Inherited Metabolic and Developmental Disorders of the Pediatric and Adult Liver


Introduction

Jaundice, which is observed in almost every newborn, is termed physiological because it clears within a few days, after hepatic activation of bilirubin conjugation. This phenomenon reflects a unique feature of prenatal life: Many functions of the liver that are required after birth for nutrition, metabolic balance, and detoxification and excretion of endogenous chemicals are provided to the developing fetus by the placenta and the mother. “Pathological” jaundice (i.e., extending beyond the usual “physiological” time frame) is the most frequent indication for liver biopsy in children, especially infants because functional immaturity is not limited to glucuronidation; intrinsic defects in many processes and structures lead to cholestasis. This is even more evident in premature infants. Not only do the first challenges to hepatobiliary function account for liver diseases that “adult” pathologists encounter very rarely, but maternal–fetal interactions are not always beneficial. Certain infections and immunologically mediated injuries are observed only in infants.

This chapter focuses on constitutional deficiencies of the liver that necessitate examination of tissue for diagnosis and treatment. Myriad chromosomal imbalances and heritable mutations that manifest with dysmorphism and multisystem disease, such as Down syndrome, may affect the liver but can be diagnosed clinically; they are included in this chapter only if they offer a challenge to diagnosis. Anatomical and synthetic defects, such as clotting factor deficiencies that do not lead to hepatobiliary dysfunction, are covered in other publications. , Against the roster of inherited conditions described in this chapter, it is worth noting the conditions included in United States recommended uniform screening panel for newborns ( Table 55.1 ) ; only a subset of this chapter’s disorders are included. Hence evaluation of hepatic disease in the pediatric population extends well beyond information obtained from the routine constitutional screening performed at the time of birth.

TABLE 55.1
Uniform Screening for the Newborn
Downloaded for Anonymous user (n/a) at North Shore University Hospital at Manhasset from ClinicalKey.com by Elsevier on Nov 25, 2019. From Urv TK, Parisi MA. Newborn screening: beyond the spot. Adv Exp Med Biol . 2017;1031:323–346; Advisory Committee on Heritable Disorders in Newborns and Children. Recommended uniform screening panel. US Department of Health and Human Services. https://www.hrsa.gov/advisory-committees/heritable-disorders/recommendations-reports/reports/index.html .
Recommended Uniform Screening Panel: Core Conditions
Organic Acid Conditions

    • Propionic academia (PROP)

    • Methylmalonic academia (MUT)

    • Methylmalonic academia (Cbl A,B)

    • Isovaleric academia (IVA)

    • 3-Methylcrotonyl-CoA carboxylase deficiency (3MCC)

    • Glutaric academia type I (GA1)

    • 3-Hydroxy-3-methylglutaric aciduria deficiency (HMG)

    • β-Ketothiolase deficiency (BKT)

    • Holocarboxylase synthetase (multiple carboxylase) deficiency (MCD)

Fatty Acid Oxidation Disorders

    • Carnitine uptake defect/carnitine transport defect (CUD)

    • Medium-chain acyl-CoA dehydrogenase deficiency (MCAD)

    • Very-long-chain acyl-CaA dehydrogenase deficiency (VLCAD)

    • Long-chain L-3 hydroxyacyl-CoA dehydrogenase deficiency (LCHAD)

    • Trifunctional protein deficiency (TFP)

Amino Acid Disorders

    • Classic phenylketonuria (PKU)

    • Maple syrup urine disease (MSUD)

    • Homocystinuria (HCY)

    • Tyrosinemia type I (TYR)

    • Arginosuccinic academia (ASA)

    • Citrullinemia type I (CIT)

Endocrine Disorders

    • Primary congenital hypothyroidism (CH)

    • Congenital adrenal hyperplasia (CAH)

Hemoglobin Disorders

    • S,S disease (sickle cell anemia) (Hb SS)

    • S, β-thalassemia (Hb S/bTh)

    • S,C disease (Hb S/C)

Other Disorders

    • Critical congenital heart disease (CCHD)

    • Cystic fibrosis (CF)

    • Classic galactosemia (GALT)

    • Hearing loss (HEAR)

    • Severe combined immunodeficiency (SCID)

Recommended Uniform Screening Panel: Secondary Conditions
Organic Acid Conditions

    • Methylmalonic academia with homocystinuria (Cbl C,D)

    • Malonic academia (MAL)

    • Isobutrylglycinuria (IBG)

    • 2-Methylbutyrylglycinuria (2MBG)

    • 3-Methylglutaconic aciduria (3MGA)

    • 2-Methyl-3-hydroxybutyric aciduria (2M3HBA)

Fatty Acid Oxidation Disorders

    • Short-chain acyl-CoA dehydrogenase deficiency (SCAD)

    • Medium/short-chain L-3-hydroxyacyl-CoA dehydrogenase deficiency (M/SCHAD)

    • Glutaric academia type II (GA2)

    • Medium-chain ketoacyl-CoA thiolase deficiency (MCAT)

    • 2,4 Dienoyl-CoA reductase deficiency (DE RED)

    • Carnitine palmitoyltransferase type I deficiency (CPT-1A)

    • Carnitine palmitoyltransferase type II deficiency (CPT II)

    • Carnitine acylcarnitine translocase deficiency (CACT)

Amino Acid Disorders

    • Argininemia (ARG)

    • Citrullinemia type II (CIT II)

    • Hypermethioninemia (MET)

    • Benign hyperphenylalaninemia (H-PHE)

    • Biopterin defect in cofactor biosynthesis (BIOPT BS)

    • Biopterin defect in cofactor regeneration (BIOPT REG)

    • Tyrosinemia type II (TYR II)

    • Tyrosinemia type III (TYR III)

Hemoglobin Disorders

    • Various other hemoglobinopathies (Var Hb)

Other Disorders

    • Galactoepimerase deficiency (GALE)

    • Galactokinase deficiency (GALK)

    • T-cell–related lymphocyte deficiencies

We have incorporated a practical approach to liver biopsy that is derived from Jevon and Dimmick’s classification of the histological pattern of pediatric liver biopsies, which identify six dominant patterns. Additionally, we emphasize the progress made during the past two decades with regard to decoding the genetic basis of disease that has resulted in improved therapeutics as well as reclassification and renaming of disease entities, genes, and proteins. In the preparation of this chapter, we have benefited from Online Mendelian Inheritance in Man (OMIM; www.omim.org ), an online compendium of human genes and phenotypes maintained by the Johns Hopkins University and developed by the National Center for Biotechnology Information (NCBI). This searchable database provides a unique accession number for each entity and incorporates all alternative disease names and gene nomenclature. Throughout the text and in the tables, we have provided OMIM numbers for heritable conditions to assist readers.

Pediatric Liver Biopsies

Indications for Liver Biopsy in Children

In addition to prior liver or bone marrow transplantation (see Chapter 43 ), the most common indications for liver biopsy in the pediatric age group are conjugated hyperbilirubinemia in young infants ( Table 55.2 ); tumor diagnosis (see Chapter 56 ); and assessment of liver injury, inflammation, and fibrosis. Metabolic diseases may manifest with fetal demise immediately after birth or at any age thereafter ( Table 55.3 ).

TABLE 55.2
Liver Disease in Infants
  • Bile duct stricture

  • Biliary atresia

  • Choledochal cyst

  • Caroli syndrome

  • Alagille syndrome

  • Neonatal infection

    • Cytomegalovirus

    • Herpesvirus

    • Hepatotropic viruses (hepatitis E)

    • Human immunodeficiency virus

    • Parvovirus B19

    • Paramyxovirus

    • Enteric viruses (echoviruses, coxsackieviruses, adenoviruses)

    • Rubella (congenital)

    • Bacterial sepsis

    • Urinary tract infection

    • Listeriosis

    • Toxoplasmosis (Toxoplasma gondii)

    • Syphilis (congenital)

  • Neonatal hemochromatosis

  • Disorders of carbohydrate metabolism

    • Glycogen storage disease, types I and IV

    • Galactosemia

    • Fructosemia

  • Disorders of amino acid metabolism

    • Tyrosinemia

  • Disorders of glycolipid and lipid metabolism

    • Niemann-Pick disease types A, B, and C

    • Gaucher disease

    • Hurler disease

    • Wolman disease

  • Disorders of glycoprotein metabolism

    • Congenital disorder of glycosylation type Ib

  • Peroxisomal disorders

    • Zellweger syndrome

  • Mitochondrial cytopathies, Reye syndrome

  • Urea cycle disorders

  • Hereditary disorders of bilirubin metabolism

    • Crigler-Najjar syndrome

    • Gilbert syndrome

    • Dubin-Johnson syndrome and Rotor syndrome

  • Hereditary disorders of bile formation and transport

    • Progressive familial intrahepatic cholestasis, types 1 to 5

    • Disorders of bile acid biosynthesis

  • Disorders of protein biosynthesis and targeting

    • α 1 -Antitrypsin deficiency

    • Cystic fibrosis

  • Miscellaneous inherited disorders

    • Trisomy 21

    • Aagenaes syndrome

    • Citrullinemia, type II

    • X-linked adrenoleukodystrophy

  • Miscellaneous nonneoplastic conditions

    • Shock/hypoperfusion (as from congestive heart failure in congenital heart disease)

    • Parenteral nutrition

    • Fetal alcohol syndrome

    • Drugs

    • Budd-Chiari syndrome

    • Multiple hemangiomas (with high-output cardiac failure)

  • Idiopathic neonatal hepatitis/syncytial giant cell hepatitis

  • Neonatal sclerosing cholangitis with ichthyosis

  • Neonatal lupus

  • Neoplasia

    • Neonatal leukemia

    • Neuroblastoma

    • Hepatoblastoma

    • Langerhans cell histiocytosis

    • Hemophagocytic lymphohistiocytosis

TABLE 55.3
Presentation of Metabolic Diseases That Involve the Liver
Age Hepatic Failure Encephalopathy (±Bleeding) Jaundice (Hepatitis) Failure to Thrive and/or Hepatomegaly Portal Hypertension (Ascites, Bleeding, Splenomegaly)
(Hypoglycemia) (Normal Sugar)
Newborn Galactosemia, mitochondriopathies, urea cycle defects, glutaric aciduria II Crigler-Najjar syndrome type I Leprechaunism, fructose 1.6 diphosphatase deficiency
First 2 months Wolman disease, tyrosinemia, perinatal hemochromatosis α 1 -Antitrypsin deficiency, NPD type C GSD 1a, Ib Zellweger syndrome GSD IV
First 6 months Hereditary fructose intolerance, LCAD deficiency, carnitine deficiency, propionic acidemia Byler disease, Alagille syndrome, THCA, 3β-HSD, isomerase deficiency GSD III Lysinuric protein intolerance, MPS, other storage diseases
First 2 years MCAD deficiency, mitochondriopathies Cystic fibrosis, Rotor syndrome GSD VI and IX, congenital disorder of glycosylation type Ib, glycoprotein
Up to 6 years Cholesterol ester storage, NPD types A and B, cystinosis, hereditary fructose intolerance
Puberty/and adolescence Wilson disease, erythropoietic porphyria Gilbert syndrome, Wilson disease, Dubin-Johnson syndrome α 1 -Antitrypsin deficiency, Wilson disease, lipoatrophic diabetes
Adults Gaucher disease, citrullinemia, hemochromatosis
GSD, Glycogen storage disorder; 3β-HSD, 3β-hydroxysteroid dehydrogenase; LCAD, long-chain acyl-coenzyme A dehydrogenase; MCAD, medium chain acyl-coenzyme A dehydrogenase; MPS, mucopolysaccharidoses; NPD, Niemann-Pick disease; THCA, trihydroxycholestanoic acid.

For young infants with conjugated hyperbilirubinemia, biliary atresia is the most important and common condition that is amenable to surgical treatment. Choledochal cysts and other rare causes of duct obstruction that lead to jaundice shortly after birth are rare and are typically diagnosed by imaging studies rather than liver biopsy. Biliary atresia must be recognized quickly if surgical hepatic portoenterostomy is to be successful in reestablishing biliary drainage. Biopsy specimens from these patients are often obtained before the results of noninvasive studies, such as protease inhibitor typing, are available. Even in infants with probable biliary atresia, a liver biopsy may be performed to exclude other potential causes of jaundice. Therefore clinical management decisions rely heavily on morphological assessment of liver biopsy specimens ( Table 55.4 ). After exclusion of biliary atresia and infections, consideration should be given to the possibility of an inherited disease as the cause of the patient’s illness.

TABLE 55.4
Pathologist’s Role in Infantile Cholestasis
Action Examples
Find treatable condition Large duct obstruction, especially biliary atresia
Galactosemia
Prevent inappropriate treatment α 1 -Antitrypsin deficiency mimics BA
No surgery for Alagille syndrome; must recognize lack of large duct obstruction pattern in Alagille syndrome
Secure samples for diagnosis Urine for FAB-GC/MS for bile acids
Frozen liver for enzymes and molecular studies
PCR for virus
Provide data for the family Hereditable conditions
Monitor course of treatment and prognosis After Kasai portoenterostomy or transplantation, biopsies and serial α-fetoprotein levels
Elucidate pathogenesis (with a goal of prevention) Role of fasting versus TPN
BA , Biliary atresia; FAB-GC/MS , fast atom bombardment gas chromatography/mass spectrometry; PCR , polymerase chain reaction; TPN , total parenteral nutrition.

In older children, hepatomegaly, liver tumors, or chronic liver disease may prompt a liver biopsy. When liver disease appears after the neonatal period, clinical studies are typically used to identify the specific cause of the disease. Clinically diagnosed disorders include hepatitis B virus (HBV) and hepatitis C virus (HCV) infection, Wilson disease, reticuloendothelial storage disorders, steatosis, drug-induced hepatitis, autoimmune hepatitis, and cholangiopathy. Unusual causes include Alagille syndrome and metabolic storage disorders. On occasion, liver tissue may be obtained from a child with portal hypertension in whom none of these conditions is suspected. In such cases, congenital vascular anomalies (see Chapter 52 ) and congenital hepatic fibrosis should be considered. Liver biopsies are used to assess the severity of disease and the response to treatment in all of these disorders.

Pediatric Liver Biopsy Specimen

Because of the broad range of diseases in children, evaluation of liver biopsies in these patients is distinct from that in adults. To perform all potentially necessary tests on a liver biopsy specimen, prior arrangements should be in place to ensure adequate specimen processing ( Table 55.5 ).

TABLE 55.5
Bedside Processing of Pediatric Liver Biopsy Specimens
Snap-freezing Use liquid nitrogen; core tissue 2 cm in length; air-sealed specimen vial
Electron microscopy Use electron microscopy fixative; core tissue 0.3 cm in length
Formalin fixation Use neutral-buffered formalin; core tissue at least 1 cm (1-2 cm) in length
Do not use a biopsy sponge.
Do not place tissue in saline or transport media.
Do not let the specimen sit exposed to air.

Formalin-fixed specimens should be processed for routine light microscopy. Serial sections should be obtained. For example, a ribbon of 20 sections may be placed on 10 slides, with two tissue sections per slide. The first and last slides should be stained with hematoxylin and eosin (H&E) stain. Periodic acid–Schiff (PAS) stain with and without diastase digestion, trichrome stain, Perls iron stain, and reticulin techniques may be used on intervening slides. The remaining unstained slides may be held for possible future use.

Normal and Potentially Misleading Features of the Liver in Infants and Children

Some key features in the liver of infants and children differ from those in the adult liver ( Table 55.6 ). Variations occur in architecture, specific cell populations, content of hepatocytes, and response to injury.

TABLE 55.6
Normal and Potentially Misleading Features of Pediatric Liver Tissue
Architecture Physiological hyperplasia: liver cell plates two cells thick
Residual ductal plate architecture, particularly at periphery of liver
Absence of bile ducts Recognize apparent paucity in early biopsies of extreme preterm infants
Extramedullary hematopoiesis Portal tracts: granulocytic extramedullary hematopoiesis
Parenchyma: erythropoietic extramedullary hematopoiesis
Hepatocyte contents Hemosiderin granules
Copper deposits

Architecture

The liver undergoes substantial growth after birth. It normally doubles in weight within the first month of life, doubles again during the first year of life, and does not reach its mature size until late adolescence. The portal tract system grows in parallel with the liver. Therefore the most peripheral aspects of the liver may exhibit developmental residua of fetal histology. For instance, residual bile duct plates may rim the portal tracts, and the latter contain a more cellular mesenchyme and a centrally placed portal vein ( Fig. 55.1 ). The dimensions of hepatic lobules remain constant with growth. However, hepatocyte cords may remain two cells thick well into the fourth postnatal year. This should not be misinterpreted as regenerative hyperplasia in response to tissue injury.

FIGURE 55.1, Histology of the perinatal liver.

Cell Populations

Hematopoietic elements are commonly present in liver biopsy specimens obtained during the postnatal months. Granulopoiesis predominates in portal tracts, whereas erythropoiesis is common in the parenchyma.

Hepatocyte Content

Until a postnatal age of approximately 3 months, hepatocytes normally contain copper-binding protein and copper (demonstrable by orcein and rhodanine techniques, respectively) and granules of hemosiderin, particularly in periportal hepatocytes. These deposits are considered to be physiological and disperse with time. Conversely, hepatocyte alterations characteristic of various storage disorders may be inconspicuous in early infancy because of the time required to accumulate abnormal substances, such as α 1 -antitrypsin (A1AT). One dramatic exception to the concept of physiological iron deposition occurs in newborns who exhibit liver failure at birth, which is usually attributable to severe liver injury in utero. In this scenario, marked iron deposits may be present in hepatocytes at birth, giving rise to the term neonatal iron storage disease or perinatal hemochromatosis . A severe degree of necrosis and fibrosis is also present in patients with this condition. The extrahepatic reticuloendothelial system does not exhibit iron accumulation, highlighting the primacy of the liver injury. The finding of severe perinatal hepatic siderosis is nonspecific and indicates the development of liver injury during gestation. A lesser degree of hemosiderosis, with reticuloendothelial system deposits, may be seen in cases of maternal–fetal blood group incompatibility with significant hemolysis.

Response to Injury

Giant multinucleated hepatocytes, with or without bile pigment, are often present in infants with liver disease, regardless of the etiology. This change is considered nonspecific and reactive. Multinucleated hepatocytes are formed by syncytial breakdown of cell-to-cell borders, but with partial preservation of the canalicular aspects of the cell membrane. Canalicular remnants with retained bile may be observed within the cytoplasm. Giant cells exhibit multiple nuclei, either scattered throughout the cytoplasm or clustered toward one pole of the cell. This reaction may persist well into childhood if the inciting disorder is not resolved. Multinucleated giant cell change is unusual in older children and adults, but it may occur in some disorders such as autoimmune hepatitis and paramyxovirus hepatitis. ,

The histological spectrum of neonatal hepatitis includes giant cell change in hepatocytes, intralobular cholestasis, necrosis of hepatocytes, and intrahepatic hematopoiesis. All of these features are nonspecific events in infancy and can be observed in biliary atresia, A1AT storage disorder, and many other conditions ( Table 55.7 ). With advances in biochemistry and molecular genetics, many conditions formerly grouped within the category of neonatal giant cell hepatitis can now be specifically diagnosed, including progressive familial cholestasis types 2 and 3 and various bile salt synthetic defects.

TABLE 55.7
Etiology and Differential Diagnosis of Neonatal Hepatitis
Infantile Cholestasis with Giant Cells
Normal or Low GGT Elevated GGT
PFIC type 2 (BSEP disease) PFIC 4 (TJP2 defect) PFIC 5 (FXR defect) MYO5B defect
Bile salt synthetic defects ARC syndrome
Familial hypercholanemia CALFAN syndrome
  • PFIC type 3 (ABCB4/MDR3 disease)

  • Niemann-Pick disease type C

  • Alagille syndrome

  • Perinatal hemochromatosis

  • Rubella, cytomegalovirus, herpesvirus type 6, parainfluenza

  • α 1 -Antitrypsin deficiency

  • McCune-Albright syndrome

  • Navajo neurohepatopathy

  • Biliary atresia

  • Hemophagocytosis syndromes

Infantile Cholestasis without Giant Cells (±Bile Duct Damage)
  • PFIC type 1 (Byler disease, Greenland cholestasis)

  • Alagille syndrome

  • Cytomegalovirus

  • Hypopituitarism

  • Cystic fibrosis

  • Citrullinemia type II

  • Smith-Lemli-Opitz syndrome

  • Prematurity, fasting, total parenteral nutrition

  • North American Indian childhood cirrhosis

  • Jeune syndrome

  • Trisomy 21 (Down syndrome)

  • Trisomy 18

  • Neonatal lupus

  • Septo-optic dysplasia

Infantile Cholestasis Plus Necrosis (±Steatosis)
  • Galactosemia

  • Hereditary fructose intolerance

  • Tyrosinemia type I

  • Pearson mitochondrial DNA deficiency

  • Perinatal hemochromatosis

  • Bacterial (i.e., gram-negative) sepsis

  • Hepatitis B virus

  • Cytomegalovirus

  • Echovirus

  • Herpes simplex virus types 1 and 2

  • Hemophagocytic syndrome

  • Bile salt synthetic defects

ARC, Arthrogryposis, renal dysfunction, and cholestasis; BSEP, bile salt export pump; GGT, γ-glutamyltransferase; PFIC, progressive familial intrahepatic cholestasis.

Prompt medical intervention is possible and can be lifesaving, protect the central nervous system, and avert transplantation.

Medical intervention is possible.

Approach to the Diagnosis of Pediatric Liver Disorders in Liver Biopsies

When evaluating pediatric liver biopsy specimens, a careful review of patient age at disease onset (see Table 55.3 ), clinical manifestations, and routine laboratory workup findings is essential ( Table 55.8 ). If the presentation includes hepatomegaly, awareness of extrahepatic involvement is also helpful ( Table 55.9 ). It is useful to initially classify the histological pattern of disease into one of the six patterns of injury described initially by Jevon and Dimmick. Although these patterns often overlap, it is usually possible to define the predominant pattern in an individual case. , This section describes an algorithmic approach to the diagnosis of liver disorders, beginning with the histological patterns of tissue injury (see Boxes 55.1 to 55.6 ). A detailed description of the major entities is found later in this chapter.

TABLE 55.8
Workup of Neonatal Cholestasis
Soon After Birth Short Delay Insidious Onset
  • Bacterial cultures

  • TORCH antibodies

  • CBC differential, platelets

  • Reticulocyte count

  • Coombs test

  • Urine sediment for inclusions

  • Radiography of skull, bones

  • Variable symptoms

  • Syphilis serology

  • Urinary reducing sugars, FeCl 3 , amino acids

  • Sweat test

  • Ophthalmic examination

  • Skin examination for hemangiomas

  • Abdominal radiographs for free air, ascites

  • Upper gastrointestinal radiographs

  • Darkening of diaper

  • Pale stool

Feeding offSlow growth

  • Prematurity or small for dates

  • Many cases with GCT have this history

  • Abnormal feeding history, intravenous amino acids

CBC differential, Complete blood cell count with differential; GCT, giant cell transformation; TORCH, t oxoplasmosis, o ther agents, r ubella, c ytomegalovirus, and h erpes simplex.

Acute presentation: bleeding, seizures, and vomiting.

TABLE 55.9
Hepatomegaly with Extrahepatic Associations—Diagnostic Studies
Hepatomegaly Splenomegaly Mental Retardation Neurodegeneration
Steatohepatitis (liver biopsy) Biliary cirrhosis (liver biopsy) Sly (fibroblast culture; β-galactosidase) GSD type IV (liver; branching enzyme)
Budd-Chiari syndrome (MRI) NPD type C (fibroblast culture; cholesterol esterification) Wolman disease (fibroblast culture; acid lipase) GSD type VIII (liver; phosphorylase B)
GSD type VI (liver; phosphorylase) Cholesteryl-ester storage disease (fibroblast culture; acid lipase) NPD type C (fibroblast culture; cholesterol esterification) Sialidosis (fibroblast culture; neuraminidase)
GSD type IX (liver; phosphorylase kinase) NPD type B (leukocyte; acid sphingomyelinase) Mannosidosis (fibroblast culture; α-mannosidase)
GSD type X (liver; complement C3 and C5, AMP-dependent kinase) Fucosidosis (fibroblast culture; α-fucosidase)
Congenital disorder of glycosylation type Ib (serum transferrin) NPD type A (leukocyte; acid sphingomyelinase)
AMP, Adenosine monophosphate; GSD, glycogen storage disorder; NPD, Niemann-Pick disease.

BOX 55.1
Diagnostic Algorithm for Cholestatic Pattern
EM, Electron microscopy; GGT, γ-glutamyltransferase; GRACILE, g rowth r etardation, a mino aciduria, c holestasis, iron overload, lactic acidosis, and e arly death; PAS, periodic acid–Schiff; RER, rough endoplasmic reticulum.

Normal-Ggt Cholestasis

  • 1.

    Giant cell hepatitis

    • Consider progressive familial intrahepatic cholestasis type 2 (PFIC2) → EM: amorphous canalicular bile → confirm with ABCB11 gene sequencing

    • Consider PFIC4, PFIC5, and MYO5B disease → confirm with cholestatic gene panel

  • 2.

    Dilated canaliculi with pale bile

    • Centrilobular fibrosis → progressive familial intrahepatic cholestasis type 1 (PFIC1) → EM: coarse granular canalicular bile → ATP8B1 gene sequencing (30%-40% positive)

    • No fibrosis → benign recurrent intrahepatic cholestasis → confirm with ATP8B1 or ABCB11 gene sequencing

  • 3.

    EM: dense amorphous canalicular bile

    • PFIC2 → as above

    • Arthrogryposis, renal dysfunction, and cholestasis (ARC) syndrome → in the setting of arthrogryposis, perform VPS33B and VIPAR gene sequencing

    • Familial hypercholanemia (FHCA) TJP2 and BAAT and EPHX1 gene sequencing

High-Ggt Cholestasis

  • 1.

    Bile duct/portal tract ratio > 0.9

    • Giant cell change ± cirrhosis → consider progressive familial intrahepatic cholestasis type 3 (PFIC3) → confirm with ABCB4 gene sequencing

    • Cirrhosis with prominent bile ductular proliferation → North American Indian childhood cirrhosis (NAIC) → confirm with CIRH1A gene sequencing

    • Hepatocellular siderosis → confirm with iron stain → GRACILE syndrome → confirm with BCS1L gene sequencing

    • Giant cell hepatitis ± cirrhosis → PAS-positive diastase-resistant hepatocyte inclusions → A1AT staining by immunohistochemistry → α 1 -Antitrypsin ( A1AT) deficiency → EM: homogeneous inclusions within RER → confirm with Pi typing

    • Giant cell hepatitis ± cirrhosis → PAS negative → consider Niemann-Pick disease type C → EM: membrane-bound laminated structures and dense osmiophilic bodies → confirm with filipin staining in cultured fibroblasts and cholesterol esterification assays or with NPC1 and NPC2 gene sequencing

    • Neonatal sclerosing cholangitis with ichthyosis caused by claudin-1 ( CLDN1 ) mutation

  • 2.

    Bile duct/portal tract ratio < 0.9 (ductopenia)

    • Giant cell hepatitis and cirrhosis → consider Alagille syndrome (AGS) → confirm with JAG1 (and NOTCH2 ) gene sequencing

    • Giant cell hepatitis with bridging fibrosis or cirrhosis → PAS-positive diastase-resistant hepatocyte inclusions → A1AT staining by immunohistochemistry → A1AT deficiency → as above

BOX 55.2
Diagnostic Algorithm for Steatotic Pattern
CESD, cholesteryl-ester storage disease; EM , Electron microscopy; mtDNA , mitochondrial DNA.

Microvesicular Steatosis

  • 1.

    Cholestasis

    • Consider mitochondrial electron transport chain (ETC) disorders → confirm with specialized testing (enzymatic activity of ETC complexes; mtDNA analyses)

    • Oncocytic transformation → Consider mtDNA depletion syndrome (MDS) → EM: enlarged pleomorphic mitochondria with unusual cristae → confirm with specialized testing including genotyping dGUOK, POLG, and so on

    • EM: enlarged mitochondria with increased cristae → consider fatty acid oxidation (FAO) disorders → hypoketotic hypoglycemia → confirm with specialized testing including gene sequencing for ACADM, ACADL, and so on (see Table 55.11 )

Macrovesicular Steatosis

  • 1.

    Cirrhosis

    • Hepatocyte vacuolation, foamy Kupffer cells, macrophages with positive lipid stains → consider Wolman disease → see Box 55.3

    • Hepatocyte stainable copper → consider Wilson disease → see Box 55.4

    • Inspissated material within bile ducts → consider cystic fibrosis (CF) → confirm with CFTR gene sequencing, sweat chloride testing

  • 2.

    Cholestasis without cirrhosis

    • Consider hereditary fructose intolerance (HFI) → confirm with fructose-1-phosphate aldolase assay on liver tissue and ALDOB sequencing

  • 3.

    Hepatocyte cytoplasmic storage

    • Consider glycogen storage disease (GSD) types I, III (see Box 55.3 ), VI, and IX

    • Consider Chanarin-Dorfman syndrome (ABHD5 mutation)—mixed steatosis with skin disease

    • Consider lysosomal lipase deficiency diseases (Wolman disease and CESD)

BOX 55.3
Diagnostic Algorithm for Storage Pattern
EM, Electron microscopy; GSD, glycogen storage disease; PAS, periodic acid–Schiff; RES, reticuloendothelial system.

Membrane-Bound (Lysosomal) Inclusions by Electron Microscopy

  • 1.

    Hepatocyte microvesicular steatosis

    • Lipid stains positive → Wolman disease/cholesterol-ester storage disease (CESD) → EM: cholesterol crystals → confirm with enzyme activity and LIPA gene sequencing

  • 2.

    Predominantly RES involvement with hepatocyte sparing

    • Histiocytes, Kupffer cells with “crinkled-paper” inclusions → Gaucher disease → EM: tubular structures → confirm with enzyme activity and GBA gene sequencing

    • Foamy histiocytes and lipogranulomas → Farber disease → EM: curvilinear structures → confirm with enzyme activity and ASAH1 gene sequencing

  • 3.

    Foamy histiocytes and hepatocytes

    • PAS negative → Lipid stains positive → Niemann-Pick disease type A → EM: myelin-like figures → confirm with enzyme activity and SMPD1 gene sequencing

    • PAS positive → EM: monoparticulate glycogen ( Pompe disease [GSD type II] ); fibrillogranular material or empty vacuoles ( mucopolysaccharidoses or GM1 gangliosidosis ) → confirm with enzyme activity and gene sequencing

  • 4.

    Cirrhosis

    • Wolman disease or Gaucher disease → as above

Exclusively Cytoplasmic Storage by Electron Microscopy

  • 1.

    Hepatic adenomas

    • PAS positive, diastase sensitive → von Gierke disease (GSD type I) → confirm with enzyme activity on fresh liver tissue

  • 2.

    Cirrhosis

    • PAS positive, diastase resistant → colloidal iron positive → EM: amylopectin-like fibrillary structures → Andersen disease (GSD type IV) → confirm with enzyme activity and GBE1 gene sequencing

    • PAS positive, diastase sensitive → GSD type III → confirm with enzyme activity

    • PAS positive, diastase resistant → α 1 -antitrypsin— EM: endoplasmic reticulum

    • Cytoplasmic eosinophilic globules → Fibrinogen defect— EM: endoplasmic reticulum

    • Fingerprint inclusions

BOX 55.4
Diagnostic Algorithm for Hepatitic Pattern
EM , Electron microscopy; PAS , periodic acid–Schiff.

  • 1.

    Giant cell hepatitis

    • PAS-positive diastase-resistant hepatocyte inclusions → A1AT staining by immunohistochemistry → consider α 1 -Antitrypsin ( A1AT) deficiency → see Box 55.1

  • 2.

    Cholestasis, steatosis

    • Consider mitochondrial hepatopathies → see Box 55.2

  • 3.

    Increased hepatocyte stainable copper

    • Consider Wilson disease → EM: pleomorphic dilated mitochondria → confirm with hepatic copper content >250 μg/g dry weight; urinary copper excretion after penicillamine challenge >25 μmol/24 hour → DNA haplotype analysis and/or ATP7B gene sequencing in affected families

BOX 55.5
Diagnostic Algorithm for Cirrhotic Pattern
EM, Electron microscopy; GGT, γ-glutamyltransferase; PAS, periodic acid–Schiff; RES, reticuloendothelial system.

Steatosis

  • 1.

    Hepatocyte vacuolation and foamy Kupffer cells, macrophages

    • Lipid stains positive → consider Wolman disease → EM: cholesterol crystals → confirm with enzyme activity and LIPA gene sequencing

  • 2.

    Increased hepatocyte stainable copper

  • 3.

    Increased hepatocyte iron

    • Consider hereditary hemochromatosis (HH) → measure hepatic iron (hepatic iron index > 2) → confirm with HFE testing

  • 4.

    Giant cell hepatitis

    • PAS-positive diastase-resistant hepatocyte inclusions → A1AT staining by immunohistochemistry → consider α 1 -Antitrypsin ( A1AT) deficiency → see Box 55.1

    • PAS negative → consider tyrosinemia → confirm with elevated blood/urine succinylacetone levels

    • Cholestasis, steatosis → consider mitochondrial hepatopathies → see Box 55.2

Hepatocyte And/Or Res Inclusions

  • 1.

    Exclusively cytoplasmic inclusions on EM

    • PAS positive, diastase resistant, colloidal iron positive → EM: amylopectin-like fibrillary structures → Andersen disease (GSD type IV) → see Box 55.3 and Fig. 55.23

    • PAS positive, diastase sensitive → GSD type III → see Box 55.3

  • 2.

    Membrane-bound (lysosomal) inclusions on EM

    • Histiocytes, Kupffer cells with “crinkled-paper” inclusions → Gaucher disease → see Box 55.3

    • PAS-positive inclusions → EM: fibrillogranular material or empty vacuoles (mucopolysaccharidoses) → see Box 55.3

Cholestasis

  • 1.

    Normal GGT cholestasis

    • Consider progressive familial intrahepatic cholestasis type 2 (PFIC2 ) or PFIC1 (also consider PFIC4, PFIC5, and MYO5B disease) → see Box 55.1

  • 2.

    High GGT cholestasis

    • Consider PFIC3, North American Indian childhood cirrhosis (NAIC), and Alagille syndrome → see Box 55.1

    • Giant cell hepatitis → PAS negative → consider Niemann-Pick disease type C → see Box 55.1

BOX 55.6
Diagnostic Algorithm for Metabolic Diseases Predisposing to Hepatocellular Neoplasms

Hepatic Adenoma

  • 1.

    Consider glycogen storage disease (GSD) type I → see Box 55.3

  • 2.

    Alagille syndrome

Hepatocellular Carcinoma

  • 1.

    Noncirrhotic

    • Storage pattern → consider GSD type I → see Box 55.3

    • Consider α 1 -Antitrypsin (A1AT) deficiency → see Box 55.1

    • Increased hepatic iron → consider hereditary hemochromatosis (HH) → see Box 55.5

  • 2.

    Cirrhotic liver with cholestasis

    • Consider progressive familial intrahepatic cholestasis (PFIC), especially types 2, 3, and 4; Wilson disease; Alagille syndrome; and A1AT deficiency → see Box 55.1

  • 3.

    Cirrhotic liver without cholestasis

    • Consider GSD type III and GSD type IV → see Box 55.3

    • Consider HH → as above

    • Consider tyrosinemia

Cholestatic Pattern

The differential diagnosis of cholestatic disease in childhood is extremely broad and includes extrahepatic biliary obstruction (biliary atresia, choledochal cyst), infections, immune regulatory defects such as Langerhans cell histiocytosis, genetic disorders, metabolic disorders, total parenteral nutrition (TPN), and toxin exposures (see Table 55.2 ). Liver biopsies to determine the cause of cholestasis should be performed only after completion of a thorough radiological and laboratory workup, including ultrasound, hepatobiliary scintigraphy, viral serology, Pi typing for A1AT deficiency, and sweat chloride testing to rule out the more common causes of cholestasis in this age group.

When confronted with a predominantly cholestatic pattern of liver injury in a biopsy specimen, a useful starting point is the serum level of γ-glutamyltransferase (GGT) ( Box 55.1 ). Serum levels of GGT, a canalicular membrane protein, are usually low in disorders of defective bile acid synthesis or bile salt secretion. These entities ( Table 55.10 ) are discussed later in this chapter. Although the histological features differ among some of these entities (e.g., lack of giant cells in progressive familial intrahepatic cholestasis type 1 [PFIC1] compared with PFIC2 and PFIC3), ultrastructural examination, specialized enzymatic assays, and genetic testing are crucial in diagnosing these disorders. Among the cholestatic disorders with normal or low serum GGT, congenital defects in bile acid synthesis are commonly diagnosed by urinary mass spectrometry. Peroxisomal biogenesis disorders, such as Zellweger syndrome, neonatal adrenoleukodystrophy, and infantile Refsum disease, typically manifest with cholestasis, necrosis, and siderosis. These disorders are caused by mutations in multiple peroxin (PEX) genes. Biochemical diagnosis involves measurement of very-long-chain fatty acids in plasma and erythrocyte plasmalogen. Whole-genome sequencing is providing novel mechanistic and diagnostic insights for peroxisomal disorders.

TABLE 55.10
Genetic Defects and Available Testing for Inherited Disorders That Manifest with a Cholestatic Pattern
Disorder Gene Protein Inheritance Secondary Pattern Confirmatory Testing
Progressive Familial Intrahepatic Cholestasis (PFIC) Syndrome
PFIC1 (allelic disorder: BRIC) ATP8B1 ATPase, class I, type 8B, member 1 AR Cirrhotic Gene sequencing
PFIC2 ABCB11 ATP-binding cassette, subfamily B, member 11 AR Hepatitic
Cirrhotic
Gene sequencing
PFIC3 ABCB4 (MDR3) ATP-binding cassette, subfamily B, member 4 AR Cirrhotic Serum LPX, genotyping
PFIC4 TJP2 Tight junction protein 2 AR Hepatitic Gene sequencing
PFIC5 NR1H4 (FXR) Nuclear receptor subfamily 1, group H, member 4 AR HepatiticCirrhotic Gene sequencing
CALFAN syndrome SCYL1 Homolog of Saccharomyces cerevisiae ?AR Acute liver failure, neuropathy, cerebellar atrophy
Congenital Bile Acid Synthetic (CBAS) Defects
CBAS1 HSD3B7 3β-Hydroxy-Δ5-C 27 -steroid dehydrogenase AR Hepatitic Blood spot ESI-MS or urine MS
CBAS2 AKR1D1 Δ4-3-Oxosteroid 5β-reductase AR Hepatitic
Steatotic
Blood spot ESI-MS or urine MS
Peroxisomal Biogenesis Disorders
Zellweger syndrome PEX genes Peroxisomal biogenesis factors AR Hepatitic
Cirrhotic
Steatotic
↑ Plasma VLCFA by GC
Neonatal adrenoleukodystrophy PEX Peroxisomal biogenesis factors AR ↑ Plasma VLCFA by GC
Infantile Refsum disease PEX Peroxisomal biogenesis factors AR ↑ Plasma VLCFA by GC
Others
North American Indian childhood cirrhosis CIRH1A Cirhin AR Cirrhotic R565W (c.1741C→T) genotyping
Alagille syndrome JAG1; NOTCH2 Jagged1; Notch-2 AD Cirrhotic
Hepatitic
Gene sequencing
Niemann-Pick disease type C NPC1; NPC2 Niemann-Pick disease types C1 and C2 AR Hepatitic
Storage
Filipin staining in fibroblasts
AD, Autosomal dominant; AR, autosomal recessive; ATP, adenosine triphosphate; ATPase, adenosine triphosphatase; BRIC, benign recurrent intrahepatic cholestasis; ESI, electrospray ionization; GC, gas chromatography; LPX, lipoprotein X; MS, mass spectrometry; OMIM, Online Mendelian Inheritance in Man ( www.omim.org ); VLCFA, very-long-chain fatty acids.

Congenital hepatic fibrosis and Caroli disease are two rare disorders of ductal plate malformation that deserve mention here. Both manifest with cholestasis and cholangitis and often with portal hypertension. Both are associated with autosomal recessive polycystic kidney disease (ARPKD), and both carry mutations in PKHD1 (fibrocystin). The gene is defective in up to 30% of cases of ARPKD.

The current approach to diagnosis of neonatal cholestatic infants is by genetic testing using several comprehensive commercially available panels performed using next-generation sequencing techniques. , This includes most genes that have been identified in various childhood and adult genetic diseases and includes all genes for PFICs including TJP2 and MYO5B , as well as genes in Alagille syndrome and peroxisomal disorders, to name a few. For the desperately ill neonate, rapid whole-genome sequencing may be critically important for clinical decision making. Thus the advent of genetic testing has facilitated interpretations of liver biopsies, and the latter is now used more often to stage the degree of parenchymal involvement by respective diseases rather than to diagnose specific entities. Liver biopsies, however, continue to help in diagnosis of extrahepatic biliary atresia (EHBA) and to differentiate from paucity of bile ducts.

Steatotic Pattern

A steatotic pattern of injury is present when there is a prominent and diffuse distribution of fat vacuoles within hepatocytes. Steatosis is a common histopathological finding in several types of inherited disorders that affect the liver; those in which other histological features predominate are discussed separately. When one is considering the differential diagnosis of a primary steatotic pattern of liver injury, the most useful feature is the type of fat accumulation: microvesicular, macrovesicular, or mixed microvesicular and macrovesicular ( Box 55.2 ).

Microvesicular steatosis results from perturbation of mitochondrial metabolism, fatty acid β-oxidation (FAO), or electron transport chain function, through either a genetic defect or drug-induced inhibition of the pathways. The latter mechanism may result from a variety of drugs, including aspirin, ibuprofen, valproate, and zidovudine (see Chapter 49 for details on Reye syndrome). The diagnostic workup of genetic defects in FAO or electron transport chain function is often based on the clinical presentation and relies on specialized biochemical and metabolic testing of plasma, urine, and biopsied muscle tissue. , , Pathological features in liver biopsy specimens, such as microvesicular steatosis, cholestasis, fibrosis, cirrhosis, abnormal mitochondrial ultrastructure, and immunohistochemical demonstration of mitochondrial enzyme deficiency, support the diagnosis. Fresh-frozen liver is essential for specialized biochemical and genetic assays, particularly for analysis of the ratio of mitochondrial DNA (mtDNA) to nuclear DNA by Southern blotting (used for diagnosis of mtDNA depletion syndrome). Whole-exome sequencing is also making inroads in diagnosis of these disorders. Also, histological examination of the liver is highly relevant in the postmortem examination of patients with suspected mitochondrial disorders.

Diffuse macrovesicular steatosis or mixed microvesicular and macrovesicular steatosis can develop in several inherited and acquired conditions ( Table 55.11 ). Two inborn errors of carbohydrate metabolism, galactosemia and hereditary fructose intolerance, are classically associated with steatosis in newborns and infants. Galactosemia is diagnosed through urine biochemical testing and red cell enzyme assays. Liver biopsy, if performed, shows macrovesicular steatosis with fibrosis and cirrhosis. A liver biopsy is indicated in hereditary fructose intolerance for confirmatory aldolase B enzyme assays on fresh-frozen tissue, although molecular assays to detect ALDOB mutations are now available. The pathological features mimic those of galactosemia, except that cirrhosis is usually absent. Steatosis, with or without biliary cirrhosis, is common in cystic fibrosis (CF). Liver disease is relatively uncommon in young patients with CF but can lead to significant morbidity. In patients in whom the diagnosis was confirmed by positive sweat chloride testing and mutations in the CF transmembrane conductance regulator gene (CFTR), the diagnosis is suspected when eosinophilic material is present within bile ducts in liver biopsies (see later).

TABLE 55.11
Genetic Defects and Available Testing for Inherited Disorders That Manifest with a Steatotic Pattern
Disorder Gene(s) Protein(s) Inheritance Secondary Pattern Confirmatory Testing
Mitochondrial Disorders
FAO disorders, ETC disorders, mtDNA depletion syndrome, etc. Multiple
ACADM, ACADL, etc.
Multiple Mostly AR Plasma acylcarnitine and gene sequencing
Inborn Errors of Carbohydrate Metabolism
Hereditary fructose intolerance ALDOB Aldolase B AR Cholestatic Liver enzyme activity and sequencing
Galactosemia GALT Galactose-1-phosphate uridyltransferase AR Cirrhotic RBC GALT assay
Cystic fibrosis CFTR Cystic fibrosis transmembrane conductance regulator AR Hepatitic
Cholestatic
Sweat chloride, gene sequencing
AR, Autosomal recessive; ETC, electron transport chain; FAO, fatty acid oxidation; mtDNA, mitochondrial DNA; RBC, red blood cell.

Organic acidurias and urea cycle disorders may occasionally demonstrate steatosis or focal glycogenosis, but these disorders are diagnosed through urine chromatographic analyses. In some cases, progression to fibrosis and even cirrhosis may occur. Other disorders may demonstrate steatosis as a secondary feature. Biopsies of glycogen storage disease (GSD) types I and III often reveal steatosis. The liver in Wilson disease may be steatotic, but the predominant pattern is hepatitic or cirrhotic or both, with positive staining of copper using rhodanine to confirm.

Storage Pattern

A storage pattern is characterized by the presence of enlarged, swollen, and pale hepatocytes and/or reticuloendothelial cells, including sinusoidal Kupffer cells and portal macrophages. The stored material results from specific enzyme deficiencies in various metabolic pathways. A diverse group of disorders (>30) result in the development of a storage pattern within the liver, most of which cause hepatomegaly (see Table 55.9 ). Most, but not all (e.g., not the X-linked disorders, Fabry disease, or Hunter disease), are inherited in an autosomal recessive fashion. These disorders often demonstrate variable penetrance and expressivity, with different clinical and histological manifestations among family members with the same genetic defect. For a comprehensive discussion of these disorders, readers are referred to several excellent review articles. , ,

For pathologists confronted with biopsy specimens revealing a storage pattern, the most efficient approach involves a pediatric geneticist and a clinical biochemist because the patient’s clinical presentation and laboratory findings often suggest the most likely diagnosis. The storage pattern can be subclassified as either lysosomal or cytoplasmic . Because reticuloendothelial cells are rich in lysosomes, Kupffer cells and histiocytes are typically more involved in lysosomal storage diseases than in disorders with cytoplasmic storage (e.g., GSD types I, III, and IV). However, this distinction is not absolute because diffuse hepatic and extrahepatic involvement of the reticuloendothelial system is a well-documented feature in some GSDs, particularly GSD type IV. Distinguishing lysosomal (membrane-bound) from cytoplasmic storage disorders by electron microscopy is therefore quite useful.

Hepatic involvement in Pompe disease (GSD type II) is variable. The liver architecture is usually intact. PAS-positive diastase-sensitive inclusions are present within histiocytes and hepatocytes ( Box 55.3 ). Diagnostic confirmation is usually established by measurement of acid α-glucosidase activity in muscle or fibroblasts. Additional confirmation can be done by GAA gene sequencing.

The differential diagnosis of cytoplasmic storage disorders includes GSD types I, III, IV, VI, and IX ( Table 55.12 ). These disorders are discussed later in this chapter.

TABLE 55.12
Features of Important Glycogen Storage Disorders That Affect the Liver
Disorder Inheritance Gene Protein Storage Material Liver Histology Other Features Ultrastructure Diagnostic Testing
Parenchyma RES
GSD Ia (von Gierke disease) AR G6PC Glucose-6-phosphate, catalytic subunit Glycogen PAS+, diastase-sensitive cytoplasmic glycogen Steatosis, adenomas, HCC Fresh liver enzyme activity, genetic testing
GSD Ib AR SLC37A4 Glucose-6-phosphate transporter
GSD II (Pompe disease) AR GAA Acid α-glucosidase Glycogen PAS+ vacuoles Membrane-bound monoparticulate glycogen Fibroblast and/or muscle enzyme activity, genetic testing
GSD III AR AGL Glycogen debranching enzyme Glycogen PAS+, diastase-sensitive cytoplasmic glycogen Steatosis, cirrhosis, HCC Fresh liver or muscle enzyme activity, genetic testing
GSD IV (Andersen disease) AR GBE1 Glycogen debranching enzyme Glycogen PAS+, diastase-sensitive cytoplasmic glycogen Kupffer cell inclusions± Cirrhosis, HCC Amylopectin-like cytoplasmic glycogen Fresh liver or fibroblast enzyme activity, genetic testing
AR, Autosomal recessive; GSD, glycogen storage disorder; HCC, hepatocellular carcinoma; PAS, periodic acid–Schiff stain; RES, reticuloendothelial system.

The differential diagnosis of lysosomal storage disorders with foamy histiocytes includes lipidoses ( Table 55.13 ), chiefly Gaucher disease, Farber disease, Niemann-Pick disease (NPD) types A and B, GSD type II, mucopolysaccharidoses, and GM1 gangliosidosis. In Gaucher disease, the most common lysosomal storage disorder, there is a characteristic diffuse infiltration by engorged histiocytes (Gaucher cells) containing PAS-negative cytoplasmic (“crinkled-paper”) inclusions of glucosylceramide. Hepatocytes are typically spared. On demonstration of tubular structures by electron microscopy, one should, for confirmation, measure the level of leukocytic or fibroblastic acid β-glucosidase activity and sequence the GBA gene for mutations.

TABLE 55.13
Major Disorders of Lipid Metabolism That Affect the Liver
Disorder Inheritance Gene Protein Storage Material Liver Histology Other
Features
Ultrastructure Diagnostic Testing
Parenchyma RES
Gaucher disease AR GBA Acid β-glucosidase Glycosylceramide Gaucher cells (20-100 μm): eosinophilic corrugated “crinkled-paper” cytoplasm Fibrosis, rarely cirrhosis Spindled tubular structures Enzymes activity (l, f) ± GBA mutations
Niemann-Pick disease types A and B AR SMPD1 Acid sphingomyelinase Sphingomyelin Vacuolated hepatocytes Niemann-Pick cells (25-75 μm): foamy histiocytes, lipofuscin+ ORO+, LXB+, PAS− Laminated myelin-like figures Enzymes activity (l, f)
Niemann-Pick disease type C AR NPC1
NPC2
NPC1, NPC2 Cholesterol Cholestasis, giant cell transformation Sea-blue histiocytes Cirrhosis Whorled aggregates Filipin staining (f) + NPC1, NPC2 mutations
Farber disease AR ASAH1 Acid ceramidase Ceramide Lipogranulomas, foamy histiocytes Fibrosis Curvilinear Farber bodies Enzymes activity (l, f, p)
GM1 gangliosidosis AR GLB1 β-galactosidase 1 Glycosphingolipids Vacuolated hepatocytes Finely vacuolated Fibrillogranular material Enzymes activity (l, f, p)
Wolman disease
CESD
AR LIPA Acid liposomal lipase Cholesterol esters Microvesicular droplets Enlarged vacuolated, periportal foamy histiocytes ORO+, cirrhosis, (Wolman disease) Cholesterol crystal profiles Enzymes activity (l, f) + LIPA mutations
AR, Autosomal recessive; CESD, cholesteryl-ester storage disease; f, fibroblast enzyme activity; l, leukocyte enzyme activity; LXB, luxol-fast blue; ORO, oil red O; p, plasma enzyme activity; PAS, periodic acid–Schiff; RES, reticuloendothelial system.

Hepatitic Pattern

The hepatitic pattern, in infants, reveals hepatocellular unrest (variability in cell and nuclear size and shape) with or without necrosis, diffuse giant cell transformation of hepatocytes, extramedullary hematopoiesis, and prominent cholestasis (see Table 55.7 ). In older children, portal, interface, or lobular inflammation (or some combination of these) is typically present, but usually without cholestasis. Chief among the inherited disorders that manifest with a hepatitic pattern are A1AT deficiency and Wilson disease. A number of other inherited disorders with characteristic histological patterns of injury (e.g., NPD type C, Alagille syndrome with cholestasis) can also manifest with a superimposed hepatitic pattern on liver biopsies. These entities are discussed later in this chapter.

When investigating biopsy specimens with a hepatitic pattern for a suspected inherited disorder, it is crucial to rule out acquired causes of hepatitis (e.g., infection, toxin or drug exposure, TPN) that are far more common in this age group. It is also important to evaluate the clinical presentation and laboratory results. For example, A1AT deficiency liver disease can manifest either in the neonatal period with hepatitis or later in childhood with cirrhosis, but clinical manifestations of Wilson disease are rare before 5 years of age (see Table 55.3 ). On biopsy, the presence of PAS-positive, diastase-resistant inclusions within zone 1 hepatocytes is characteristic of A1AT deficiency, but immunostaining is more sensitive and specific. Importantly, the neonate with A1AT-associated hepatitis may not exhibit hepatocyte inclusions on liver biopsy; serum A1AT phenotyping by isoelectric focusing (Pi typing) is the confirmatory assay for diagnosis of A1AT deficiency in cases of suspected neonatal hepatitis, and it is particularly valuable in cases that mimic EHBA (i.e., bile duct proliferation, no biliary excretion on scintigraphy). Diagnosis of Wilson disease is less straightforward; screening for decreased serum ceruloplasmin levels (<20 mg/dL) can be problematic because both decreased (hypoceruloplasminemia or aceruloplasminemia) and elevated levels (acute-phase reaction) may be seen in other conditions ( Box 55.4 ). A low serum alkaline phosphatase level may be a useful clue to the etiology, as well as increased unconjugated bilirubin, which reflects hemolysis in children with acute liver decompensation.

Cirrhotic Pattern

Cirrhosis, an end-stage response to chronic liver injury, is common to several different types of inherited disorders. Therefore the differential diagnosis of liver biopsies with a cirrhotic pattern rests largely on the presence or absence of other characteristic microscopic findings ( Box 55.5 ). Metabolic disorders that lead to cirrhosis also carry an increased risk of neoplasia ( Box 55.6 ). Advanced stages of congenital hepatic fibrosis (discussed earlier) may be confused with cirrhosis; however, bile duct ectasia is a characteristic feature of the former. Although mitochondrial hepatopathies may lead to several different patterns of liver injury, it is most often steatosis. Less often, these disorders may manifest with hepatitis and cirrhosis.

Tyrosinemia, caused by a deficiency of fumarylacetoacetate hydrolase, can lead to cirrhosis in early life (infancy), hepatic failure, hepatocellular necrosis, or giant cell hepatitis.

Hereditary hemochromatosis (HH) is caused by mutations in the hemochromatosis (HFE) gene and leads to cirrhosis and hepatocellular carcinoma (HCC). Although the defect in iron metabolism is present at birth, the clinical manifestations of HH are rarely apparent before adulthood, when long-term effects of chronic iron overload typically manifest (see later discussion).

Neoplastic Pattern

Several types of inherited disorders predispose the child to the development of focal nodular hyperplasia, hepatic adenoma, and HCC. In contrast, hepatoblastoma, the most common malignancy in the liver of children, is infrequently associated with inherited metabolic disorders, although trisomy 18, neurofibromatosis, and congenital hepatic fibrosis have been linked to hepatoblastoma. The increased risk for HCC is chiefly caused by the development of cirrhosis, but HCC can arise in noncirrhotic patients with A1AT deficiency, hemochromatosis, or GSD type I. , The histological features and biological behavior of HCC and adenomas arising in patients with a metabolic disorder are similar to those of neoplasms that arise in cirrhosis resulting from other causes. Cirrhosis and neoplasms that arise in inherited metabolic disorders are indicative of advanced disease (not necessarily advanced age), and the diagnostic features of the underlying disorder can usually be identified in nonneoplastic areas of the liver tissue (see Box 55.6 ).

Most patients with GSD type I have hepatic adenomas by 15 years of age, although adenomas may be present in early childhood. Dysplastic changes and HCC within individual adenomatous nodules have also been reported in this condition. ,

Among all of the metabolic disorders of the liver, hereditary tyrosinemia (discussed later) carries the highest risk for development of HCC (13% to 15% incidence). , Typically, hepatocellular dysplasia and foci of HCC develop in a background of mixed micronodular and macronodular cirrhosis, but treatment should begin in infancy as soon as succinyl acetone is detected in the urine to abort the mutagenic process. Not all infants manifest overt liver dysfunction with time.

The incidence of HCC in patients with HH (discussed later) is approximately 10%. Most cases develop in a background of cirrhosis. Because of the widespread availability of testing for the HFE gene mutations C282Y and H63D and sensitive transferrin-iron screening tests, a biopsy diagnosis is required only in cases with a negative genotype or high ferritin levels.

Other inherited disorders that are less commonly associated with HCC include A1AT deficiency, PFIC2, PFIC3, PFIC4 (TJP2 defect), Wilson disease, Alagille syndrome, and GSD types I, III, and IV. The characteristic biopsy features were described earlier. A1AT deficiency is a precursor to HCC. In some cases, PiZ heterozygotes were found to have HCC (and cholangiocarcinoma) in noncirrhotic livers. Fanconi anemia, familial adenomatous polyposis, and Beckwith-Weidemann syndrome are other syndromes that predispose to cancer. Three percent of patients with Fanconi anemia develop adenomas or HCC, and hepatoblastoma has a well-known association with both familial adenomatous polyposis and Beckwith-Weidemann syndrome. Finally, cholangiocarcinoma is a rare complication of Wilson disease and congenital hepatic fibrosis. , This tumor was recently described in two children with PFIC2. Cases of HCC in an MDR3 explant liver and association with a TJP2 defect have also been recently reported. ,

Liver Diseases of Infancy

Acute Life-Threatening Illness and Sudden Death

Although sudden death within 2 to 3 days after birth is usually caused by nonmetabolic conditions such as sepsis or congenital heart disease, some inborn errors of metabolism are also associated with acute life-threatening illness ( Box 55.7 ). As mentioned earlier, decision making on behalf of a desperately ill neonate may be critically informed by rapid whole-exome sequencing, including the decision to provide comfort measures only.

BOX 55.7
Most Common Inborn Errors of Metabolism Associated with Acute Life-Threatening Illness

  • Organic acidurias

  • Congenital lactic acidurias

    • Pyruvate oxidation defects

    • Gluconeogenesis defects

    • Krebs cycle defects

    • Respiratory chain defects

  • Mitochondrial fatty acid β-oxidation disorders

    • Defects of membrane-bound enzymes

    • Defects of matrix enzymes

  • Urea cycle defects

  • Amino acid disorders

    • Maple syrup urine disease

    • Nonketotic hyperglycinemia

  • Molybdenum cofactor deficiency

FAO defects lead to cardiac arrhythmias and can cause sudden death (see Mitochondrial Cytopathies). At autopsy, excess droplets of fat may be present in the liver and heart of patients with an FAO defect. If an FAO disorder is suspected, tissue specimens should be obtained as soon as possible after death, before autopsy, for metabolic testing , ( Table 55.14 ). Both liver and muscle tissue should be obtained for analysis. In addition, urine and cerebrospinal fluid should be snap-frozen and stored for further analysis. Blood spots should be obtained for analysis of acylcarnitines. Whole-blood specimens should be placed in an ethylenediaminetetraacetic acid (EDTA) tube for DNA extraction and in a lithium heparin tube (spun and separated within 20 minutes of collection) for metabolite analysis. A full-thickness skin biopsy should be performed under sterile conditions within 12 hours of death for fibroblast culture and archiving.

TABLE 55.14
Investigation of Acute Life-Threatening Disease and Sudden Death
Collect Blood, urine, CSF, vitreous humor (dodecanoic acid = MCAD deficiency)
Bile (carnitine and acylcarnitines = FAO disorders)
Freeze Liver, skeletal, cardiac muscle
Sample for cell culture Skin fibroblasts (DNA and enzyme analyses)
CSF, Cerebrospinal fluid; FAO, fatty acid oxidation; MCAD, medium chain acyl-coenzyme A dehydrogenase.

Sudden and unexplained death in an infant or young child is often the first manifestation of medium chain acyl-coenzyme A dehydrogenase (MCAD) deficiency, the most common FAO disorder. MCAD deficiency manifests with hepatomegaly and steatosis and may be confused with Reye syndrome. A particular Lys304Glu mutation in the ACADM gene is highly prevalent in some populations. Since the institution of newborn screening, early diagnosis has led to prospective management of acute episodes of hypoketotic hypoglycemia.

Neonatal Hemochromatosis

Clinical Features

Neonatal hemochromatosis (NH), also termed neonatal iron storage disease, is a rare syndrome that is characterized by the presence of congenital cirrhosis and fulminant liver failure. This condition exhibits abundant iron deposition in the liver and in other organs, but not in the reticuloendothelial system (spleen or bone marrow). Clinically, patients with NH, either before birth or shortly thereafter, exhibit liver failure, including hypoglycemia, coagulopathy, hypoalbuminemia, ascites, and hyperbilirubinemia. Although some infants recover with exchange transfusion and some survive to transplantation, most die of their disease. Most cases of NH belong to the group of gestational alloimmune liver disease (GALD).

Pathogenesis

NH has been described in association with various conditions, including metabolic disorders (tyrosinemia, Δ4-3-oxosteroid 5β-reductase deficiency, mtDNA mutations, and Zellweger syndrome); infections (echovirus 9, cytomegalovirus [CMV], herpes simplex virus, rubella, and parvovirus B19); and karyotypic disorders (Down syndrome). , As a result, some authors regard NH as a final common phenotype of any gestational insult that culminates in abnormal iron metabolism. , At least three patterns of disease transmission have been described: transmission of maternal alloantibodies, autosomal recessive inheritance, and matrilineal inheritance. In the last instance, several reports have documented women with more than one affected child, but the children were fathered by different men. Although gonadal mosaicism in these mothers has not formally been excluded, the possibility of mitochondrial inheritance also has not been excluded. NH is not associated with mutations in the HFE gene, which is involved in most cases of HH (see later discussion).

In 2004, Whitington and Hibbard first reported giving high-dose intravenous γ-globulin to pregnant women with a history of a previously affected infant, and this treatment prevented recurrence in a small series. Their findings have since been confirmed in many centers, and it is now imperative to determine the etiology of all perinatal forms of acute liver injury. Mimicry of hemochromatosis has been seen in cases of mtDNA depletion and other circumstances, which presumably do not benefit from intravenous immunoglobulin therapy.

The antibody responsible for NH has yet to be determined. Complement fixation induced by immunoglobulin G of maternal origin is detected by immunofluorescence for the membrane attack complex on hepatocytes. One patient with cirrhosis at birth did not have hemosiderosis and survived without specific therapy. However, the usual scenario is fatal necrosis with extensive parenchymal siderosis without reticuloendothelial iron, suggesting dysfunction of macrophages. This has been observed even prenatally: Three of eight stillborn or very premature infants had no extrahepatic siderosis. , Recently, Whitington and colleagues have provided insight into the severe degree of parenchymal siderosis that accompanies the hepatocellular injury. The injured livers have significantly reduced hepcidin, hemojuvelin, and transferrin gene expression compared with normal livers.

Antibodies to mitochondrial proteins are responsible for primary biliary cirrhosis, a disease that affects women disproportionately. Two patients transmitted this disease transplacentally to their infants, producing transient liver injury. One infant was born with ascites and conjugated hyperbilirubinemia. In the other infant, the presentation was more insidious. His biopsy, at 5 weeks, showed portal inflammation involving bile ducts and ductules, mild portal fibrosis, and multinucleated giant hepatocytes typical of neonatal hepatitis. Immunofluorescence was able to detect immunoglobulin G deposits surrounding hepatocytes. Within 3 months, both infants showed no evidence of liver dysfunction, and the antibodies were undetectable.

NH resembles neonatal hepatitis in infants with systemic lupus erythematosus, in which the mother has high titers of antinuclear anti Ro (SSA) and anti La antibodies.

Pathological Features

The liver in patients with NH (usually seen at postmortem examination) reveals cirrhosis and cholestasis ( Fig. 55.2 ). Confluent areas of hepatocellular loss and a variable degree of hepatocyte regeneration are typical. Residual hepatocytes may demonstrate giant cell or pseudoacinar transformation. Iron deposition is typically coarsely granular and located predominantly within hepatocytes, sparing Kupffer cells. Extrahepatic sites of siderosis include the parenchymal cells of the heart, thyroid, pancreas, adrenal glands, kidneys, and the submucosal glands of the gastrointestinal and upper respiratory tracts. It has now been shown that examples of GALD exist without evidence of hepatic siderosis. While the diagnosis of this entity has been a challenge, the recognition of the deposition of the C5b-9 complex by immunohistochemistry on necrotic hepatocytes in 2010 has led to an increase in recognition of this entity. Dubruc et al. showed 100% expression in GALD though only 26% of their cases showed staining in more than 75% of hepatocytes. This has improved the outcome of future pregnancies because of the ability to administer IV-IG in the prenatal period of subsequent pregnancies. Having said this, there are issues with this staining technique because recent literature suggests overlap of staining in necrotic livers as a result of viral or other etiology, as well as suspected cases of GALD with negative C5b-9 staining. It is therefore important to exclude other causes, especially viral causes, of neonatal acute liver failure.

FIGURE 55.2, Perinatal hemochromatosis.

Natural History

NH carries a high risk of death, and a high index of suspicion is required to diagnose this disorder at first presentation, as this has impact on management of subsequent pregnancies for the mother. NH remains in the differential diagnosis of severely ill neonates who survive beyond the first days of life. Demonstration of siderosis in biopsy samples from oral submucosal glands is a rapid diagnostic method. T2-weighted magnetic resonance imaging (MRI) can be used to assess the presence of iron in various tissues. Siderosis of the liver and extrahepatic organs may also accompany other conditions, such as erythropoietic disorders, sickle cell disease, thalassemia, and erythroblastosis from blood group incompatibility. However, hemosiderosis affects the reticuloendothelial system primarily. These other diseases need to be excluded by other clinical and laboratory studies.

Neonatal Jaundice

Heritable disorders of bilirubin conjugation can rarely produce liver dysfunction (see later discussion). Liver diseases of infancy most often manifest with jaundice owing to conjugated hyperbilirubinemia. This occurs because of the relative immaturity of hepatic secretory and excretory functions in early life. Some of the disorders that cause neonatal jaundice are listed in Table 55.2 . In all cases of neonatal jaundice, the possibility of biliary atresia or hepatic damage caused by drug exposure (including inadvertent drug overdose) should be excluded. Infants requiring TPN are at risk for cholestatic liver disease (see Chapter 49 ). Infectious causes of liver disease in infancy include enterovirus, parvovirus B19, and adenovirus, which cause direct cytopathic cell death, as well as bacterial sepsis, urinary tract infection, and intrauterine exposure to maternal infections (of the “TORCH” acronym), all of which cause liver damage to neonates and infants (see Chapter 47 ). Acquired syphilis is an exceedingly uncommon cause of perinatal liver disease.

Inherited metabolic disorders of carbohydrate, amino acid, and lipid and glycolipid metabolism, along with disorders of the biosynthetic and secretory pathways for bile acids, should always be considered in the differential diagnosis of neonatal jaundice (see Table 55.7 ). Storage of abnormal A1AT and abnormal function of the CFTR compromise hepatic formation of bile. Defects in the transporters responsible for bile formation give rise to progressive familial intrahepatic cholestasis. Inherited defects of peroxisomal and mitochondrial function can cause neonatal jaundice. Shock may cause cholestatic liver damage in neonates. Alagille syndrome may manifest in early infancy as “giant cell hepatitis”; it can mimic biliary atresia and may lead to Kasai portoenterostomy (KPE). The nonspecific designation of “neonatal hepatitis/giant cell hepatitis” applies to the 25% of patients in which a specific etiology for neonatal jaundice remains undetermined.

Biliary Atresia

Biliary atresia manifests as a fibrosing destruction of extrahepatic and intrahepatic bile ducts of unknown etiology, presenting usually in the initial weeks after birth. Biliary atresia has long been classified as extrahepatic on the basis of involvement of that portion of the biliary tree. However, this concept is imprecise because the anatomy of abnormal bile ducts in affected patients varies markedly. A recommended terminology is obliterative cholangiopathy, with two major types: noncystic and cystic. The cystic disorders include different types of choledochal cysts (described later in this chapter) and cystic biliary atresia. The noncystic forms include the different variants and presentations of noncystic biliary atresia (the predominant form of biliary atresia) and neonatal sclerosing cholangitis.

The widespread use of screening fetal ultrasound performed during pregnancy has resulted in increased detection of cystic lesions in the hilum of the fetal liver, with the differential diagnosis of cystic biliary atresia versus choledochal cyst. As many as 10% of infants ultimately diagnosed with biliary atresia have prior fetal ultrasound indicating this cystic form of biliary atresia. The postnatal pathology of the extrahepatic biliary tract in these cases is not notably different than in other patients with EHBA. , The proximal biliary remnants of these patients with cystic biliary atresia exhibit cysts which lack epithelium and inflammation and exhibit myofibroblastic hyperplasia interposed with atretic segments of the biliary tree, especially caudad to the cyst. This is in contrast with choledochal cysts, which have preserved uninjured epithelium and no subepithelial cicatrix. That being said, the frequency of biliary atresia in infants with choledochal cysts is 13% to 44%; choledochal cyst (with preserved epithelium) is found in 8% to 11% of infants with biliary atresia, suggesting a shared pathogenesis with a proposed continuum between the two entities. Moreover, prenatal nonvisualization of the fetal gallbladder during the second trimester in neonates subsequently diagnosed with biliary atresia provides further support for the premise that some cases of biliary atresia are of fetal rather than perinatal onset. Indeed, in an infant suspected of having biliary atresia, preoperative ultrasound demonstrating a cyst >5 mm in the hilum with no patent gallbladder is associated with favorable postoperative outcomes following portoenterostomy. Persistent nonvisualization of the fetal gallbladder on second-trimester ultrasound requires consideration of other conditions as well, including CF, Alagille syndrome, and chromosomal anomalies. ,

Clinical Features

Biliary atresia accounts for more than 30% of all cases of cholestasis in neonates. This disorder has an incidence of 1 in 5000 to 19,000 live births, occurring more frequently in Asian countries such as Taiwan and Japan when compared with North America and Europe. Most cases are sporadic and do not reveal a positive family history of neonatal cholestasis. A series of 30 sets of twins revealed only 2 sets with both infants affected by EHBA, and both pairs were dizygotic. There are reports of a 20-year-old mother who underwent a portoenterostomy for EHBA at 64 days of age and subsequently gave birth to a daughter with EHBA and of a family cluster of 5 children in which 2 dizygotic twin sisters and a third sibling all had EHBA. Variation in epigenetic modifications of genomic DNA has been suggested for these occasional familial presentations. Genes related to bile duct dysmorphogenesis (including ciliopathies) overlapping with features of biliary atresia in both humans and nonhuman model systems have been proposed, sparking continued interest in identifying potential causative and modifying genes relevant to patients with biliary atresia.

Infants presenting with biliary atresia are usually of normal gestational age and birth weight. Cholestatic jaundice is the main clinical presentation. It typically develops in the first few weeks of life and does not remit, unlike the mild physiological jaundice of early infancy. Furthermore, in physiological jaundice, the mildly elevated serum bilirubin is primarily unconjugated, and the serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) are normal. The progressive biliary obstruction that characterizes biliary atresia leads to a progressive increase in serum bilirubin levels in which conjugated bilirubin represents 50% to 80% of the total. A recent study of conjugated or direct-reacting bilirubin in the first 48 hours of life revealed significant elevations in all infants with biliary atresia compared with aged-matched controls, even though the total bilirubin level was not increased. Serum levels of GGT are increased several times above normal. Liver biosynthetic function, as indicated by serum albumin levels and prothrombin time, is usually normal at initial presentation.

In keeping with the aforementioned discussion, ultrasound should be performed in suspected cases of biliary atresia to exclude the presence of an extrahepatic biliary tract anomaly, such as a choledochal cyst. Hepatobiliary scintigraphy is also useful to assess the status of biliary tract function, but in many hepatocellular diseases of infants, there also is an absence of excretion of the labeled molecule into the intestine. Ultrasound shear wave elastography has also been shown to be discriminatory for biliary atresia.

Percutaneous liver biopsy is used to determine whether there is histological evidence of large bile duct obstruction. Biopsy features of atresia may also occur with other extrahepatic forms of biliary obstruction ( Box 55.8 ). The overall accuracy rate of percutaneous liver biopsy for diagnosis of biliary atresia has recently been shown to be around 90.1%, with a sensitivity of 88.4% and a specificity of 92.7%. The decision to proceed with a biliary tract exploratory surgical procedure does not rest solely on the pathological findings in a liver biopsy specimen. The presence of acholic stools, an undetectable or irregular contour of the gallbladder on sonographic studies, and failure to excrete into the intestine the radioactive tracer iminodiacetic acid by hepatobiliary scintography (HIDA scan) are findings that lend support to a diagnosis of biliary atresia, but may not be sufficiently discriminating. Magnetic resonance cholangiopancreatography (MRCP) is not optimal for visualization of the extrahepatic biliary tract in children younger than 3 months of age. Intraoperative cholangiography, whether laparoscopic or during open laparotomy, is a confirmatory procedure before actual performance of KPE portoenterostomy.

BOX 55.8
Biliary Tract Obstruction

Extrahepatic Biliary Obstruction Without Atresia—Rare Events In Infancy

  • Choledochal cyst

  • Spontaneous perforation of bile duct

  • Bile plug syndrome

  • Segmental cystic dilation of biliary ducts

  • Duodenal atresia (more common in Down syndrome)

  • Peptic ulceration secondary to duplication of intestine

  • Compression by enlarged lymph node

  • Hemangioendothelioma and other neoplasms of head of pancreas

Extrahepatic Obstruction—Liver Histology

  • Portal tract and periportal fibrosis

  • Bile duct and ductular proliferation

  • Bile duct “thrombi” or plugs

  • ±Giant cell transformation

  • ±Portal inflammation, mixed

  • ±Persistent extramedullary hematopoiesis

  • Beware of α 1 -antitrypsin, timing of biopsy

Pathogenesis

Biliary atresia is not a single disease with a defined etiology. An all-encompassing hypothesis is that a genetically susceptible individual undergoes inflammatory destruction of the extrahepatic biliary system in response to as yet undetermined environmental factors. Potential pathogenetic mechanisms include a genetic defect in morphogenesis with or without defective prenatal hepatic circulation, environmental triggers such as intrauterine or perinatal viral infection or toxin exposure, and immunological dysregulation in the perinatal period ( Table 55.15 ). These hypotheses have been put forth on the basis of epidemiological studies and molecular analyses of tissue specimens from human patients. Mouse models of biliary atresia have provided some additional clues.

TABLE 55.15
Biliary Atresia: Pathogenesis
Proposed Mechanisms Syndromic vs. Nonsyndromic Associated Malformations
  • Genetic or epigenetic susceptibility, involving abnormal development of maturing biliary tract , , ,

  • Defect in fetal circulation

  • Occult viral infection , ,

  • Rubella

  • Cytomegalovirus

  • Retrovirus

  • Reovirus type 3

  • Rotavirus

  • Toxin exposure in utero

  • Disorder of immunologic-inflammatory system (including maternal chimerism) ,

  • 15% are syndromic, having associated malformations , , ,

  • 85% are nonsyndromic with no malformations

  • Polysplenia or asplenia

  • Intestinal malrotation or atresia, anomalies of the portal vein and hepatic artery, abdominal situs inversus

  • Congenital cardiovascular disease including absence of the vena cava

  • Genitourinary anomalies

Morphogenesis and Genetics

Systemic dysregulation of morphogenesis in patients with biliary atresia is well-documented, such as the coexistence of nonhepatic embryological abnormalities, evidence for abnormal development of the ductal plate of the maturing intrahepatic biliary tract, and overexpression of certain regulatory genes in children who have an early form of biliary atresia. , Developmental abnormalities with which biliary atresia is associated include polysplenia or asplenia (biliary atresia splenic malformation syndrome); cardiovascular defects including absence of the inferior vena cava, abdominal situs inversus, intestinal malrotation, or atresias (duodenal atresia, esophageal atresia with tracheoesophageal fistula); and anomalies of the portal vein and hepatic artery (see Table 55.15 ). , In a large multi-institutional North American study, biliary atresia occurred in a syndromic fashion with laterality defects and spleen anomalies in ∼10% of cases, with at least one malformation but without laterality defects in ∼5%, and as nonsyndromic biliary atresia in the remaining ∼85% of cases .

Mutations in CFC1 and ZIC3 lead to laterality defects and biliary atresia in some patients. The Inv mouse that develops situs inversus also develops biliary obstruction. The JAG1 gene has also been implicated in the pathogenesis of biliary atresia because affected patients with a poor outcome also show a high frequency of JAG1 single-nucleotide polymorphisms. The persistent expression of HES1 protein in the nuclei of biliary epithelial cells specimens of biliary atresia obtained up to 3 months after birth offers more evidence for disorderly Notch signaling in this disease. In normal development, such expression is silenced by 16 weeks of gestation. Genome-wide association studies in Chinese children have identified variants of the ADD3 gene, and its knockdown in zebrafish has shown biliary abnormalities. , ADD3 is expressed in hepatocytes and biliary epithelia and is involved in the assembly of spectri-actin membrane protein networks at sites of cell-to-cell contact. Defective ADD3 could lead to excessive deposition of actin and myosin, leading to biliary fibrosis. Biliary atresia also has been reported in a premature neonate with 1p36 deletion syndrome; a chromosome with no prior reports of genes linked to biliary atresia but with associated gastrointestinal abnormalities including intestinal malrotation and anomalies in pancreatobiliary anatomy.

The ciliopathies responsible for polycystic disease and congenital hepatic fibrosis (discussed later) may have a new and potentially meaningful relationship to biliary atresia. Other ciliary dysfunction contributes to laterality defects in embryonic development, including syndromic biliary atresia. Hartley and colleagues used immunohistochemistry and found that expression of fibrocystin was missing in the biliary epithelium of the biliary atresia patients, suggesting a role for PKHD1 or genes involved in primary ciliogenesis in this disease. More recently, a variant in the primary cilia protein PKD1L1 has been reported. Chu and associates initially and Karjoo et al. showed that the cilia in biliary epithelium of five affected children were fewer, shorter, and abnormally oriented. Two other patients with biliary atresia who had no other malformations displayed alterations similar to those in epithelium of patients with other cholestatic diseases. Therefore the defects may be secondary and nonspecific. Hence tantalizing evidence continues to accumulate regarding genetic and epigenetic influences on the development of biliary atresia.

Environmental Triggers

The concept of a hepatotropic virus capable of causing cholangiolar and structural damage as a common factor causing “infantile obstructive cholangiopathy” was introduced in the 1970s. Identification in 1992 of higher titers of antibodies against reovirus type 3 in jaundiced infants with biliary atresia compared with those without biliary atresia and in 1998 of reovirus RNA in liver and/or biliary tissues of infants with biliary atresia and choledochal cysts sparked further interest in a potential infectious etiology. No evidence for reovirus as an associated agent has been found in subsequent studies. , However, rhesus reovirus inoculated intraperitoneally within 12 hours of birth in the mouse is now an established experimental model of a biliary atresia-like condition. Other viral agents (CMV, rotavirus) also are able to cause inflammatory destruction of bile ducts when introduced into neonatal mice. The weakness of the viral hypothesis is that viral genomic material can be found in a substantial minority of infants diagnosed with biliary atresia, without clear evidence that such transient infections can incite the powerful inflammatory response characteristic of this disorder.

An outbreak of ovine biliary atresia in New South Wales, Australia, in 1964 born to dams that had grazed on weeds of the genus dysphania glomulifera (red crumweed or pigweed) raised the possibility that in utero fetal exposure to an environmental toxin might cause biliary atresia. Recently, four potentially toxic isoflavonoids isolated from extracts of dysphania spp. have been tested in a zebrafish system model of early bile duct development. One, now named biliatresone, caused fish biliary maldevelopment. Work with a neonatal mouse model shows that this effect is not species specific, raising the possibility that even human biliary atresia could arise as a result of maternal exposure to this or other toxins.

Inflammation

The most striking feature of biliary atresia is the inflammatory and fibrotic destruction of bile ducts, definitional for an obliterative cholangiopathy. Hypotheses about exposure of neoantigens on the biliary epithelium as a result of exposure to cholangiopathic toxins or viruses have abounded. The occurrence of biliary atresia in the child with progressive familial intrahepatic cholestasis 3 (PFIC 3) in which bile salts are secreted into bile without accompanying secretion of phosphatidylcholine and cholesterol suggest that biliary atresia may be an extreme outcome of exposure of the biliary tree to toxic biophysical properties of an abnormal bile.

Abnormal expression of intercellular adhesion molecular 1 (ICAM-1), vascular cell adhesion molecular 1 (VCAM-1), E-selectin, and P-selectin on endothelial cells and biliary epithelium in livers of infants with biliary atresia , indicate triggering of a strong inflammatory reaction in biliary atresia. Prevention of experimental inflammatory destruction of bile ducts occurs in mice that are deficient in interferon-γ. Studies of human liver specimens at different phases of disease progression point to the presence of a proinflammatory commitment of lymphocytes with a predominant type 1 helper T cell (Th1) phenotype. Molecular profiling of liver tissue from children with biliary atresia has revealed a unique proinflammatory footprint related to activation of lymphocytes, particularly natural killer cells. Further work has shown that the T-cell infiltrate is oligoclonal in nature, suggestive of a specific provocation. CD8+ T cells are the predominant cell line in the infiltrate, with the suggestion from murine studies that there is a limited time window for an imbalance between cytotoxic T cells and an absence of regulatory T cells (T-regs, which suppress and inhibit natural killer cell expansion) to render the liver susceptible to biliary tract damage. In the mouse model of reovirus-induced biliary atresia, prevention of proliferation of T lymphocytes and the activation of NK cells through depletion of dendritic cells prevented the development of biliary atresia. Collectively, these findings support the premise that triggering of the cellular immune response is critical for development of biliary atresia.

Biliary atresia may thus represent a final common pathway of perinatal inflammatory destruction of the extrahepatic and potentially intrahepatic biliary tree. Indeed, neonatal systemic lupus can mimic biliary atresia. , It is not surprising that analysis of the initial biopsies of 47 infants with biliary atresia showed overexpression of genes associated with inflammation or fibrosis or both. The patients with a fibrosis signature were older and had decreased duration of transplant-free survival following KPE. When serum samples collected at the same time from 19 infants with biliary atresia and 19 with other forms of neonatal cholestasis were subjected to gel electrophoresis and tandem mass spectrometry, a combination of 11 proteins was able to discriminate between the two groups. , Among them were apolipoproteins CII and E, whose genes on chromosome 19 are regulated by farnesoid X receptor, which in turn is responsive to bile acids. Proinflammatory “positive” acute phase reactants, such as complement C3, were upregulated, and there was a relatively reduced level of “negative” acute-phase proteins such as prealbumin. The humoral response also may be activated, based on evidence from the murine model of biliary atresia for increased levels of antibodies to alpha-enolase, an enzyme ubiquitously expressed on a variety of cells, including biliary epithelial cells and hepatocytes.

Prompted by the similarities between biliary duct inflammation and graft-versus-host disease (GVHD), in 1992 Suskind and colleagues were first to describe maternal “microchimerism” in the livers of infants with biliary atresia. Human leukocyte antigen (HLA) class I matching was significantly more prevalent in 57 maternal-child pairs with biliary atresia than in 50 control pairs (odds ratio, 2.46), possibly providing for greater survival of the chimeric lymphocytes. This has led to the hypothesis that a “first hit” for biliary atresia is a GVHD-like interaction of maternal effector chimeric T lymphocytes engrafted within the fetus, with target fetal tissues. This hypothesis has been challenged by the absence of maternal microchimerism in regional lymph nodes of children with nonsyndromic biliary atresia. Regardless, the concept of some form of autoimmunity being active in biliary atresia remains of interest, supported by the occurrence of autoimmune disorders in 44% of family members of patients with biliary atresia.

Pathological Features

A diagnosis of biliary atresia is favored if a liver biopsy specimen exhibits bile ductular proliferation (neocholangioles), portal edema, and fibrosis but lacks sinusoidal fibrosis. Neutrophilic cholangitis and pericholangitis may or may not be present. The presence of bile plugs within bile duct lumina (which are distinct from the lumina of neocholangioles at the margins of portal tracts), portal edema, and increased numbers of bile duct profiles are helpful diagnostic findings with the largest odds ratio for predicting biliary atresia versus nonbiliary atresia ( Fig. 55.3 ). In fact, the extent of ductular reaction in wedge liver biopsies of children with biliary atresia, obtained at the time of portoenterostomy, is positively associated with improved 1-year survival of the native liver following KPE. Fifteen percent of biliary atresia cases show giant cell transformation of hepatocytes ( Fig. 55.4 ). The progressive fibrosis observed in biliary atresia may be abetted in part by epithelial-mesenchymal transition. ,

FIGURE 55.3, The portal region in an advanced case of extrahepatic biliary atresia demonstrates extensive ductular reaction with inflammation as well as severe cholestasis in the periportal parenchyma.

FIGURE 55.4, Lobular changes of “neonatal hepatitis” in extrahepatic biliary atresia.

The classic “obstructive” findings of bile duct proliferation and inspissated bile plugs are not always the result of atresia (see Box 55.8 ). Conversely, some cases of biliary atresia may show nonclassic features such as prominent portal tract arteries with medial hypertrophy, associated with disappearance of interlobular bile ducts and an absence of bile duct proliferation ( Fig. 55.5A ,B). , In keeping with the developmental theory of biliary atresia, a subset of affected patients exhibit ductal plate malformation (see Fig. 55.5C ). Recapitulating immature fetal portal tracts, this latter entity consists of portal tracts without an interlobular bile duct but with a centrally located hypertrophic hepatic artery and a peripheral rim of bile ductular structures. However, this phenomenon is not limited to the 20% to 25% of patients with a laterality defect (biliary atresia splenic malformation). As a result, biliary atresia can be difficult to diagnose at the time of percutaneous liver biopsy, even for the most experienced pediatric hepatopathologist (see Fig. 55.5D ). The age of the child at biopsy does not help resolve this difficulty. Nevertheless, the pathologist must advise the clinical team whether the histology of the liver biopsy justifies the patient being subjected to surgical intervention, with or without the performance of confirmatory intraoperative cholecystographic imaging before performance of the KPE. The alternative is that the pathologist advises the clinical team that the differential diagnosis of nonsurgical cholestatic disorders is of sufficient concern as to justify further noninvasive studies, rather than moving quickly to surgical intervention.

FIGURE 55.5, Nonclassic features of portal tracts in extrahepatic biliary atresia.

On performance of a KPE, the bile duct remnant is submitted for pathological examination. Typical findings in the remnant include fibrosis and obstruction of the lumen, a variable degree of periductal inflammation, and apoptotic degeneration of residual bile duct epithelium ( Fig. 55.6 ). These findings do not differ in syndromic cases of biliary atresia, cases discovered by prenatal ultrasound, and nonsyndromic postnatal biliary atresia, supporting the concept of a shared pathogenetic pathway. ,

FIGURE 55.6, Biliary atresia at surgery.

Natural History and Treatment

Before 1959, when the KPE procedure was introduced, biliary atresia was considered to be uniformly fatal by 2 years of age, with a median age at death of 10 months. The best survival rate in KPE patients with a native liver is 53% at 10 years, although reported native liver survival rates at 10 years are more commonly in the 35% range. , In the first report of the U.S. Biliary Atresia Research Consortium in 2006, a decline in total bilirubin at 3 months after KPE to less than 2 mg/dL was seen in 36.5% of children, and a favorable response (to <6 mg/dL) was seen in another 29%. Most patients in the excellent bilirubin response group had more favorable outcomes (as measured by “native liver survival”), a finding confirmed in more recent reports. , The 25% of children with a laterality defect fared more poorly, even if treated earlier. , Atresia of the common hepatic duct or ducts at the liver hilum (Ohi types 2 and 3) had a poorer prognosis than an atretic common bile duct (Ohi type 1), similar to the prognosis in biliary atresia patients with other malformations, ascites at surgery, or delay in surgery beyond day 75. Children operated on within the first 30 days had a 2-year 74% native liver survival rate. At King’s College Hospital in London, 56% of infants with EHBA cleared jaundice, and the 2-year survival rate in patients with their native liver was 65%. The 51 patients with biliary atresia splenic malformation or biliary cysts tended to be operated on earlier had poorer outcomes if the procedure was delayed. Nevertheless, the same group reported that even when surgery was performed on day 100 or even later, 45% of patients were alive with their native liver at 5 years, and 40% at 10 years, suggesting that patients may postpone transplantation by the KPE. In a report of native liver histology in 23 patients after a clinically successful KPE with an average follow-up of 4.2 years, resolution in histological cholestasis occurred in a majority of patients (83%), but fibrosis and bile duct proliferation persisted. In these “native liver survivors,” fibrosis had progressed to cirrhosis (stage 4 METAVIR) in 52% of patients, associated with the presence of portal hypertension.

Infants who do not respond to the KPE suffer from recurrent episodes of ascending cholangitis or sepsis caused by the immediate proximity of a bowel segment to the porta hepatis of the residual biliary tree. Regardless of the presence or absence of ascending cholangitis, the liver has a higher likelihood of progression cirrhosis with portal hypertension. Large bile lakes may form at the hilum, indicating a persistent effort at bile secretion by hepatocytes ( Fig. 55.7 ). The pathogenesis of progressive fibrosis in chronic cholestatic diseases, including biliary atresia, PFIC, and Alagille syndrome, has been linked to persistence of Hedgehog (Hh) signaling, which normally promotes embryonal duct differentiation and then shuts off as the liver matures. Immunohistochemical colocalization of the Hh transcription factor Gli2 and the mesenchymal markers vimentin and FSP1 in some of the reactive ductular epithelial cells in these diseases is evidence of reversal of mesenchymal-epithelial transition. Similar colocalization of FSP1 and cytokeratin 7 was shown in ductular epithelium, and this ductal cell derangement may lead to fibroplasia. Growth failure, malnutrition, deficiencies of lipid-soluble vitamins, and altered protein–energy and nutrient utilization are expected complications.

FIGURE 55.7, Liver specimens after Kasai portoenterostomy (KPE).

A failed KPE in a patient with biliary atresia is the most frequent indication for liver transplantation in infants and young children, accounting for 32.3% of all pediatric liver transplants in 2016. In such instances, prior performance of a KPE does not adversely affect outcomes of liver transplantation in children with biliary atresia, although patients subjected to laparoscopic KPE are less likely to have adhesions at the time of liver transplantation, when compared with those whose KPE was by open laparotomy. Patient survival after orthotopic transplantation for biliary atresia in the U.S. United Network for Organ Sharing from 1988 to 2003 was 85.8% at 10 years, with graft survival of 72.7%. The decision to transplant a patient with biliary atresia who has progressed to end-stage liver disease is critical; entry of a patient into the “intent-to-transplant” pathway is associated with excellent overall outcomes, with 97% survival at 5 years in one published series. Pathological examination of explanted livers for occult malignancy is crucial for all forms of chronic liver disease in children.

Neonatal Sclerosing Cholangitis

In addition to neonatal sclerosing cholangitis associated with ichthyosis (OMIM 607626) caused by CLDN1 mutations, several infants of consanguineous matings have been described with progressive sclerosing cholangitis from infancy. , An 8-month-old Arab boy reported by Bar Meir and associates had elevated immunoglobulins and anti–smooth muscle titers, but there were no such abnormalities in his mother. These few patients serve to highlight the concept of an intrahepatic form of biliary “atresia,” but they also manifest extrahepatic ductal lesions by imaging, similar to older patients with progressive sclerosing cholangitis. , Langerhans cell histiocytosis can lead to sclerosing cholangitis as part of a systemic process in young children (see Paucity of Intrahepatic Bile Ducts) ( Fig. 55.8 ). Therefore it may be most useful to consider that there is a continuum of inflammatory and sclerosing processes that may have a heritable basis in susceptibility and that show a variable degree of ductal obliteration. More recently DCDC2 mutations, a gene located on the cilia, has been shown as a cause of neonatal sclerosing cholangitis. ,

FIGURE 55.8, Langerhans cell histiocytosis in a 6-week-old infant with rash and jaundice.

Neonatal Hepatitis Syndrome

A neonatal hepatitis syndrome resembling biliary atresia was first described by Burns in 1817. By the mid-1950s, two distinct disease categories of cholestasis of infancy were established: biliary atresia, an inflammatory sclerosing syndrome of the extrahepatic biliary tract with hepatic features of bile duct obstruction, and neonatal hepatitis, a nonobstructive type of neonatal cholestatic syndrome with characteristic hepatitic features. At the time, neonatal cholestasis resulting from disorders of bile formation and transport was not yet recognized. In subsequent decades, enormous progress was made in the molecular characterization of neonatal cholestatic disorders. This entity is now best considered a clinicopathological syndrome in which there are many potential etiologies, including infections, anatomical or structural defects, metabolic and inherited disorders, hormonal insufficiency, and vascular, toxic, and immune causes (see Table 55.7 ).

A review of all biopsies designated neonatal giant cell hepatitis at the Johns Hopkins and University of Chicago tertiary hospitals from 1984 to 2007 found 62 cases (75% boys; average age, 2 months). Of the cases with clinical follow-up, half proved to be idiopathic. Ultimately, 8% of patients were found to have biliary atresia, 6% Alagille syndrome, 6% bile salt synthetic defects, and 16% hypopituitarism. At the Texas Children’s Hospital in Houston, 151 infants up to 3 months of age had a biopsy performed because of persistent conjugated hyperbilirubinemia. Fifty-nine percent were boys. However, among the 80 patients (53%) who were found to have extrahepatic biliary obstruction, 60% were girls. Forty-eight patients had neonatal hepatitis, with or without giant cells, one of whom proved to have CMV infection. One infant had A1AT deficiency, one hypopituitarism, one McCune-Albright syndrome, and one Langerhans cell histiocytosis. Sixteen had paucity of intrahepatic bile ducts, but none was syndromic (i.e., Alagille syndrome) at this early age. Disorders without a specific identifiable etiology now account for about 25% of the total.

Clinical Features

Persistent unconjugated hyperbilirubinemia raises the possibility of an inherited defect in bilirubin conjugation or hemolysis. Diagnostic assessment and therapeutic intervention is then critically important to avoid kernicterus and permanent neurological damage. In contrast, predominantly conjugated hyperbilirubinemia in newborns should prompt a rigorous diagnostic investigation for biliary atresia. An approach to the workup of neonatal hepatitis was presented earlier (see Cholestatic Pattern and Table 55.8 ).

Pathological Features

The hallmark histological finding of many neonatal liver disorders is the formation of large multinucleated hepatocytes (giant cells), which are part of the broader spectrum of lobular disarray in neonatal hepatitis ( Fig. 55.9 ). Nuclear inclusions in hepatocytes may suggest DNA viruses ( Figs. 55.10 and 55.11 ); those in red cell precursors may suggest parvovirus B19. In some cases, most of the liver parenchyma is transformed into giant cells, and this is referred to as syncytial giant cell hepatitis. When this histological picture predominates, electron microscopy can be extremely helpful and even pathognomonic if NPD type C is the cause. Of 40 infants evaluated for neonatal giant cell hepatitis in a Denver Children’s Series, 27% had NPD type C. Splenomegaly was a useful clue.

FIGURE 55.9, Neonatal hepatitis.

FIGURE 55.10, Cytomegalovirus (CMV) hepatitis.

FIGURE 55.11, Herpes simplex virus infection.

In neonatal hepatitis, the relative proportion of inflammatory cells is not considered informative because ongoing apoptosis may lead to extensive hepatocyte destruction and accumulation of residual lymphocytes and macrophages. In some cases, parenchymal inflammation is minimal or absent. In addition, parenchymal neutrophils are an uncommon finding. Occasional islands of erythropoiesis are a normal finding. In all instances, the biopsy should be examined carefully for evidence of viral cytopathic change.

Portal tracts may be inconspicuous, whether normal or abnormal. In general, at least five to seven portal tracts are required to consider the tissue sample adequate for histological evaluation. Ten or more portal tracts are preferred. Bile duct hypoplasia was more common among infants with hypopituitarism, compared with other conditions, in the Johns Hopkins/University of Chicago series. No other biopsy finding was helpful in distinguishing possible etiologies in that series. Exclusion of biliary atresia or A1AT storage disorder requires verification that most, if not all, portal tracts contain a terminal bile duct, a companion hepatic artery, and a portal vein and that bile ductular proliferation, edema, neutrophilic inflammation, and fibrosis are absent. Although the portal tracts in patients with neonatal hepatitis may be expanded by a mixed inflammatory infiltrate, this should not be confused with normal granulocytic hematopoiesis occasionally found within portal tracts.

Occasionally, massive hepatic necrosis, with marked accumulation of parenchymal iron, may be observed in patients with Down syndrome. One infant in the Texas Children’s Hospital series had an exceptional degree of hepatocellular necrosis, and the cause was pyruvate kinase deficiency. The observation of massive necrosis should also prompt consideration of NH (discussed earlier; see Table 55.7 ).

Differential Diagnosis

Once biliary atresia and A1AT mutations have been excluded on the basis of both clinical findings and percutaneous liver biopsy, then the differential diagnosis of neonatal hepatitis syndrome must be pursued. Infections of the newborn have been mentioned, and Chapter 47 addresses some of these disorders. Extensive lobular disease, with or without giant cells, may be seen in patients with biliary atresia (see Fig. 55.4 ). Therefore lobular changes should not divert attention from the features in the portal tracts. Second, nonclassic features of biliary obstruction may be observed in biopsy specimens from patients with biliary atresia, in which ductular reaction has not yet occurred and hepatic arteries exhibit hypertrophy of the media smooth muscle (see Fig. 55.5A ,B). Sometimes, alert detection of acholic stools prompts an early biopsy, but the time of onset and rate of progression can vary greatly. (For a detailed discussion of the differential diagnosis, see Cholestatic Pattern and Table 55.7 .)

Natural History and Treatment

The natural history of neonatal hepatitis depends heavily on the specific etiology ( Table 55.16 ). For instance, pharmacological intervention is imperative for many of the infectious disorders. Many of the other disorders, including hypopituitarism, tyrosinemia, and bile salt synthetic defects, benefit dramatically from prompt intervention. In about a quarter of all infants, no cause is found. The prognosis for “idiopathic” neonatal hepatitis syndrome is considered favorable; the mortality rate is 13% to 25%. Predictors of a poor clinical outcome include severe or prolonged (>6 months) jaundice, acholic stools, familial occurrence, and persistent hepatomegaly. Peak bilirubin level is not predictive of outcome. Sepsis is a devastating complication and is associated with a poor outcome. For infants whose liver disease resolves, the prognosis is quite favorable; most have no residual liver dysfunction. Many children benefit from a cholestatic gene panel evaluation that may pinpoint the exact defect (see Box 55.1 ).

TABLE 55.16
Natural History of Common Causes of Neonatal Jaundice
Etiology Natural History
Biliary atresia Death by 1 to 2 years of age without Kasai procedure or liver transplantation
α 1 -Antitrypsin storage disorder Progressive chronic liver disease with risk of cirrhosis
Neonatal infection Resolution of infection required for survival
Disorders of Carbohydrate Metabolism
Glycogen storage disease Maintenance of blood sugar levels enables survival
Risk of chronic renal disease
High probability of hepatic adenomatosis
Liver transplantation is curative in some forms
Galactosemia Dietary galactose restriction enables survival
Neurodevelopmental complications may persist
Fructosemia Avoidance of dietary fructose enables survival
Lifelong risk of metabolic crises on fructose exposure
Disorders of Amino Acid Metabolism
Tyrosinemia Pharmacological treatment enables survival
High lifetime risk of hepatocellular carcinoma
Disorders of Glycolipid and Lipid Metabolism
Niemann-Pick disease types A and C Type A: fatal outcome of progressive neurological disease
Type C: survival into adulthood possible; neurological compromise may occur
Hereditary Disorders of Bile Formation
Progressive familial intrahepatic cholestasis Biliary diversion may ameliorate; liver transplantation for cirrhosis, neoplasia; persistent diarrhea in type 1
Disorders of bile acid biosynthesis Dietary treatment with bile acids can ameliorate effects of disease
Idiopathic neonatal hepatitis Highly variable, most likely representing undiscovered genetic abnormalities
Cholestatic liver disease may persist into later childhood

The management of idiopathic neonatal hepatitis syndrome is supportive and includes adequate nutrition. Dietary measures, such as use of lactose-free, low-protein formulas, may mitigate further liver damage until the results of tests for galactosemia, hereditary tyrosinemia type I, and hypopituitarism, for example, become known. Elemental formulas containing medium-chain triglycerides help maintain caloric intake in severely ill patients. Infants with chronic cholestasis require fat-soluble vitamin supplementation. Pruritus associated with chronic cholestasis is difficult to treat in many patients.

Syncytial Giant Cell Hepatitis

In 1991, a putative novel form of hepatitis was first described in patients 5 months to 41 years of age. Clinical features of a severe type of hepatitis required liver transplantation in five patients; five others died of their liver disease. Liver histology revealed replacement of the parenchyma with abundant large syncytial multinucleated (giant) hepatocytes that contained up to 30 nuclei per cell. Electron microscopy revealed intracytoplasmic structures consistent with paramyxovirus nucleocapsids ( Fig. 55.12 ). Injection of liver homogenate from an affected patient into two chimpanzees led to an increase in the titer of paramyxovirus antibodies in one.

FIGURE 55.12, Paramyxovirus hepatitis in 4-year-old with 8 days of jaundice progressing to fulminant hepatic failure.

One year later, a separate report concluded that syncytial giant cell hepatitis may have a variety of causes, such as autoimmune hepatitis, hepatitis A virus (HAV) or HBV infection, Epstein-Barr virus infection, or, potentially, HCV infection. Three of the patients had fulminant hepatic failure; two others developed severe chronic hepatitis. However, a further seven patients fared quite well. Therefore syncytial giant cell hepatitis represents a tissue reaction pattern that may develop as a result of multiple causes and does not always imply an ominous prognosis. This interpretation has been supported by subsequent reports in which generalized bacterial sepsis, viral infection, toxoplasmosis, syphilis, listeriosis, and even tuberculosis were shown to induce a similar histological pattern of injury. Viral causes include CMV, rubella, herpes simplex virus, human herpesvirus 6, varicella, coxsackievirus, echovirus, reovirus 3, parvovirus B19, human immunodeficiency virus (HIV), enteroviruses, paramyxovirus, HAV, HBV, or (rarely) HCV; some adult patients were coinfected with HIV and HCV.

The Paramyxoviridae are divided into the subfamily Paramyxovirinae, containing Paramyxovirus (Sendai virus, parainfluenza virus type 3), rubella, mumps, parainfluenza virus type 2, and Morbillivirus (measles), and the subfamily Pneumovirinae, containing the Pneumovirus genus (respiratory syncytial virus). Therefore, even by ultrastructural examination, the differential diagnosis for syncytial giant cell hepatitis is quite broad.

Paucity of Intrahepatic Bile Ducts

Neonates with conjugated hyperbilirubinemia may exhibit paucity of small intrahepatic (interlobular) bile ducts. A variety of disorders can cause nonsyndromic duct paucity ( Table 55.17 ), including A1AT storage disorder and hypopituitarism. Langerhans cell histiocytosis may cause severe cholangitis with a dense infiltrate of eosinophils as well as CD207-positive and CD1a-positive histiocytes (see Fig. 55.8 ). In patients who survive infancy, duct loss can be severe. Sclerosing cholangitis caused by Langerhans cell histiocytosis has been reported in an adult. Some cases of paucity are idiopathic. Among congenital infections, CMV infection is the most important; viral inclusions may be found within bile duct epithelial cells (see Fig. 55.10 ). In severe neonatal hepatitis, paucity of bile ducts may be evident on liver biopsy specimens. Clinical presentation of severe cholestasis, either early in life or later in childhood, should always raise the possibility of syndromic paucity of bile ducts (Alagille syndrome).

TABLE 55.17
Causes of Nonsyndromic Paucity of Bile Ducts
Modified from Roberts EA. Neonatal hepatitis syndrome. Semin Neonatol . 2003;8:357–374.
Infants Adolescents and Adults
  • Prematurity

  • Immune-mediated causes

  • Infection

    • Primary biliary cirrhosis

    • Cytomegalovirus

    • Primary sclerosing cholangitis

    • Rubella

    • Liver allograft rejection

    • Syphilis

    • Graft-versus-host disease

    • Hepatitis B virus

  • Metabolic

  • Metabolic disorders

    • Cystic fibrosis

    • α 1 -Antitrypsin storage disorder

  • Toxic insults

    • Niemann-Pick disease type C

    • Cystic fibrosis

    • Zellweger syndrome

    • Mitochondrial cytopathies

    • Byler syndrome

    • Prune-belly syndrome

    • Hypopituitarism

    • MYO5B liver disease

  • Genetic: chromosomal disorders

    • Trisomy 18, trisomy 21

    • Partial trisomy 11

    • Monosomy X

    • Neonatal sclerosing cholangitis-ichthyosis

  • Immune-mediated causes

    • Liver allograft rejection

    • Graft-versus-host disease

  • Severe idiopathic neonatal hepatitis

  • End-stage extrahepatic atresia

α 1 -Antitrypsin Deficiency

A1AT is a hepatic storage disorder that mostly manifests later in life (see later discussion). However, 11% of individuals with PiZZ mutations in SERPINA1 , the A1AT gene, may develop conjugated hyperbilirubinemia in infancy.

A1AT storage disorder can mimic the histological appearance of biliary atresia, with portal tract changes that include edema, scant acute inflammation, proliferating bile ductules, and bile plugs within bile duct lumina ( Fig. 55.13 ). Characteristic globular inclusions of staining material (PAS) with diastase digestion (d-PAS) in periportal hepatocytes are typically not present for weeks or months, but immunohistochemistry for the protein is more sensitive, as is electron microscopy for demonstration of the protein within endoplasmic reticulum. Bile pigment, hemosiderin, and copper-binding protein may be present within periportal hepatocytes, all of which are highlighted as fine granules in tissue sections stained with d-PAS. Protease inhibitor typing by serum immunoelectrophoresis is required for diagnosis.

FIGURE 55.13, α 1 -Antitrypsin storage disease, acute.

Syndromic Paucity of Bile Ducts: Alagille Syndrome

Clinical Features

Some infants and young children with conjugated hyperbilirubinemia have associated facial dysmorphism and cardiovascular, vertebral, and ocular malformations. The combination of extrahepatic abnormalities and deficiency of interlobular bile ducts is termed Alagille syndrome . Alagille syndrome is an autosomal dominant disorder with an estimated frequency of 1 in 70,000 live births, increased to an incidence of 1 in 30,000 live births in the molecular era. Sporadic cases account for 45% to 50%. The mortality rate is 15% to 20%. Deaths are mostly caused by hepatic or cardiovascular complications.

Before the advent of genetic testing, a diagnosis of Alagille syndrome required demonstration of paucity of interlobular bile ducts and clinical evidence of at least three major of the following abnormalities: characteristic facies, posterior embryotoxon, butterfly vertebrae, renal disease, and cardiac anomalies. However, bile duct paucity is evident in only 60% of infants who undergo biopsy before 6 months of age. Furthermore, there is wide phenotypic variability in patients with this disorder, ranging from mild subclinical findings to complete liver failure and complex congenital heart disease. , Most patients with hepatic involvement are seen clinically within the first 6 months with jaundice, hepatomegaly, pruritus, and failure to thrive. Laboratory studies reveal conjugated hyperbilirubinemia and increased serum levels of GGT, alkaline phosphatase, bile acids, and cholesterol. Serum ALT and AST levels may be normal or mildly elevated. Currently, genetic testing is almost always conclusive and is included as part of the cholestatic gene panel.

Pathogenesis

Mutations in Jagged1 (JAG1), a ligand in the Notch signaling pathway, have been identified in 94% of patients with Alagille syndrome. Mutations in the NOTCH2 gene, which encodes a receptor for Jagged1, have been identified in some JAG1 mutation–negative patients. , It is hypothesized that defects in JAG1 result in an arrest of branching and elongation of bile ducts during postnatal liver growth. In support of this theory, Libbrecht and associates reported a case in which an explanted liver from a 16-year-old patient with Alagille syndrome demonstrated paucity of bile ducts in peripheral liver parenchyma but normally developed bile ducts in the perihilar areas. Deletions in a single JAG1 allele are sufficient to cause ALGS, suggesting haploinsufficiency as the disease-causing mechanism. Recent mouse models with mutant JAG1 that have been developed by Andersson et al. and Adams et al. will help better define the mechanism of ALGS as it showed binding of NOTCH2 rather than NOTCH1 by JAG 1 Ndr in these mice. The interaction of the NOTCH and WNT pathways in biliary epithelial development is of great interest for aberrant organogenesis and response to injury.

Pathological Features

To diagnose paucity of bile ducts, the liver biopsy specimen should contain at least 7 portal tracts, and preferably at least 10. Normally, on routinely stained tissue sections (H&E or Masson trichrome) of mature liver tissue, approximately 90% of portal tracts contain an interlobular bile duct, either as a single duct or as multiple duct profiles, paired in close apposition to branches of the hepatic artery of similar outside diameter. The presence of multiple portal tracts containing arteries and veins but not bile ducts should prompt consideration of bile duct paucity, which is defined by an absence of, or marked reduction in, the number of interlobular bile ducts within portal tracts. In pediatric patients, a bile duct-to-portal tract ratio of less than 0.8 is often used as a cutoff point between normal and abnormal, although the number of bile ducts may be normally (physiologically) low in preterm infants. However, in any age group, a ratio of less than 0.4 is strongly suggestive of bile duct paucity. When making this assessment, large portal tracts should not be considered in the equation. Furthermore, incomplete portal tracts that are partially transected by the biopsy should be excluded from the analysis because absence of a bile duct may simply represent a sampling artifact. Specimens from patients with Alagille syndrome are conspicuous for their lack of portal tracts and parenchymal inflammation, but they may have abundant giant cells, as in neonatal hepatitis ( Fig. 55.14 ).

FIGURE 55.14, Alagille syndrome.

Immunohistochemical staining for cytokeratin 7 or 19 is useful to highlight bile duct epithelium. Because hepatocytes in cholestatic livers often acquire cytokeratin 7 immunoreactivity (see Fig. 55.14D ), cytokeratin 19 is preferred for quantitating bile ducts. Immunoreactive ductular profiles at the margins of portal tracts should not be confused with true interlobular bile ducts (see Fig. 55.1 ). A persistent lack of canalicular CD10 immunopositivity, although physiological in infants younger than 2 years of age, is a characteristic finding in Alagille syndrome. The Masson trichrome stain is also helpful in identifying small portal tracts because of its ability to detect delicate amounts of connective tissue. These small portal tracts should be included in the portal tract count when one is assessing bile ducts.

In the evaluation of a liver biopsy specimen for potential paucity of bile ducts, the pathology report should indicate how many portal tracts and bile ducts were identified. The staining method should also be reported. Paucity of interlobular bile ducts is not a specific disorder; instead, it represents a pathological feature that may have a variety of causes.

Differential Diagnosis

When Alagille syndrome cannot be distinguished from biliary obstruction on liver biopsy, and the clinical and radiological features fail to detect the extrahepatic manifestations in an infant with Alagille syndrome, patients have at times still been treated by KPE. Even at a major center with extensive experience with these diseases, 4.4% of patients were mistakenly operated on, and afterward their liver disease progressed more rapidly to end-stage cirrhosis. Alagille histology may show an overlap with EHBA with marked ductular proliferation and fibrosis but shows absence of interlobular bile ducts. Because Alagille disease progression is highly variable, every effort must be made to avoid surgery.

Caution is also required in the interpretation of hepatobiliary scintigraphy and cholangiographic studies. Typically, both extrahepatic and intrahepatic bile ducts in patients with Alagille syndrome are markedly hypoplastic; therefore on hepatobiliary scanning, one may not see excretion of radioisotope into the intestines (mimicking biliary atresia). Similarly, intraoperative cholangiography may not necessarily demonstrate opacification of the proximal extrahepatic ducts. These diagnostic pitfalls underscore the importance of a careful and full physical examination and radiological evaluation in patients with suspected Alagille syndrome.

No genotype-phenotype correlation with severity for JAGGED1 mutations was found among 33 patients with Alagille syndrome who were older than 10 years of age. Those children with elevated total bilirubin (>6.5 mg/dL), conjugated bilirubin (>4.5 mg/dL), and cholesterol (>520 mg/dL) before 5 years of age had significantly worse liver disease later. Sequencing of the entire coding region of JAG1 is now available and should be used in patients in whom there is a strong clinical suspicion and in family members of probands with mutations.

Natural History

Alagille syndrome is a benign illness in many children. However, young children with protracted severe jaundice usually have a poorer prognosis. Between 10% and 50% of patients with Alagille syndrome eventually progress to cirrhosis and liver failure. Cases of HCC in Alagille syndrome have been described, as in every chronic cholestatic disease of infants. The overall mortality rate from Alagille syndrome is estimated to be 20% to 25%; death occurs mainly from cardiac disease, intercurrent infection, or progressive liver disease. Liver transplantation is warranted for patients with severe hepatic disease. Because of the presence of multiple organ defects in Alagille syndrome, the outcomes of liver transplantation are poorer than for patients with EHBA, especially with regard to death during the first 30 postoperative days. No pretransplant factors were identified among the 87% of patients who survived to 1 year, compared with those who did not.

Hereditary Disorders of Bilirubin Metabolism

Bilirubin is the end product of heme degradation, which is derived predominantly from the breakdown of senescent erythrocytes by the mononuclear phagocytic system. Heme oxygenase within reticuloendothelial cells oxidizes heme to biliverdin, which is then reduced to bilirubin by biliverdin reductase. Bilirubin is released into the circulation and binds to serum albumin. Uptake of bilirubin by hepatocytes occurs via a carrier-mediated system at the level of the sinusoidal membrane. Bilirubin is then conjugated with either one or two molecules of glucuronic acid by bilirubin uridine diphosphate (UDP)-glucuronosyltransferase (i.e., UGT1A1) within the endoplasmic reticulum. It is then excreted as water-soluble, nontoxic bilirubin glucuronides into bile.

Crigler-Najjar Syndrome and Gilbert Syndrome

Classification and Pathogenesis

The hepatic conjugating enzyme UGT1A1 is a product of the UGT1A1 gene, which is located on chromosome 2q37. It is a member of a family of UDP-glucuronosyltransferases (UGTs) that catalyze the glucuronidation of an array of substrates such as steroid hormones, carcinogens, and drugs. The various types of UGTs are distributed within a wide range of tissues, including liver, kidney, intestines, skin, lung, olfactory epithelium, and testis. UGT1A1 is located primarily within the smooth and rough endoplasmic reticulum of hepatocytes as a single isoform that catalyzes the glucuronidation of bilirubin to form its monoglucuronidated and diglucuronidated forms. In humans, two members of the UGT1 family possess the capability to glucuronidate bilirubin in vitro, but only one isoform is physiologically relevant in vivo. The bilirubin glucuronidating isoform is termed UGT1A1 because it is generated from the exon 1A of the UGT1 gene locus. Multiple mutations in UGT1A1 cause hereditary unconjugated hyperbilirubinemia: Crigler-Najjar syndrome types I and II and Gilbert syndrome.

Clinical and Pathological Features

In Crigler-Najjar syndrome type I, the liver UGT1A1 enzyme is completely absent. Colorless bile contains only trace amounts of unconjugated bilirubin. Serum unconjugated bilirubin reaches very high levels, leading to severe jaundice and icterus. Without liver transplantation, this condition is invariably fatal (because of kernicterus), usually within 18 months of birth. In a recent review of 22 patients undergoing liver transplant for Crigler-Najjar syndrome type I, the pathological features showed canalicular cholestasis and significant pericentral sinusoidal, periportal, and mixed patterns of fibrosis in almost 41% of the patients. The fibrosis appeared to be progressive and more in older children at the time of transplant. This suggests the need for early recognition of this disease and intervention to arrest progression of fibrosis and need for earlier liver transplantation. , This has led to attempts to use AAV8 gene therapy to ameliorate the changes with preliminary success in mouse models.

Crigler-Najjar syndrome type II is a less severe, nonfatal disorder in which the hepatic level of UGT1A1 enzyme activity is greatly reduced but not absent, and the enzyme is capable of forming only monoglucuronidated bilirubin. In contrast with the type I syndrome, the only major clinical consequence is the presence of extraordinarily yellow skin caused by moderate to high levels of circulating unconjugated bilirubin. Phenobarbital treatment may improve bilirubin glucuronidation by inducing hypertrophy of the hepatocellular endoplasmic reticulum.

Gilbert syndrome is a relatively common benign, inherited condition. Affected patients have mild, fluctuating hyperbilirubinemia in the absence of hemolysis or liver disease. Typically, hepatic bilirubin glucuronidating activity is approximately 30% of normal levels. In most patients, the genetic defect consists of two extra bases (TA) in the TATAA element (TATA box) in the promoter region of the UGT1A1 gene. This creates an A(TA) 7 TAA element, rather than the normal A(TA) 6 TAA of the UGT1A1 gene (“an extra TA in the TATA box”), and results in reduced expression of UGT1A1. In some cases, patients are heterozygous for missense mutations in the UGT1A1 gene. Gilbert syndrome affects 3% to 10% of the population. Mild hyperbilirubinemia may go unrecognized for many years and is not associated with functional derangements of the liver. When detected in adolescents or adults, it is typically in association with an unrelated stress, such as intercurrent illness, strenuous exercise, or fasting, that reduces hepatic levels of the obligate cofactor for UGT1A1, UDP-glucuronic acid. Gilbert syndrome has no clinical consequence except for anxiety related to persistent jaundice. A 2007 study suggested that diabetic patients with coexistent Gilbert syndrome have a lower incidence of vascular complications and a reduction in serum markers of oxidase stress and inflammation, possibly because of the antioxidant effect of bilirubin.

In all disorders of bilirubin conjugation other than Crigler-Najjar type I, the liver is morphologically normal by both light and electron microscopy.

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