Skeletal Muscle and Peripheral Nerve Disorders


Skeletal muscle, which represents 40 % of the weight of a reasonably fit human, has one major function: converting food into force. The unit of contraction in skeletal muscle is the sarcomere, a 2.5 × 1 × 1 µm lattice of interdigitating thick (predominantly myosin) and thin (predominantly actin) filaments, the latter anchored to Z-discs. Z-discs (visible by light microscopy as cross striations; see Fig. 11.3C ) keep adjacent sarcomeres aligned and are linked via desmin to the outer membrane of the muscle fiber (sarcolemma). This structure enables the rapid and factorial generation and relaxation of force that is absent in other contractile actomyosin systems such as smooth muscle. A myofibril consists of a series of sarcomeres (10,000 per inch) attached to each other beginning at a muscle’s origin and ending at its insertion (a myofibril in your biceps muscle is 1 sarcomere thick and about 100,000 sarcomeres long). Each ∼50-µm diameter muscle fiber cross section that we recognize under the light microscope ( Fig. 11.1A–B ) is a multinucleated syncytium consisting of roughly parallel arrays of a few thousand myofibrils. Grossly visible muscles are composed of roughly parallel arrays of thousands to millions of muscle fibers (the gastrocnemius muscle, for example, contains about one million fibers; the bicep muscle contains about 200,000). Although the number of muscle fibers within a muscle is fairly static, the number of myofibrils within each of these muscle fibers is dynamic and underlies changes in muscle size with activity, inactivity, and aging.

Figure 11.1
A , Normal (frozen) muscle, H&E. Note the uniformity of muscle fiber size and shape, peripherally located multiple small nuclei, and the absence of endomysial fibrous tissue. Freezing (ice crystal) artifact produces a waffle-like appearance in some of the muscle fibers. B , Normal frozen muscle, Gomori trichrome stain. Fibers stain bluish-green; perimysial and endomysial collagen stains green; nuclei are dark. There are no aggregates of any color within the sarcoplasm. The ice crystal artifact is still seen within the same fibers as on the H&E stain. C , H&E section containing small groups of angulated atrophic fibers characteristic of denervation atrophy. Several “target fibers” are present ( arrows ), indicative of reinnervation. D , Paraffin-embedded muscle from a patient with Charcot-Marie-Tooth disease showing features indistinguishable from dystrophic myopathy, including endomysial fibrosis.
E Severely atrophic muscle fibers (nuclear bags) may resemble inflammatory cells ( arrows ). On careful examination, cytologic features are distinctive, including intense eosinophilic cytoplasm and multinucleation. F , Fiber atrophy of immature muscles, which is classically encountered in infantile spinal muscular atrophy, results in large groups of either hypertrophic or markedly atrophic fibers with rounded rather than angulated profiles. G , Reinnervated fibers assume biochemical characteristics dictated by the parent neuron, resulting in loss of the normal mosaic pattern on ATPase histochemistry (fiber-type grouping). H , Rounded atrophic fibers are usually an indicator of myopathic stress, ranging from disuse to various primary muscle diseases. Immunohistochemical staining with antibodies to the slow myosin heavy chain reveals selective atrophy of type 1 fibers in a patient with mitochondrial disease.

Muscle contraction is the result of adenosine triphosphate (ATP)-dependent movement of actin along myosin. Very little ATP is stored within muscle. Therefore efficient metabolism of glycogen and lipids is necessary to support ongoing muscle activity. Two major types of muscle fibers have evolved: those capable of generating large amounts of force quickly (fast twitch) and those capable of sustaining force over a more protracted interval (slow twitch). Slow-twitch muscle fibers, being more dependent on a steady supply of oxygen, contain greater amounts of myoglobin and thus appear redder than fast-twitch fibers (in Thanksgiving lingo, “dark meat versus white meat”). Slow-twitch fibers also contain more lipids and mitochondria than fast-twitch fibers; the latter contain more glycogen and myofibrillary adenosine triphosphatase (ATPase). Although fiber types are often designated as type 1 or type 2 based on the stability of myofibrillary ATPase isoenzymes in acid and alkaline incubation, the speed and stability of contraction are primarily determined by myosin-heavy chain isoforms, which can be reliably detected using specific antibodies. Muscles composed of single fiber types are found in some animals (type 1 muscles allow clams to secure their entrance against intruders, and type 2 muscles allow hummingbirds to hover while eating), but in humans, all muscles include both fiber types in varying proportions.

Each muscle fiber is innervated by a motor axon emanating from an anterior horn cell neuron within the spinal cord. A single anterior horn cell innervates anywhere from several hundred to several thousand muscle fibers, depending on the precision and force needed to accomplish required muscle functions. These motor units are interspersed among each other within muscle to form a mosaic pattern, easily visualized by immunohistochemical staining for myosin-heavy chain isoforms or ATPase histochemical staining. The rate and pattern of neuronal firing determine fiber type. Neuromuscular continuity is also required to maintain sarcomeric balance. In the absence of innervation (or artificial stimulation), the 1 % to 2 % of muscle protein that degenerates daily is not replaced, and fibers within the motor unit atrophy. Muscle fibers reinnervated by axonal sprouting from adjacent motor units (within about a 2-month window) can regain normal size and shape, although their type will conform to that of the reinnervating motor unit. Reinnervation is manifested electrophysiologically as large polyphasic motor unit action potentials and pathologically as fiber-type grouping (i.e., interruption of the normal mosaic pattern by single-type fiber groups).

Although we are now well into the molecular age of myology, patients will continue to have symptoms and signs of muscle dysfunction, clinicians will almost certainly continue to evaluate these patients using anatomic and physiologic algorithms, and pathologists will indubitably continue to receive biopsy specimens without any of this information. Nevertheless, efficient pathologic diagnosis of muscle disease is only possible with some knowledge of clinical manifestations. Age at onset, duration of symptoms, and distribution of dysfunction are probably the most important, followed closely by whether symptoms are predominantly progressive or episodic.

Tremendous advances in our understanding of the molecular bases of muscular diseases have led to several conceptual shifts in our approach to the clinicopathologic diagnosis of muscle biopsy specimens.

Our current understanding of neuromuscular physiology allows us to associate skeletal muscle diseases with disorders of innervation and neuromuscular transmission, as well as dysfunction of proteins involved in each of the steps required for effective locomotion: excitation–contraction coupling, muscle contraction, and mechanical transduction ( Table 11.1 ). In addition, as locomotion requires large amounts of energy, disorders of energy metabolism are often expressed as compromised neuromuscular function. Finally, skeletal muscle is not exempt from the dysregulation of the immune system, and autoimmune inflammatory myopathies comprise a large proportion of diagnostic muscle biopsies.

Table 11.1
Intrinsic disorders of skeletal muscle
Physiology Pathology Major Proteins Helpful Stains
Excitation-Contraction Coupling Periodic paralyses Calcium channels H&E, trichrome
Myotonic dystrophies Chloride channel, insulin receptor H&E
Core myopathies Ryanodine receptor
Selenoprotein N
Oxidative
Contraction Nemaline myopathy Nebulin, actin Trichrome
Quadriplegic myopathy Myosin Myosin IHC
Z-disc myopathies Myotilin, ZASP H&E, trichrome
Mechanical Transduction Desminopathies Desmin, αβ-crystallin Oxidative
Duchenne dystrophy
Becker dystrophy
Dystrophin Dystrophin IHC
Limb-girdle dystrophies Calpain, dysferlin, dystrophin-associated proteins Specific Abs
Congenital dystrophies Laminin α 2 , α-dystroglycan Specific Abs
Abs , antibodies; H&E , hematoxylin and eosin; IHC , immunohistochemistry.

One cannot get far in the discussion of muscle diseases before entering the somewhat arcane world of specimen processing, histochemical staining, immunohistochemical staining, and electron microscopy. These techniques should be seen as important aids for diagnosis rather than as obstacles to understanding.

Ideally, processing of a muscle biopsy specimen consists of dividing the muscle into four portions: a small aliquot fixed in glutaraldehyde or half-strength Karnovsky glutaraldehyde/paraformaldehyde solution, and three roughly equal quantities of muscle embedded transversely in OCT embedding media (Miles Scientific) and flash-frozen in liquid nitrogen–cooled isopentane (2-methylbutane) for histochemistry; placed in a sterile container or aluminum foil, flash-frozen in liquid nitrogen, and stored at –80°C; or fixed in formalin and embedded longitudinally in paraffin for routine light microscopy. Because focal abnormalities (e.g., inflammatory infiltrates) are best visualized within paraffin-embedded specimens, these specimens are serially sectioned at approximately 100-µm intervals with alternate sections stained with hematoxylin-eosin (H&E) or left unstained on charged slides for possible immunohistochemical studies.

Which of the multitude of stains is worth performing routinely (i.e., on every muscle biopsy)? We would venture that each of the authors contributing to this text would answer this question differently, although many would agree on the essentials. The EURO-NMD pathology working group recommends that all new biopsies get four histologic stains (H&E, Gomori trichrome, oil red O or Sudan black, periodic acid–Schiff [PAS]), seven histochemical stains (NADH-tetrazolium reductase [NADH-TR], combined succinate dehydrogenase [SDH]/cytochrome oxidase [COX], acid phosphatase, and myofibrillary ATPase at 3 pH levels, although these last three may be replaced by myosin-heavy chain immunostaining), and six immunohistochemical stains (four myosin-heavy chains [neonatal/fetal, developmental/embryonic, fast, and slow/beta cardiac], major histocompatibility complex [MHC] class I, and p62). In our laboratories, we routinely perform H&E, Gomori trichrome, NADH-TR, and cytochrome oxidase; assess fiber type using antibodies for fast and slow myosin-heavy chains; and perform immunohistochemical staining for CD56, which highlights both activated satellite cells and acutely denervated muscle fibers. We also routinely examine 1-µm plastic-embedded sections for structural and storage abnormalities and follow up with electron microscopy as needed.

As in all of surgical pathology, initial assessment begins with the H&E-stained slide. We begin by looking for atrophy and/or degeneration of muscle fibers. If atrophy is identified, we try to determine whether it is distributed throughout the biopsy. If so, we use myosin immunohistochemistry to ascertain whether the atrophy is restricted to type 2 fibers (nonspecific) or type 1 fibers (suggestive of several metabolic and structural muscle diseases). If not, we determine whether the atrophic fibers appear angulated and grouped (denervation atrophy) or reside predominantly adjacent to perimysium (suggesting dermatomyositis). If there appears to be muscle fiber degeneration, we confirm our suspicions by finding associated macrophage infiltration, evidence of myofiber regeneration (CD56 immunostaining is especially useful for this), or increased numbers of fibers containing centrally located nuclei. We then assess the specimen for endomysial fibrosis (dystrophic myopathies); lymphocytic inflammation within the endomysium, perimysium, or epimysial blood vessel walls; and/or vacuolar changes in muscle fibers (plastic-embedded sections). Confronted with an entirely normal H&E-stained muscle biopsy, we peruse our trichrome-stained section for evidence of tubular aggregates or sarcomeric abnormalities, such as nemaline rods and rimmed vacuoles, and our cytochrome oxidase–stained section for evidence of mitochondrial dysfunction, proliferation, or maldistribution (central cores, minicores, “rubbed-out” fibers). If our initial battery of stains fails to provide diagnostic information, we consult the clinical history for evidence of myasthenia gravis, abnormalities in excitation–contraction coupling, or diseases of energy metabolism, and pursue histochemical and ultrastructural evaluations as indicated in the discussions that follow.

It has been stated that what separates the novice from the master pathologist is the ability to recognize artifacts. Muscle biopsy interpretation is frequently complicated by one or more of the following:

  • 1.

    Hypercontraction of fibers caused by handling of the muscle before fixation or freezing. These fibers appear identical to acutely necrotic fibers. In bona fide muscle disease, accompanying regenerating muscle fibers can usually be found, although in rare instances definitive resolution may not be possible.

  • 2.

    Ice crystal artifact as a result of poor (slow) freezing. When mild, holes left in muscle fibers may be confused with vacuolar degeneration. Vacuoles associated with disease are usually more varied in size and are also visible within paraffin- and/or plastic-embedded sections. Severe freezing artifact can render the biopsy specimen uninterpretable; thawing and refreezing the muscle may ameliorate the problem.

  • 3.

    A single focus of atrophic or degenerated fibers in an otherwise normal-appearing biopsy specimen. The former is usually due to the proximity of the sampled muscle to a tendinous insertion; the latter may indicate muscle trauma prior to biopsy (e.g., biopsy of a muscle previously used for electrophysiologic testing).

We now turn to a survey of skeletal muscle pathology.

DISORDERS OF INNERVATION (DENERVATION ATROPHY)

Clinical Features

Although denervation atrophy is not a disease unto itself, it is among the most common diagnoses found in muscle biopsy specimens. Elderly patients with radiculopathies and/or neuropathies often have clinical and laboratory features suggestive of muscle disease: weakness that is not obviously distal, elevated creatine kinase, and unreliable physical findings.

DENERVATION ATROPHY—FACT SHEET

Definition

  • n

    Sarcomeric loss secondary to loss of innervation

Incidence and Location

  • n

    Common; specific incidence depends on underlying disease

  • n

    Pattern of weakness corresponds to myotomes involved if radiculopathy

  • n

    Weakness due to neuropathy depends on pattern of involvement (multifocal and asymmetric, length-dependent or nonlength-dependent)

Sex and Age Distribution

  • n

    Dependent on cause: peaks include infancy (spinal muscular atrophy) and advancing age (peripheral neuropathies, radiculopathies, motor neuron disease)

Clinical Features

  • n

    Muscle weakness that is predominantly distal (except in spinal muscular atrophy)

  • n

    Gross muscle atrophy in severe cases

  • n

    Decreased deep tendon reflexes for diseases that are lower motor neuron only

  • n

    Accompanying sensory loss (in most neuropathies and radiculopathies)

Prognosis and Treatment

  • n

    Dependent on cause

Pathologic Features

The cardinal histopathologic feature of active denervation is small group atrophy: clusters of a few to several angulated muscle fibers in proximity (but not necessarily adjacent) to each other (see Fig. 11.1C ). These fibers are smaller than the intervening normal (or occasionally hypertrophied) muscle fibers, and all of the fibers within a group appear atrophied to approximately the same extent. On reinnervation, sarcomeric reconstitution is initially accompanied by target fiber formation. Once reinnervation is complete, the muscle fibers will appear normal on H&E-stained slides; however, myosin heavy chain immunostaining will show large groups of fibers lacking the normal mosaic fiber type pattern, referred to as fiber type grouping. It is not uncommon to encounter occasional degenerating (and regenerating) muscle fibers in patients with denervation, and in long-standing denervation, such as accompanies some hereditary neuropathies, histopathologic features indistinguishable from primary myopathy may be encountered and are often referred to as “pseudomyopathic changes of chronic denervation” (see Fig. 11.1D ). Severely atrophic fibers, having lost all of their sarcomeres, may resemble lymphocyte aggregates on superficial inspection (see Fig. 11.1E ). In infantile spinal muscular atrophy, denervated fibers are rounded rather than angulated and may resemble macrophages (see Fig. 11.1F ).

DENERVATION ATROPHY—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Groups of small angulated (adult) or rounded (infant) fibers

  • n

    Bimodal fiber size distribution

Histochemical Findings

  • n

    ATPase: small group atrophy, fiber type grouping

  • n

    Target fibers (variable)

  • n

    Oxidative enzyme stains: confirm findings on ATPase

Immunohistochemical Findings

  • n

    Myosin heavy chains: small group atrophy, fiber type grouping

  • n

    CD56: acutely denervated fibers

Ultrastructural Features

  • n

    Redundant basal lamina

Genetics

  • n

    Dependent on cause. Most infantile and childhood cases of motor neuron disease are autosomal recessive (i.e., Werdnig–Hoffmann disease, also known as type 1 spinal muscular atrophy); most adult cases are sporadic or acquired

Pathologic Differential Diagnosis

  • n

    Type 2 fiber atrophy secondary to disuse

  • n

    Type 1 fiber atrophy in some metabolic myopathies

Ancillary Studies

Myosin immunohistochemistry and ATPase histochemistry demonstrate that atrophic fibers within small groups are all of the same fiber type and that small groups of both fiber types are present (although type 2 fiber groups usually predominate because of their greater reliance on phasic neural input results in more rapid atrophy after denervation). After complete reconstitution, the reinnervated fibers assume the fiber type characteristics dictated by the parent neuron, resulting in abnormally large groups of similarly typed fibers (see Fig. 11.1G ). On ultrastructural examination, loss of sarcomeric proteins with preservation of sarcolemmal membranes results in cells with folded, redundant basal lamina.

Differential Diagnosis

It is important to distinguish denervation atrophy from type-specific atrophy encountered commonly in disuse (type 2 fiber atrophy) or uncommonly in some primary muscle diseases (type 1 fiber atrophy; see Fig. 11.1G–H ). Although myosin immunohistochemistry or ATPase histochemistry is required for a definitive differential diagnosis, type-specific atrophy almost never appears as small group atrophy. It is usually distributed evenly within the muscle. Although much has been written concerning distinguishing targets from cores, it is likely that absolute distinctions are neither possible nor necessary, as the contexts in which the two are encountered provide more distinguishing information than histochemical and ultrastructural analyses. The current consensus is that target fibers are probably a type of core encountered during muscle fiber reinnervation.

Prognosis and Therapy

The prognosis and treatment depend on the specific disease responsible for the loss of muscle innervation. (Many of these diseases are discussed in the second part of this chapter.)

DISORDERS OF NEUROMUSCULAR TRANSMISSION (AUTOIMMUNE MYASTHENIA, MYASTHENIA GRAVIS)

Clinical Features

Myasthenia gravis is an autoimmune disease produced by antibodies against postsynaptic antigens at the neuromuscular junction, which often presents as ptosis and double vision that worsens as the day wears on. A history of fluctuating weakness can usually be elicited, which on further investigation generally represents fatigability with activity rather than fixed weakness. A more rapid onset of symptoms can also occur, where a patient may have first time symptoms that develop over hours to days following a body stressor such as an infection that triggers presentation of this autoimmune condition. The degree of weakness is highly variable, both from day to day and muscle to muscle. Oculobulbar symptoms are often a feature in the majority of patients with primary autoimmune myasthenia gravis. Around 80 % of patients with myasthenia gravis have antibodies directed against the acetylcholine receptor, 5 % present antibodies against muscle-specific tyrosine kinase, and about 15 % are seronegative, although new antibodies are continually being uncovered in this group of patients. Early-onset myasthenia gravis (under the age of 50 at disease onset) is more frequent among females with thymic hyperplasia, while late-onset myasthenia gravis has been associated with thymoma and more severe disease manifestations.

AUTOIMMUNE MYASTHENIA (MYASTHENIA GRAVIS)— FACT SHEET

Definition

  • n

    Acquired autoimmune disease of the (postsynaptic) neuromuscular junction

Prevalence and Location

  • n

    100 to 200 cases per million

  • n

    Oculobulbar symptoms usually predominate

Sex and Age Distribution

  • n

    Female preponderance in children and young adults

  • n

    Male preponderance in patients older than 50 years

Clinical Features

  • n

    Insidious onset of weakness and fatigability beginning in the oculobulbar muscles in more than 50% of patients

  • n

    Acetylcholine receptor antibodies in 50% (ocular) to 90% (generalized) of patients

  • n

    Thymic hyperplasia in 65% of patients

  • n

    Thymoma in 10% of patients

Prognosis and Treatment

  • n

    Cholinesterase inhibitors for symptom control

  • n

    Immunosuppressive therapy

  • n

    Plasma exchange, intravenous immunoglobulin (IVIG) (useful for myasthenic exacerbations and crises, but effects are short lived)

  • n

    Thymectomy for specific patient populations

  • n

    Eculizumab, a complement inhibitor, approved only for acetylcholine receptor positive myasthenia gravis

Pathologic Features

Several nonspecific light microscopic changes have been described in limb muscle biopsies from patients with myasthenia gravis, including type II fiber atrophy, type I fiber atrophy, and grouped fiber atrophy suspected to be secondary to functional denervation at the motor endplate. Large collections of lymphocytes (lymphorrhages) within muscle have been reported, which is believed to be more likely in biopsies taken early in the disease process and/or prior to the initiation of corticosteroid therapy. A recent study of nearly 1000 patients with myasthenia gravis found evidence of “clinically defined” inflammatory myopathy in less than 1 % of the study subjects.

AUTOIMMUNE MYASTHENIA (MYASTHENIA GRAVIS)—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Nonspecific fiber atrophy

  • n

    Lymphorrhages (very rare)

Histochemical Findings

  • n

    Fiber type or group atrophy

Ultrastructural Features

  • n

    Routine electron microscopy normal

Genetics

  • n

    Sporadic

Immunohistochemical Features

  • n

    Immunoelectron microscopy of neuromuscular junctions may demonstrate reduced density of acetylcholine receptors

Pathologic Differential Diagnosis

  • n

    Type II atrophy secondary to corticosteroid therapy

  • n

    The clinicopathologic differential diagnosis includes Lambert–Eaton myasthenic syndrome and botulism

Ancillary Studies

Immunoelectron microscopic analysis of neuromuscular junctions may demonstrate inaccessibility of acetylcholine receptors.

Prognosis and Therapy

In patients with myasthenia gravis in the absence of cancer, the prognosis is favorable. Cholinesterase inhibitors (e.g., pyridostigmine) are used for symptom relief. Thymectomy may lead to improvement in patients and decrease the overall amount of immunosuppressive needed in managing symptoms. The mainstay of therapy is immunosuppression (steroids and long-term immunosuppressants) and more recently eculizumab for refractory acetylcholine receptor positive myasthenia gravis, with plasma exchange and IVIG used in the treatment of myasthenic exacerbations and crises.

DISORDERS OF EXCITATION–CONTRACTION COUPLING (MUSCLE CHANNELOPATHIES)

PERIODIC PARALYSIS

Clinical Features

The periodic paralyses are autosomal dominant neuromuscular disorders associated with mutations in skeletal muscle sodium, calcium, and potassium channels. Attacks of muscle paralysis are caused by sarcolemmal depolarization resulting in sodium channel inactivation with reduced fiber excitability. These are often triggered by behavior or diet and may last minutes to hours or days. Certain conditions are associated with alterations in serum potassium levels. Among the periodic paralyses, the most common is hypokalemic periodic paralysis, with an estimated prevalence of 1 per 100,000 individuals; it is associated with mutation and calcium channels in about 60 % of families and with sodium channels in about 20 %. It usually starts in childhood, with attacks of weakness typically lasting hours to days, often triggered by carbohydrate ingestion. In hyperkalemic periodic paralysis, also associated with sodium channel mutations, attacks typically occur after (not during) exercise and do not last as long (minutes to hours). Ocular and respiratory muscles are usually spared. Although the frequency of attacks in both types of periodic paralysis tends to decrease with age, many patients develop permanent weakness associated with vacuolar muscle fiber degeneration. Fatal cardiac arrhythmias may complicate paralytic attacks in some individuals.

PERIODIC PARALYSIS—FACT SHEET

Definition

  • n

    Episodic muscle weakness caused by intermittent inexcitability of muscle fibers secondary to ion channel dysfunction

Incidence and Location

  • n

    ∼1 case per million per year

  • n

    Generalized limb weakness/paralysis

Sex and Age Distribution

  • n

    No sex predilection

  • n

    Generally presents in childhood or adolescence

Clinical Features

  • n

    Hypokalemic:

    • Weakness lasting 12 to 24 hours

    • Often precipitated by carbohydrate ingestion

  • n

    Hyperkalemic:

    • Weakness lasting minutes to hours

    • Often precipitated by rest after exercise

    • Cold-induced myotonia may occur

    • Frequency of attacks diminishes with age

Prognosis and Treatment

  • n

    The hypokalemic variant frequently eventuates in degenerative myopathic changes with permanent residual weakness

  • n

    Treatment is aimed at preventing tuition in serum potassium concentration

PERIODIC PARALYSIS—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    H&E staining results may be normal or may show vacuolar changes within muscle cells

Histochemical Findings

  • n

    Tubular aggregates appear as collections of red subsarcolemmal granules on trichrome stain

Ultrastructural Features

  • n

    Proliferation and dilatation of the sarcoplasmic reticulum and T-tubular system

  • n

    Tubular aggregates may be seen in otherwise unaffected fibers

Genetics

  • n

    Autosomal dominant, with reduced penetrance in females

  • n

    Hypokalemic: CACNA1S (1q32) calcium channel; SCN4A (17q23) sodium channel; KCNE3 (11q13-14) potassium channel

  • n

    Hyperkalemic: SCN4A (17q23) sodium channel

Pathologic Differential Diagnosis

  • n

    Tubular aggregates:

    • Other channelopathies, including myotonia and paramyotonia congenita; malignant hyperthermia

    • Alcoholic myopathy

    • Congenital myasthenic syndromes

    • Familial tubular aggregate myopathy

  • n

    Vacuolar degeneration:

    • Nonspecific autophagic (lysosomal) vacuoles

    • Drug-induced vacuolar degeneration

    • Lysosomal storage disorders

Pathologic Features

Except for patients in whom fixed weakness with vacuolar changes has developed, muscle biopsy specimens usually appear normal by H&E staining. Occasionally, tubular aggregates may be visible as bluish granular cytoplasmic inclusions ( Fig. 11.2A ).

Figure 11.2, A , Though not common, aggregates of bluish granular material ( arrows ) may be seen in muscle biopsies of patients with periodic paralysis. B , Trichrome staining enhances the appearance of tubular aggregates ( arrows ), which appear reddish-purple, similar to mitochondrial proliferations. Unlike the latter, these inclusions do not demonstrate cytochrome oxidase activity. C , Tubular aggregates demonstrate a distinctive ultrastructural appearance, usually including both membranous whorls and honeycomb arrays. D , Type 1 myotonic dystrophy shows diffuse type 1 fiber atrophy accompanied by large numbers of internally displaced nuclei.

Ancillary Studies

Histochemistry

Trichrome staining may disclose intermyofibrillary or subsarcolemmal aggregates of red-staining granular material (see Fig. 11.2B ). Although these aggregates may occasionally resemble “ragged red” fibers seen in mitochondrial disorders, the two processes can be distinguished by cytochrome oxidase staining, which highlights mitochondrial proliferations but shows negative staining of tubular aggregates.

Ultrastructural Features

Tubular aggregates appear as densely packed tubules arising from the sarcoplasmic reticulum, usually in a subsarcolemmal location (see Fig. 11.2C ).

Other

Specific sodium, potassium, and calcium channel mutations are associated with the periodic paralyses. The potassium level at initial evaluation, as well as the presence or absence of associated clinical findings or a family history thereof, provides a useful guide to which mutations are likely responsible. The presence of a channel protein mutation confirms the diagnosis.

Differential Diagnosis

Neither vacuolar degeneration nor tubular aggregates are pathognomonic for periodic paralysis, although they help substantiate the diagnosis in clinically suspicious cases. Tubular aggregates may be seen in association with other channelopathies, including myotonic disorders such as myotonia congenita and paramyotonia congenita and in malignant hyperthermia. They have also been reported in a variety of acquired myopathies associated with systemic disease, as well as alcoholic myopathy and congenital myasthenic syndromes, and constitute the diagnostic hallmark of familial tubular aggregate myopathy. Autophagic vacuolar degeneration may also occur as a nonspecific finding in a variety of myopathies and is commonly seen as a manifestation of lysosomal toxicity associated with drugs such as colchicine, chloroquine, and amiodarone. T-tubule–associated vacuoles identical to those of periodic paralysis may be seen in drug-related hypokalemia, such as may occur with diuretics, laxatives, and amphotericin B.

Prognosis and Therapy

Progression to permanent fixed weakness with myopathic changes occurs in a minority of patients and is more common with the hypokalemic variant. Treatment of acute attacks depends on the serum potassium level. Patients with hyperkalemic paralysis may respond to a single dose of acetazolamide and eating high-carbohydrate foods, whereas oral potassium is the treatment of choice for hypokalemic attacks. Dichlorphenamide, also an oral carbonic anhydrase inhibitor, is also available to treat primary hyperkalemic and hypokalemic periodic paralysis.

MYOTONIC DYSTROPHY, TYPE 1

Clinical Features

Myotonia is defined as delayed relaxation after muscle contraction. Patients often complain of stiffness that improves after repeated contractions (“warm-up phenomenon”). Myotonic dystrophy type 1 is unusual among the primary muscle diseases in that patients generally present with distal rather than proximal extremity weakness and wasting. Finger flexor weakness, similar to that seen in patients with inclusion body myositis, is often prominent. Temporalis muscle wasting with ptosis leads to a distinctive “hatchet face” appearance. Myotonic discharges on electromyography support the diagnosis, which can be confirmed by genetic testing. Although patients classically come to medical attention in early adulthood, the age at diagnosis and the severity of the disease correlate with the size of an unstable trinucleotide repeat expansion. Type I myotonic dystrophy (DM1) shows striking changes in presentation over generations, with age at onset typically decreasing by 20 to 30 years per generation progressing from the mild late-onset form to congenital cases in only three generations. Infants with large (greater than 1000 repeats) expansions present with neonatal hypotonia. These severe congenital cases nearly always arise from trinucleotide expansion during oogenesis within an affected mother, although rare cases of large spermatogenic expansion have been reported. In addition, age-dependent somatic expansion markedly complicates genotype phenotype correlations.

Additional clinical features of DM1 include heart block, cardiomyopathy, cataracts, hypersomnia, low IQ, gonadal atrophy, diabetes, frontal balding (in both males and females), swallowing difficulties, and constipation.

MYOTONIC DYSTROPHY (TYPE 1)—FACT SHEET

Definition

  • n

    Multisystem disease characterized by myotonia, progressive muscle wasting, and a range of systemic manifestations

Incidence and Location

  • n

    5–200 cases per million population among different ethnic populations

  • n

    Facial and distal muscle weakness predominates

Sex and Age Distribution

  • n

    No sex predominance

  • n

    Typically develops in the third or fourth decade; 10% present with neonatal hypotonia

Clinical Features

  • n

    Weakness and wasting of facial muscles with ptosis

  • n

    Extremity weakness and atrophy begin distally

  • n

    Clinical myotonia with myotonic discharges on electromyography

  • n

    Heart block, cardiomyopathy, cataracts, hypersomnia, diminished intellect, gonadal atrophy, frontal balding, dysphagia, constipation, and diabetes mellitus may develop

Prognosis and Treatment

  • n

    Diaphragmatic weakness may lead to respiratory failure

  • n

    Cardiac arrhythmias may lead to sudden death

  • n

    Early pacemaker insertion is critical; respiratory therapy is supportive

Pathologic Features

Routine light microscopy reveals striking internalized nuclei within most (but not all) muscle fibers (see Fig. 11.2D ). Longitudinal sections may show distinctive chains of nuclei within the central region of myofibers. Angular atrophic type 1 fibers are present throughout the muscle, often in association with hypertrophic type 2 fibers. Dystrophic features supervene in advanced cases.

In infants with neonatal hypotonia, the histopathologic features are nonspecific, usually type 1 fiber predominance and hypotrophy and common occurrence of type I2B fiber deficiency. Before the molecular genetic era, neonatal diagnosis was accomplished by biopsy of the baby’s mother, in whom diagnostic features are usually present even in the absence of symptoms.

MYOTONIC DYSTROPHY (TYPE 1)—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Internalized nuclei in a majority of fibers

  • n

    Widespread fiber atrophy without grouping

Histochemical Findings

  • n

    Selective type 1 fiber atrophy and internalized nuclei

Ultrastructural Features

  • n

    Nonspecific

Genetics

  • n

    CTG repeat expansion involving the DMPK gene (chromosome 19) in nearly all cases

Pathologic Differential Diagnosis

  • n

    When present in full, the histologic features are virtually pathognomonic

  • n

    In neonatal cases, the histology may be nonspecific

  • n

    In advanced cases, dystrophic features supervene

Ancillary Studies

Histochemistry/Immunohistochemistry

Myosin immunostaining or ATPase histochemical staining shows that the atrophic fibers and fibers with centralized nuclei are type 1.

Other

Genetic analysis is the preferred diagnostic modality and demonstrates CTG expansion within the DMPK gene on chromosome 19.

Differential Diagnosis

Although both internalized nuclei and muscle fiber atrophy may be seen in a wide variety of muscle diseases, the high proportion of muscle fibers with centralized nuclei and preferential type 1 fiber atrophy is unique. Severe neonatal/infantile cases may be difficult or impossible to distinguish from early-onset centronuclear (myotubular) myopathies without genetic testing. Myotonic dystrophy type 2 (DM2), also known as proximal myotonic myopathy, is a more recently described genetic disease that has many clinical similarities to DM1 but different gene expansion (untranslated CCTG repeat on chromosome 3).

Prognosis and Therapy

The severity of the disease correlates directly with the size of the trinucleotide repeat expansion. The most common cause of death is respiratory failure secondary to pneumonia, diaphragmatic weakness, or both. Cardiac arrhythmias, usually secondary to atrioventricular block, are the second leading cause of death. Although indications for pacemaker placement are not clearly defined, abnormal PR intervals or documented dysrhythmias (or both) should prompt referral to a cardiologist.

MYOTONIC DYSTROPHY, TYPE 2

Clinical Features

Also referred to as proximal myotonic myopathy, DM2 often presents in middle-aged adults with slowly progressive muscular weakness similar to that seen in many other myopathic diseases. Muscle pain is commonly present. To complicate matters further, myotonia may be both subtle and fluctuating, and patients may show hypertrophy of calf muscles. The typical facial wasting seen in DM1 is not present, and cataracts may be missed or not felt to be connected with the muscle weakness.

MYOTONIC DYSTROPHY (TYPE 2)—FACT SHEET

Definition

  • n

    Multisystem disease characterized by myotonia, progressive muscle wasting, and systemic manifestations

Incidence and Location

  • n

    Prevalence estimated at 10 per million population but is probably much higher

  • n

    Proximal muscle weakness predominates

Sex and Age Distribution

  • n

    No sex predominance

  • n

    Median onset at 40 years of age, no neonatal form

Clinical Features

  • n

    Proximal muscle pain, weakness, and atrophy

  • n

    Clinical and electrical myotonia are mild and inconsistent

  • n

    Calf hypertrophy

  • n

    Heart block, cardiomyopathy, cataracts, hypersomnia, diminished intellect, gonadal atrophy, frontal balding, dysphagia, constipation, and diabetes mellitus may develop

Prognosis and Treatment

  • n

    Disease severity is highly variable

  • n

    Usually milder course than DM1

  • n

    Cardiac arrhythmias may lead to sudden death

Pathologic Features

DM2 appears to be a disease of type 2 fibers, which have a wide range of sizes ranging from hypertrophy to terminal atrophy and preferentially demonstrate internalized nuclei (as opposed to the preferential internalization of nuclei within atrophic type 1 fibers in DM1). The inconsistent presence of tubular aggregates and/or ring fibers (see Fig. 11.2E ) is a characteristic shared with DM1. Nuclear clumps are frequently seen in patients with DM2.

MYOTONIC DYSTROPHY (TYPE 2)—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Internalized nuclei

  • n

    Increased variation in fiber size, including scattered terminally atrophic fibers (nuclear clumps)

Histochemical Findings

  • n

    Selective type 2 fiber atrophy and internalized nuclei

Ultrastructural Features

  • n

    Nonspecific

Genetics

  • n

    CCTG repeat expansion involving the zinc finger protein 9 gene

Pathologic Differential Diagnosis

  • n

    May appear nearly normal or resemble denervation atrophy

Ancillary Studies

The disease locus for DM2 is on chromosome 3 and has an unstable tetranucleotide repeat expansion within the zinc finger protein 9 gene. Extreme variability in repeat size leads to technical difficulties in genotyping, which can be time consuming and expensive. A higher incidence of mutations in chloride and sodium channel genes has also been reported in patients with DM2, leading to a more severe phenotype at an earlier age of disease onset, and it has been suggested that many patients with neuromuscular disorders of undetermined origin may be suffering from DM2 due to subtleties and complexities of clinical, pathologic, and molecular diagnoses.

Differential Diagnosis

In our experience, most patients who have diagnosis of DM2 have undergone multiple biopsies read either as normal or as showing nonspecific atrophic features. Careful attention to selective involvement of type 2 fibers both with regard to nuclear internalization and atrophy should lead to communication with clinicians regarding the possibility of DM2, along with consideration of genetic testing.

Prognosis and Therapy

DM2 generally progresses slowly and does not occur as a congenital myopathy. As with DM1, however, cardiac arrhythmias may occur, and thus preventative measures may be lifesaving. In addition, a correct diagnosis of DM2 may avoid the inconvenience and expense of repeated muscle biopsies.

CORE MYOPATHIES

Clinical Features

The core myopathies are the most prevalent subgroup of congenital myopathies, with clinical manifestations ranging from apparent normalcy to severe weakness resulting in the need for ambulatory assistance. Genetic variants causing core myopathies primarily affect proteins involved in excitation-contraction coupling, either by altering calcium transits between the sarcoplasmic reticulum and sarcoplasm or by disrupting the structure of the sarcomere. Ineffective excitation-contraction coupling causes muscle weakness and is associated with the formation of mitochondria depleted core lesions. Typical presentations include infant hypotonia and developmental delay in early childhood, with prominent involvement of limb and axial muscles. Initially, the weakness tends to be slowly progressive or static, but often improves with time (which may be responsible for an appearance of anticipation). Orthopedic abnormalities are common, particularly scoliosis and congenital hip dislocation. In typical ryanodine receptor 1 ( RYR1 ) mutation-associated central core disease (CCD), respiratory weakness and cardiomyopathy are rarely encountered. Malignant hyperthermia is highly associated with RYR1 mutation-associated central core and multi-minicore disease independent of the degree of muscle weakness. Patients with SEPN1 multi-minicore disease commonly manifest severe respiratory weakness and more severe scoliosis than patients with RYR1 -associated diseases.

CORE MYOPATHIES—FACT SHEET

Definition

  • n

    Congenital myopathy characterized by fibers containing geographic regions devoid of oxidative activity

Incidence and Location

  • n

    ∼40 cases per million children per year; true incidence not known because of wide phenotypic variability

  • n

    Proximal limb weakness with generalized hypotonia and scoliosis

Sex and Age Distribution

  • n

    No sex predilection reported

  • n

    Often clinically apparent in infancy, but may not manifest until adulthood

  • n

    Earlier onset associated with more severe course

Clinical Features

  • n

    Slowly progressive or nonprogressive weakness with hypotonia

  • n

    Often develops in association with skeletal abnormalities (hip dislocation, scoliosis)

  • n

    Intrafamilial phenotypic variability

  • n

    Respiratory insufficiency may be severe

  • n

    Highly associated with malignant hyperthermia

Prognosis and Treatment

  • n

    Functional improvement is common even in severe cases

  • n

    Treatment is supportive and preventative (avoidance of stress, including medications that might trigger episodes of malignant hyperthermia)

Pathologic Features

Core myopathies are (along with nemaline myopathy, see following) prototypical histochemically defined myopathies. Routinely stained muscle may demonstrate subtle cytoplasmic abnormalities (see Fig. 11.2F ). In addition, type 1 fiber atrophy is characteristically seen in RYR1 -related CCD. More severe cases of multi-minicore disease may demonstrate nonspecific myopathic features.

CORE MYOPATHIES—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Often normal but may demonstrate degenerative myopathic features in more severe and/or long-standing cases

Histochemical Findings

  • n

    ATPase/myosin immunohistochemistry: type 1 fiber predominance (or even uniformity)

  • n

    Oxidative enzyme stains: central or peripheral cores, multiple minicores, or no cores with uniform staining

Ultrastructural Features

  • n

    Areas devoid of mitochondria, either with preservation (structured core) or disruption (unstructured core) of sarcomeres

Genetics

  • n

    Autosomal dominant

  • n

    Linked to mutations in ryanodine receptors ( RYR1 , with 50% of cases demonstrating mutations in exons 93 to 104) and selenoprotein N ( SEPN1 )

Immunohistochemical Features

  • n

    Cores contain a wide variety of proteins, including desmin and filamin

Pathologic Differential Diagnosis

  • n

    Cores have been reported in hypothyroid myopathy

  • n

    Cores may closely resemble targets seen in neurogenic processes

Ancillary Studies

Histochemistry

CCD is named for sarcoplasmic regions devoid of oxidative enzyme activity (see Fig. 11.2G ). A second common finding is marked predominance (or even uniformity) of type 1 muscle fibers. The morphologically related, but often genetically distinct, multi-minicore disease demonstrates multiple smaller regions of absent oxidative activity within muscle fibers.

Ultrastructural Features

As suggested by their histochemical features, central cores are regions devoid of mitochondria. When the myofibrillary architecture is preserved in these areas, the cores are referred to as structured. When mitochondrial absence is accompanied by myofibrillary dissolution, the core is referred to as unstructured. The significance of such core subtypes has not yet been determined. Central cores extend longitudinally along the full length of the sarcomere; multi-minicores do not.

Other

Dominantly inherited mutations in the ryanodine receptor gene ( RYR1 ) are encountered in more than 50 % of patients with CCD. Such mutations are rare in multi-minicore disease, which has been associated with mutations in a gene associated with rigid spine muscular dystrophy ( SEPN1 ).

Differential Diagnosis

Central cores and multi-minicores must be distinguished from target fibers accompanying denervation atrophy. Such distinction is more easily described than accomplished (targets are supposed to show peripheral increases in ATPase and oxidative enzyme staining [ Fig. 11.2H ]). A diagnosis of core disease should be considered with caution in a patient showing other pathological features suggesting denervation. In the absence of clinical weakness, the mere presence of central cores is not sufficient for a diagnosis of CCD. Multi-minicores do not extend longitudinally along the muscle fiber but tend to occur as circumscribed regions devoid of oxidative enzyme activity.

Prognosis and Therapy

Functional improvement is common even in severe cases, arguing for aggressive supportive therapy and avoidance of stresses that might trigger episodes of malignant hyperthermia. Recent studies of SEPN1 -related myopathy have implicated oxidative stress as the primary factor responsible for muscle impairment, which may portend effective antioxidant therapies.

DISORDERS OF THE SARCOMERE

NEMALINE MYOPATHY

Clinical Features

Nemaline myopathy was one of the first congenital–structural myopathies described and is a well-known cause of infantile hypotonia (the “floppy infant”). In these babies, prominent facial and respiratory weakness helps distinguish nemaline myopathy from the many other causes of infantile hypotonia. Nemaline myopathy may be caused by mutations in at least 13 genes, and some cases are still molecularly unresolved. The most frequent genetic cause of autosomal recessive nemaline myopathy is a mutation in the nebulin gene, whereas heterozygous pathogenic variants in the α-skeletal actin gene are the most prevalent cause of autosomal dominant nemaline myopathy. Clinical phenotypes range from severe neonatal forms expressing the fetal akinesia sequence to sporadic late-onset nemaline myopathy (SLONM) with isolated neck extensor weakness (the dropped-head sign), dysphagia, and/or respiratory insufficiency. Thus the pathologist must always peruse the muscle biopsy specimen for these sometimes subtle, but distinctive, cytoplasmic structures. This especially applies to biopsies containing very small fibers, as rods may only be apparent under high power magnification or in plastic-embedded sections.

NEMALINE MYOPATHY—FACT SHEET

Definition

  • n

    Myopathy with rod-shaped structures within muscle fibers (Greek nema = thread)

Incidence and Location

  • n

    ∼2 cases per million people per year

  • n

    Proximal weakness predominates

Sex and Age Distribution

  • n

    No sex predominance noted

  • n

    Onset from birth through adulthood; most become clinically apparent in the neonatal period or during childhood

Clinical Features

  • n

    Classic: floppy infant with facial and respiratory weakness

  • n

    Range: severe congenital forms with fetal akinesia and neonatal respiratory insufficiency to adult-onset forms with head drop, dysphagia, and/or respiratory insufficiency

Prognosis and Treatment

  • n

    Earlier onset cases are usually more severe

  • n

    Overall mortality is ∼20%, with death caused by respiratory insufficiency during the first year of life

  • n

    Aggressive supportive therapy may result in clinical stabilization with age

Pathologic Features

Abnormal variation in fiber size is often present, and atrophy or hypotrophy of type I muscle fibers is common. There may also be hypertrophy of type II fibers, resulting in fiber type disproportion. Careful examination of atrophic fibers may reveal focal regions of increased refractivity ( Fig. 11.3A ).

NEMALINE MYOPATHY—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Atrophic fibers without grouping; may contain small subsarcolemmal areas of increased refractivity

Histochemical Findings

  • n

    Trichrome: reddish-purple thread-like inclusions within the sarcoplasm, often within atrophic fibers

  • n

    Oxidative enzymes: areas with rods may appear core-like

  • n

    ATPase: type 1 fiber predominance and atrophy/hypotrophy

Ultrastructural Features

  • n

    Intracytoplasmic (and rarely intranuclear) rod-shaped structures composed of Z-band–like material, often accompanied by sarcoplasmic disruption

Genetics

  • n

    At least 13 different genetic diseases, including autosomal recessive, autosomal dominant, and de novo autosomal dominant

Immunohistochemical Features

  • n

    Sarcoplasmic rods react with antibodies to αβ-actinin

Pathologic Differential Diagnosis

  • n

    Nemaline bodies may occur at normal myotendinous junctions, in normal extraocular muscles, in aging muscle, and occasionally in a variety of other inherited or acquired neuromuscular disorders, including CCD and inflammatory myopathies

Figure 11.3, A , Similar to cores, nemaline bodies ( arrows ) may be observed on routine H&E staining of muscle biopsies, but they are easily overlooked. B , Trichrome staining stains nemaline rods purple ( arrows ), which stand out against the otherwise pale green cytoplasm of the muscle fibers. C , Acute quadriplegic myopathy is characterized by selective loss of thick filaments, with relative preservation of Z-discs and thin filaments. D , Normal ultrastructure for comparison.

Ancillary Studies

Histochemistry

Although nemaline myopathy was first described on formalin-fixed, paraffin-embedded sections, the thread-like inclusions are much more easily seen on trichrome histochemical staining of frozen sections, where they show up as purple rods against the light blue-green color of the muscle cytoplasm (see Fig. 11.3B ). The proportion of muscle fibers containing these structures varies greatly from case to case and does not correlate with clinical severity or genotype. Nuclear rods are seen only in patients with actin gene mutations, which tend to present with severe early-onset disease.

Ultrastructural Features

Electron microscopy frequently reveals striking osmiophilic rod-shaped inclusions, often in subsarcolemmal and perinuclear locations. Rarely, intranuclear inclusions are identified and signify a worse prognosis. Sarcomeric disruption is also usually seen in patients with severe weakness.

Immunohistochemistry

Immunohistochemical analysis has demonstrated a wide variety of skeletal muscle proteins within nemaline rods, although the most constantly stained protein is α-actinin.

Differential Diagnosis

Nemaline rods may occur at normal myotendinous junctions, in normal extraocular muscles, in aging muscle, and occasionally in a variety of other inherited or acquired neuromuscular disorders, including CCD and inflammatory myopathies.

Prognosis and Therapy

The classic neonatal form of the disease carries a 20 % mortality rate, usually secondary to respiratory failure during the first year of life. The other 80 % of these infants show clinical stabilization or even improvement, thus underscoring the value of aggressive supportive therapy. Later onset cases tend to be less severe, but SLONM may be associated with severe disability and even death. There is currently no curative treatment, but supportive management aimed at maintaining muscle strength, mobility, joint movements, and independence in activities of daily living to exercise and physiotherapy is important, as is regular monitoring of respiratory function and orthopedic problems, especially scoliosis.

ACUTE QUADRIPLEGIC MYOPATHY

Clinical Features

To date, acute quadriplegic myopathy has been reported exclusively in the critical care setting, which explains its alternative name: critical illness myopathy. This myopathy is usually recognized when a critically ill patient cannot be weaned off the ventilator because of diffuse flaccid paralysis, including paralysis of the diaphragmatic and intercostal muscles. Two of the three following are usually required to cause this disorder: corticosteroid therapy, neuromuscular blockade, and severe systemic illness such as sepsis.

ACUTE QUADRIPLEGIC MYOPATHY—FACT SHEET

Definition

  • n

    Acute weakness developed in the intensive care setting

Incidence and Location

  • n

    ∼30% of critically ill patients undergoing mechanical ventilation for longer than 1 week; may exceed 70% in some subgroups with extended ICU treatment

  • n

    More likely to occur with sepsis, corticosteroid therapy, or both

  • n

    Proximal weakness predominates and may be asymmetric

Sex and Age Distribution

  • n

    Female preponderance

  • n

    Mean age of 65 years, with a wide distribution

Clinical Features

  • n

    Rapid onset of diffuse, sometimes asymmetric, weakness and muscle wasting that may lead to difficulty in ventilatory weaning

Prognosis and Treatment

  • n

    Neuromuscular dysfunction resolves within 3 weeks in approximately half of patients; in the majority of the remaining patients, it resolves within a year

  • n

    Aggressive rehabilitation therapy may facilitate recovery. Prevention is recommended through restriction of corticosteroid therapy in critically ill patients to disorders in which they have been clearly demonstrated to significantly improve patient outcome

Pathologic Features

Routinely stained sections may show features of rhabdomyolysis but more often demonstrate slight to prominent muscle fiber atrophy without necrosis.

ACUTE QUADRIPLEGIC MYOPATHY—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Minimal to marked atrophy of muscle fibers

  • n

    Necrotic fibers may be present

Histochemical Findings

  • n

    ATPase: may demonstrate focal or general loss

  • n

    Trichrome: fibers may appear more purple than green

  • n

    Oxidative: may show central pallor

Ultrastructural Features

  • n

    Selective loss of thick (myosin) filaments (A-band loss)

Immunohistochemical Features

  • n

    Deficient myosin heavy chains within pale zones

Pathologic Differential Diagnosis

  • n

    Light microscopic changes are nonspecific; electron microscopic features are diagnostic in the right clinical setting

  • n

    Selective myosin loss may be seen in myotubular myopathy, in infantile cytochrome oxidase deficiency, and occasionally in chronically denervated fibers

Ancillary Studies

Histochemistry

Muscle fibers may show central pallor on all histochemical stains.

Ultrastructural Features

A striking selective loss of thick filaments is seen within atrophic fibers (see Fig. 11.3C–D ).

Immunohistochemistry

Decreased myosin heavy chain immunostaining may be observed within affected fibers.

Differential Diagnosis

Clinically, critical illness myopathy may be indistinguishable from critical illness polyneuropathy. Light microscopic findings are nonspecific, but the ultrastructural features demonstrating preferential myosin loss without generalized myofibrillar breakdown are virtually pathognomonic. Depletion of myosin heavy chains may occasionally be encountered in chronic denervation and has been reported in myotubular myopathy and infantile cytochrome oxidase deficiency.

Prognosis and Therapy

On cessation of corticosteroid therapy or neuromuscular blockade (or both), recovery of muscle strength occurs within 3 weeks in approximately 50 % of patients; the majority of the remaining patients regain normal strength within a year. Treatment is supportive. Prevention is recommended and is achieved through the restriction of corticosteroid use in critically ill patients for disorders in which the corticosteroids show a clear and significant improved patient outcome.

LATE-ONSET MYOFIBRILLAR MYOPATHY/Z-DISC DISEASES

Clinical Features

As the name implies, these diseases usually do not show symptomatology until after the age of 40, and most affected patients do not come to medical attention until they are in their 50s, 60s, or even older—in other words, the same age as patients suffering from inclusion body myositis. To further complicate the differential diagnosis, patients with myofibrillar myopathies often manifest predominantly distal weakness, though more often in their lower extremities (unlike patients with inclusion body myositis, where distal weakness is usually confined to upper extremities). Proximal muscles may be affected, as exemplified by the integral Z-disc protein, myotilin, the mutation of which underlies both late-onset myofibrillar myopathy and limb-girdle muscular dystrophy 1A (LGMD1A). Depending on the specific Z-disc protein involved, families with late-onset myofibrillar myopathies may manifest cardiac symptoms as well as peripheral neuropathy.

LATE-ONSET MYOFIBRILLAR MYOPATHY—FACT SHEET

Definition

  • n

    A group of myopathies characterized by myofibrillar disorganization secondary to mutations involving Z-disc proteins

Incidence and Location

  • n

    Rare; exact incidence is currently unknown

  • n

    May present with proximal or distal weakness, usually starting in lower extremities

  • n

    Cardiac symptoms may present before skeletal muscle weakness

Sex and Age Distribution

  • n

    No known sex predilection

  • n

    Typical onset is after age 40

Clinical Features

  • n

    Highly variable

  • n

    Serum creatine kinase levels may be normal or slightly elevated

  • n

    May include cardiac complications, respiratory insufficiency, and peripheral neuropathy

  • n

    Other findings may include cataracts, scoliosis, rigid spine syndrome, contractures, or nail dystrophy

Prognosis and Treatment

  • n

    Typically slowly progressive

  • n

    Evaluation for cardiomyopathy

  • n

    Supportive care

Pathologic Features

Routine sections generally demonstrate myopathic features, including fiber size variability, internalized nuclei, degeneration and regeneration of muscle fibers, and mild endomysial fibrosis. Vacuoles, both rimmed and nonrimmed, and sarcoplasmic aggregates may be seen within scattered muscle fibers (see Fig. 11.3E ). Small group atrophy may be seen in patients with associated peripheral neuropathy.

LATE-ONSET MYOFIBRILLAR MYOPATHY—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Myopathic features: fiber size variability, internalized nuclei, degeneration and regeneration of muscle fibers, endomysial fibrosis

  • n

    Sarcoplasmic aggregates

  • n

    Rimmed and nonrimmed vacuoles may be present

Histochemical Findings

  • n

    Trichrome may show red or blue sarcoplasmic aggregates and red granular staining around vacuoles

  • n

    Sarcoplasmic aggregates may stain with Congo red

  • n

    Aggregates react with anti-desmin antibodies, among others

Ultrastructural Features

  • n

    Myofibrillar disorganization

  • n

    Z-disc streaming

  • n

    Filamentous inclusions

  • n

    Spheroid bodies highly suggestive of myotilinopathy

Genetics

  • n

    Several genes have been implicated

  • n

    A large percentage of cases lack demonstrable genetic abnormalities

Pathologic Differential Diagnosis

  • n

    Inclusion body myositis

Ancillary Studies

Histology

Trichrome staining highlights sarcoplasmic aggregates, which may stain blue or red, as well as red granular staining around vacuoles (see Fig. 11.3F ). Sarcoplasmic aggregates may demonstrate congophilia.

Immunohistochemistry

Aggregates and vacuoles may react with a variety of antibodies including desmin.

Electron Microscopy

Ultrastructural examination demonstrates Z-disc streaming and filamentous inclusions (see Fig. 11.3G ).

Other

Although identification of a pathogenic mutation is considered the gold standard for the diagnosis of specific Z-disc diseases, laboratories that perform these tests may still be hard to find.

Differential Diagnosis

The clinical presentation and pathologic features of late-onset myofibrillar myopathies overlap considerably with those of inclusion body myositis. The latter is defined by the presence of inflammation along with atrophy, degeneration, and rimmed vacuoles. In addition, myofibrillar myopathies (with the exception of those secondary to myotilin mutations) do not show intranuclear aggregates or excess cytochrome oxidase–negative fibers. Mitochondrial abnormalities are more commonly seen in inclusion body myositis.

Prognosis and Therapy

Weakness in these diseases tends to be very slowly progressive. Therapeutic efforts are supportive and preventative, especially in families with systemic complications.

DISORDERS OF MECHANICAL TRANSDUCTION (MUSCULAR DYSTROPHIES)

EARLY-ONSET MYOFIBRILLAR MYOPATHY (DESMINOPATHIES)

Clinical Features

The original description of early-onset myofibrillar myopathy secondary to mutations involving desmin included progressive distal or proximal weakness associated with cardiomyopathy. Common autosomal dominant mutations manifest during early and middle adulthood. Rare autosomal recessive mutations have been reported and present during childhood. Clinical manifestations vary widely, but usually include slowly progressive weakness, respiratory insufficiency, and cardiomyopathy with conduction block. Early-onset myofibrillar myopathies resulting from proteins that interact with desmin (such as αβ-crystallin) are increasingly being recognized, but as their neuromuscular manifestations are currently not easily distinguishable from those resulting from desmin mutations, myopathies primarily involving the intermediate filament system of skeletal muscle are usually collectively referred to as desminopathies.

EARLY-ONSET MYOFIBRILLAR MYOPATHY—FACT SHEET

Definition

  • n

    A group of myopathies characterized by myofibrillar disorganization secondary to mutations involving desmin and associated proteins

Incidence and Location

  • n

    Exact incidence is currently unknown

  • n

    Lower extremities, then upper limb weakness followed by axial spread and cardiomyopathy

Sex and Age Distribution

  • n

    No known sex predilection

  • n

    Onset usually during late childhood, though may be earlier

  • n

    Patients with mutations in αβ-crystallin typically demonstrate lenticular opacities

Clinical Features

  • n

    Highly variable; may present with cardiomyopathy

  • n

    Serum creatine kinase levels may be normal or elevated

  • n

    Usually include cardiomyopathy, conduction abnormalities, and respiratory insufficiency

Prognosis and Treatment

  • n

    Typically slowly progressive

  • n

    Evaluation for cardiomyopathy, arrhythmias, and respiratory insufficiency

  • n

    Supportive care

Pathologic Features

Similar to late-onset myofibrillar myopathies, desminopathies show increased fiber size variability, internalized nuclei, rimmed vacuoles, and sarcoplasmic aggregates ( Fig. 11.4A ).

EARLY-ONSET MYOFIBRILLAR MYOPATHY—PATHOLOGIC FEATURES

Microscopic Findings

  • n

    Myopathic features: fiber size variability, internalized nuclei, degeneration and regeneration of muscle fibers, endomysial fibrosis, rimmed vacuoles

Histochemical Findings

  • n

    Trichrome may show red or blue sarcoplasmic aggregates of varying morphologies

  • n

    “Rubbed-out” fibers may be seen on oxidative enzyme stains

  • n

    Aggregates react with anti-desmin antibodies, which also may show plaque-like subsarcolemmal staining

Ultrastructural Features

  • n

    Myofibrillar disorganization

  • n

    Z-disc streaming

  • n

    Granulofilamentous inclusions

Genetics

  • n

    In addition to numerous autosomal dominant and recessive desmin mutations, patients with mutations in αβ-crystallin demonstrate indistinguishable myopathology

  • n

    Many patients have no family history of muscle disease and may also lack demonstrable genetic abnormalities

Pathologic Differential Diagnosis

  • n

    Late-onset myofibrillar myopathies (Z-disc diseases)

Figure 11.4, A , Similar to late-onset myofibrillar myopathies, desminopathies show myopathic features accompanied by a variety of cytoplasmic aggregates ( arrows ). B , A variety of sarcoplasmic inclusions may be seen on trichrome staining. These are often bluish-green and may be discrete or more rarefied ( arrows ). C , Desmin immunohistochemical staining shows both discrete intracytoplasmic inclusions ( lower arrows ) and more diffuse plaque-like subsarcolemmal aggregates ( upper arrows ). D , Classically, muscular dystrophies demonstrate myopathic features accompanied by prominent endomysial and perimysial fibrosis.

Ancillary Studies

Histology and Histochemistry

Trichrome staining highlights sarcoplasmic aggregates (see Fig. 11.4B ). Oxidative enzyme stains frequently demonstrate large areas devoid of oxidative enzyme activity (sometimes referred to as rubbed-out fibers).

Immunohistochemistry

Desmin immunohistochemical staining reveals diffuse or solitary cytoplasmic inclusions, the former often localized to subsarcolemmal regions of the muscle fiber (see Fig. 11.4C ).

Electron Microscopy

Granulofilamentous inclusions, either subsarcolemmal or intermyofibrillary, are characteristic.

Other

Genetic testing is essential to establish an accurate diagnosis.

Differential Diagnosis

In addition to the similarities to late-onset myofibrillar myopathies due to abnormalities in the Z-disc, desminopathies may resemble other myopathies associated with intracytoplasmic aggregates (e.g., nemaline myopathy, mitochondrial myopathy) or loss of oxidative enzyme staining (e.g., core myopathies, mitochondrial myopathies).

Prognosis and Therapy

No specific treatments are currently available, but complications may be preventable. Investigation and treatment of cardiac conduction defects, transplantation for cardiomyopathy, and respiratory support are critical if early mortality is to be avoided.

DUCHENNE MUSCULAR DYSTROPHY

Clinical Features

Duchenne muscular dystrophy becomes clinically apparent in early childhood. Initially, these young boys are clumsy and have trouble running; later, they have difficulty climbing stairs. Hip and knee extensor weakness manifests as the Gowers sign, whereby the child climbs up his thighs with his hands to arise from a lying or sitting position. Although calf hypertrophy is considered to be one of the hallmarks of Duchenne muscular dystrophy, it is not specific. Progressive, mainly proximal, muscle weakness tends to remain relatively stable until the age of about 7 years, when more rapid progression becomes apparent, leading to wheelchair dependence in most boys by the age of 12 years, followed by scoliosis, loss of upper limb function, respiratory insufficiency, and cardiomyopathy.

DUCHENNE MUSCULAR DYSTROPHY—FACT SHEET

Definition

  • n

    X-linked dystrophy of muscle caused by mutations resulting in the absence of dystrophin

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