutaraldehyde, post-fixation in osmium tetroxide, and staining with uranyl acetate and lead citrate. Ultrafine sections must be cut on special diamond knife blades. EM can, however, demonstrate many features of diagnostic importance if used for selective muscle biopsies. Nemaline rods and disarrays of myofilaments are well shown. One of its most important contemporary applications is to demonstrate ultrastructural changes in mitochondrial cytopathies. There are several mitochondrial alterations, including simplification of the cristae, stacked and whorled cristae, increased numbers of osmophilic spheroids within the mitochondrial matrix and paracrystalline structures. The latter are an array of geometrical crystal-like structures within the mitochondria and are a highly specific finding. Within myofibers, paracrystalline structures (sometimes called “inclusions,” though this is incorrect because they are not foreign particles) often assume a brick shape with sharp corners and elongation. The ultrastructural periodicity of these structures is constant and predictable; these structures are in the space between the cristae and are made of creatine kinase. Often, fragments of preserved cristae can also be seen within the same mitochondrion that has a paracrystalline structure, confirming their location as intramitochondrial. Endothelial cells of intramuscular capillaries in young infants with mitochondrial disease also contain paracrystalline-like structures suggestive of intramitochondrial location but without preserved cristae. Normal Weibel-Palade granules of endothelial cells that contain von Willebrand factor V consist of parallel microtubules that, on oblique or longitudinal sections, have a repetitive pattern reminiscent of a crystal.

Figure 4.1, This electron micrograph (EM) shows a longitudinal view of part of a normal muscle fiber fixed at resting length. The moderately dark A-bands contain the myosin (thick) filaments. In the part of the A-bands that adjoin the I-bands, the actin (thin) filaments are interspersed with the thick filaments. The very dark Z-disc in the middle of the I-band marks the boundary of the sarcomere. The plane of section passes through the trajectory of the T-tubules, some of which are marked by arrows. The darkish zone next to the T-tubule walls marks the ryanodine receptor, which is part of the wall of each cistern of sarcoplasmic reticulum, two cisterns and the T-tubule between them forming the triads. The longitudinal part of the SR is not visible. The small round granules are glycogen. The section grazes two mitochondria where one I-band goes out of the plane of section.

Biochemical Assay of Respiratory Chain Complexes

The measurement of specific activities of each of the five respiratory chain complexes, both individually and in combination (e.g. complexes 1+III), may be performed from homogenates of frozen tissue and provides quantitative numbers. Citrate synthetase is usually examined as a control because if it is decreased, the other specific activities may be unreliable due to delayed freezing of the tissue. At times, one may find convincing histochemical and ultrastructural evidence of mitochondrial disease, associated with normal activities of the complexes, particularly in infants. This anomaly is due to the measurement of an average of the muscle tissue homogenate rather than an examination of individual myofibers. If only a few scattered myofibers are affected, they may be diluted by the normal activities of the majority of myofibers. Interpretation of these assays must be made cautiously, and clinicians must understand that normal quantitative assay results do not necessarily exclude mitochondrial disease. Similarly, decreased specific activity in one complex may be an artifactual result and, in the absence of supportive evidence in other aspects of the muscle biopsy, should not be taken as conclusive for mitochondrial cytopathy. Diminished activity in all or most complexes with normal citrate synthetase activity is a primary criterion of the “mitochondrial depletion syndrome” of infancy, a genetically heterogeneous group with many clinical phenotypical features in common.

Molecular Genetic Studies

Muscle tissue can be used for molecular genetic studies of many muscular dystrophies. It is also useful in showing both mitochondrial DNA (mtDNA) point mutations and nuclear DNA (nDNA) mutations or deletions in mitochondrial disorders. Ideally, the tissue should be freshly frozen for such studies, but DNA extraction can also be made from formalin-fixed tissue, though this is less reliable. Details of techniques and discussion of the many mutations that have been demonstrated in mitochondrial diseases are beyond the scope of this chapter.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here