Liver in Systemic Disease


Introduction

This chapter considers hepatic manifestations resulting from diseases of other organs, where liver dysfunction develops secondarily but can be of clinical or morphological significance. Those diseases will be considered here on a systemic basis. Conversely, nonspecific reactive hepatitis, granulomas and steatosis are commonly encountered hepatic morphological changes among whose diverse causes, nonhepatic diseases merit particular consideration. Steatosis is dealt with in Chapter 5 , while the others will be reviewed here, together with an account of the diversity of morphological changes encountered related to mass lesions in the liver.

Nonspecific reactive hepatitis

This is a microscopically variable and patchy nondescript inflammatory reaction associated with parenchymal turnover in the liver. There is no specific clinical manifestation or derangement of liver function tests beyond a mild increase of serum aminotransferase levels. It is a convenient descriptor rather than a diagnostic term, and use in clinical practice presupposes that reasonable clinicopathological exclusions have been made, including appropriate serological and virological screening, such as for hepatitis C virus (HCV) RNA in serum. As first coined by Popper and Schaffner, in an era predating many such diagnostic tests, typical settings included resolving hepatitis, recent febrile illness and inflammation somewhere in the splanchnic bed. Identical changes may also be localized around focal liver injuries such as vascular or space-occupying lesions.

Typically, small portal tracts contain lymphocytes and macrophages (including ceroid-laden macrophages), with characteristic variability between portal tracts ( Fig. 15.1A, B ). Granulocytes and plasma cells are absent or rare; cholangitis and interface hepatitis are absent. The lobules show prominent Kupffer cells with hypertrophy, often with ceroid-laden cytoplasm and clustering, in perivenular areas and randomly distributed foci of hepatocyte cell death ( Fig. 15.1C, D ). Hepatocyte necrosis may affect single or contiguous cells (with reticulin collapse), but without zonal predilection. Accompanying signs of increased hepatocellular turnover include occasional mitotic figures, limited regenerative plate widening and greater variability of cell and nuclear size. A major differential diagnosis is chronic viral HCV infection (see Chapter 6 ), requiring serum RNA determination for exclusion, and which may lack helpful signs such as a more diffuse portal hepatitis, appreciable interface hepatitis, lymphocytic lobular inflammation or fibrosis. Resolving drug injury may similarly present only mild nonspecific changes. Autoimmune hepatitis (especially if a flare is remitting by the time a biopsy is done) can manifest with mild portal and lobular inflammation and only sparse plasma cells, while serum autoantibodies may only appear later. Numerous infections, including those caused by bacteria, spirochaetes, protozoa and nonhepatotropic viruses (see Chapter 7 ), can appear as nonspecific reactive hepatitis if more characteristic features such as granulomas and/or microabscesses fail to be sampled. Eosinophilia merits consideration of a parasite, from which pigments (e.g. haemozoin) or parts may be evident on further scrutiny. Viral infection in other organs, such as influenza, can be accompanied by a ‘collateral’ T cell-mediated hepatitis without viral antigen directly present in liver. A nonspecific reactive hepatitis is common in human immunodeficiency virus (HIV) infection (see Chapter 7 ) and is associated with chronic inflammatory disorders of other organs/systems (e.g. coeliac disease). Finally, lymphoproliferative disease can mimic hepatitis, in the form of an atypical granulomatous portal infiltrate or an atypical sinusoidal lymphocytosis.

Figure 15.1, Nonspecific reactive hepatitis: these illustrations are from a biopsy taken from a patient with a carcinoma in the ileocaecal region. (A) Portal tract showing a very light chronic inflammatory cell infiltrate. (B) Portal tract showing a mixed lymphocyte and macrophage infiltrate with some periportal spillover. (C) Sinusoidal infiltrate of inflammatory cells with reactive Kupffer cell hyperplasia. (D) Granulomatoid aggregate of lymphocytes and macrophages. (Haematoxylin and eosin [H&E].)

Space-occupying lesions in the liver

Mass lesions produce obstructive and pressure effects on surrounding liver, which, although accompanied by nonspecific reactive changes, can be distinctive when encountered in biopsies targeting the lesion. , Small portal tracts in particular show changes that presumably reflect local interference with bile flow, comprising oedema and neutrophil infiltrates around interlobular ducts, with periportal ductular reaction and neutrophils ( Fig. 15.2A, B ). Likewise, compression may explain the typical sinusoidal dilation that is usually most striking in perivenular areas, but which is not usually accompanied by hepatocyte atrophy or hepatic vein abnormality ( Fig. 15.2C ). Moderately dense portal inflammation (with or without interface hepatitis) is another common finding in the adjacent nonlesional liver tissue surrounding metastases (for example, around metastatic colorectal adenocarcinoma in resection specimens).

Figure 15.2, Liver biopsy from a patient suspected of having metastatic tumour. (A) There is some sinusoidal dilatation and the portal tract shows oedema and inflammation which in (B) is associated with an acute cholangiolitis. (C) There is more marked sinusoidal dilatation evident in this area of the biopsy. (H&E.)

Some patients with thrombophilia or occult hepatic vascular anomalies can present incidentally with one or more ill-defined parenchymal lesions on imaging. These ‘pseudotumours’ reflect reactive parenchymal changes associated with local thrombo-obliterative changes in small veins. Microscopically there is irregular and close disposition of small portal tracts, portal venular absence, atrophy or herniation, portal or hepatic venous thrombo-obliteration and focal arterialization. Associated ductular reaction and patchy sinusoidal congestion or dilation can draw attention to the often ill-defined lesional area on biopsy.

Primary or metastatic neoplasms in liver may induce localized hyperplastic change within the surrounding parenchyma, termed ‘peritumoral hyperplasia’. The significance for pathologists is misinterpretation as liver cell adenoma or focal nodular hyperplasia when a targeted biopsy has missed the index neoplasm. The hyperplastic changes extend a few to several millimetres deep, over part of or the entire lesion, and comprise broadened plates and rosettes of hepatocytes showing glutamine synthetase immunopositivity, with capillarization of adjacent sinusoids (CD34 immunopositive). The outer margin is delimited by atrophy and congestion in surrounding parenchyma. The hyperplasia is thought to be a local response to ‘spillover’ perfusion with arterial-enriched blood from tumour-associated arteries, with a possible arteriolar buffer response compensating for upstream neoplastic occlusion of portal venules.

Localized peritumoral steatosis may also occur around insulinoma metastases in liver, attributable to the effect of tumour-secreted excess insulin on the hepatocytes. In analogous fashion, distinctive subcapsular steatosis may occur in patients receiving insulin administration through peritoneal dialysates , and can be misinterpreted radiologically as infarcts. Aberrant venous drainage from gallbladder or pancreas may produce so-called ‘pseudolesions’ of focal fatty change, or focal fatty sparing near the gallbladder bed in steatotic liver, which can be mistaken radiologically for invasive tumour. Adrenal rest tumour in liver may also mimic metastasis on imaging. Liver abscess usually originates with biliary or abdominal infection, including after surgical intervention, and is facilitated by immunosuppressive states. Some examples follow trauma or secondary infection of a pre-existing mass lesion such as metastasis or cyst.

Mass lesions can cause more widespread changes due to compression of major vascular or biliary structures. Diffuse infiltration of liver parenchyma by carcinoma or non-Hodgkin lymphoma can cause acute liver failure in previously asymptomatic patients; imaging may resemble cirrhosis and while massive hepatomegaly can be a clue, prospective diagnosis often requires transjugular liver biopsy. Paraneoplastic hepatic manifestations of malignancy include vanishing bile duct syndrome in Hodgkin disease (see Chapter 9 ). Chemotherapy or other modalities of treatment can induce changes in the nontumourous liver parenchyma, including steatohepatitis, nodular regenerative hyperplasia and sinusoidal obstruction syndrome (see Chapter 12 ); it is possible that those microvascular injuries may facilitate subsequent development of focal hyperplastic lesions. Transarterial chemoembolization (TACE) for liver malignancy can be complicated by hepatic artery branch rupture or thrombosis ( Fig. 15.3 ) and subsequent ischaemic cholangitis or liver abscess (see Chapters 9 and 12 ). Liver resections for malignancy can show atrophy of the nontumourous liver due to planned preoperative portal vein branch embolization (using methods including injected coils, particles or glues). This is done to stimulate growth of the future liver remnant to a size sufficient to permit safe resection. Box 15.1 summarizes the potential liver changes secondary to space-occupying lesions with or without iatrogenic intervention.

Figure 15.3, Segment II hepatic artery rupture and thrombosis from a prior transarterial chemoembolization procedure.

Box 15.1
Potential liver changes related to tumours and other space-occupying lesions

Parenchyma

  • Peritumoral compression effect (portal oedema, cholangiolitis, sinusoid dilatation)

  • Peritumoral hyperplasia

Biliary injury

  • Bile duct obstruction

  • Vanishing bile duct syndrome (e.g. Hodgkin disease)

Occlusive venopathy

  • Inferior vena cava, hepatic vein

  • Veno-occlusive disease (lympho/myeloproliferative disorders)

  • Portal vein obstruction

Uncommon injury

  • Diffuse infiltration by malignancy—acute liver failure (e.g. breast carcinoma)

Changes attributable to chemotherapy, local ablation therapy or embolization

  • Nodular regenerative hyperplasia

  • Steatohepatitis

  • Sinusoidal obstruction syndrome

  • Ischaemic cholangitis

  • Abscess

  • Lobar/segmental atrophy

Hepatic granulomas

Liver granulomas may arise as part of primary liver disease, or multisystem granulomatous disease, or may be a nonspecific response to extrahepatic disease. Granulomas are organized clusters of mature macrophages, which develop in response to persistent stimuli that are required to maintain them. Granulomas can be considered according to the turnover rate of constituent macrophages as high or low turnover, relating to the toxicity of the inciting agent. Most foreign body granulomas are low turnover, although exceptions include Beryllium (see Chapter 12 ), while infectious granulomas tend to be high turnover. Different histological appearances of granulomas and their correlations are summarized in Table 15.1 . So-called microgranulomas—loose lobular clusters of a few to several macrophages—are very nonspecific.

Table 15.1
Histological patterns of granuloma
Pattern Description Cause
Epithelioid granuloma Discrete with distinct edges, with or without necrosis (fibrinoid, caseous, eosinophilic). Presence of eosinophils suggests drug, parasites, but common in primary biliary cirrhosis
Fibrosis formation in sarcoid; reticulin preservation not specific for sarcoid
Infectious: tuberculosis, brucellosis, Mycobacterium avium , listeriosis, tuberculoid leprosy, tertiary syphilis, schistosomiasis, fungal infection, viral infection
Noninfectious: drug reaction, foreign body, sarcoidosis, primary biliary cholangitis, Hodgkin disease, chronic granulomatous disease
Microgranuloma Small aggregates of histiocytes and lymphocytes with apoptotic debris, often seen as part of nonspecific reactive hepatitis Listeriosis, typhoid fever, any of the above
Suppurative Stellate microabscess or mixed suppurative-granulomatous inflammation Cat-scratch disease ( Bartonella ), Yersiniosis, tularaemia (typically stellate microabscess), listeriosis, melioidosis, actinomycosis, fungi
Fibrin ring granuloma Fat vacuole within the granuloma and surrounding ring of fibrin Q fever, toxoplasmosis, salmonellosis, CMV, EBV, leishmaniasis, noninfectious (drug, lupus), Hodgkin disease
Lipogranuloma Granulomas containing lipid Steatohepatitis, mineral oil in food
Foamy macrophages Aggregates of foamy macrophages Mycobacterium avium intracellulare (immunocompromised), Whipple disease, lepromatous leprosy
CMV , Cytomegalovirus; EBV , Epstein–Barr virus.
Modified from Lamps LW. Hepatic granulomas, with an emphasis on infectious causes. Adv Anat Pathol. 2008;15(6):309–318, with permission.

Lipogranulomas associated with liver steatosis , represent a macrophage reaction to fat spilled from injured or necrotic lipid-containing hepatocytes. They comprise loose clusters of macrophages with extracellular and intracellular lipid, associated with scattered small lymphocytes in areas of hepatocyte steatosis ( Fig. 15.4A ). Multinucleate giant cells are rare. Mineral oil lipogranulomas in nonfatty liver occur in portal tracts more often than parenchyma, where they are commonly perivenular. They appear as a cluster of variably sized extracellular lipid droplets (sometimes larger than a hepatocyte), surrounded by a light infiltrate of lymphocytes and macrophages, sometimes with focal fibrosis ( Fig. 15.4B ). Spleen and lymph nodes can also be involved. The lipid may come from processed food. , Gold granules have been identified in lipogranulomas of patients with rheumatoid arthritis (RA), the lipid component being attributed to oily vehicle for the gold. These lesions are generally of little consequence and are often incidental findings. However, Keen et al. described two patients in whom extensive mineral oil lipogranulomatosis led to venous outflow obstruction. Disseminated mineral oil granulomatosis has also followed cosmetic self-administration of mineral oil injections. In contrast, lipopeliosis refers to the accumulation of coalescent lipid droplets within sinusoids and the space of Disse, related to profuse release from necrotic fatty liver. This phenomenon is well described due to preservation–reperfusion injury at transplantation ( Fig. 15.5 ) (see Chapter 14 ), but as early as 1929 was shown to cause fatal hepatogenic fat embolism to lung after self-poisoning with carbon tetrachloride by an alcoholic with fatty liver. Hepatogenic pulmonary fat embolism is also reported after other nontraumatic injuries to very fatty liver, such as systemic hypotension or fulminant acute hepatitis.

Figure 15.4, Hepatic granulomas. (A) Lipogranuloma in a fatty liver; the larger one above comprises a central fat globule surrounded by a cuff of inflammatory cells. (H&E.) (B) Mineral oil granulomas, found incidentally at autopsy, are present both in a portal tract and in the perivenular zone; there is also chronic inflammation of the portal tract, and some increase in fibrous tissue is present around the parenchymal collection of lipid-laden macrophages. (Haematoxylin-phloxine-saffron.) (C) Fibrin ring granuloma from a patient with Q fever; there is a cuff of inflammatory cells in which neutrophils are prominent. (Martius Scarlet Blue stain.)

Figure 15.5, Lipopeliosis. Extracellular release of lipid-rich material from necrosis of perivenular steatotic hepatocytes; this is an allograft with primary nonfunction attributed to preservation-related injury, the effects of which have been exacerbated by the donor fatty liver disease. (H&E.)

Fibrin-ring (or doughnut) granulomas are a distinctive but nonspecific response to injury, first noted in association with Q fever. There is a shell of circumferential fibrin ( Fig. 15.4C ) within or at the margin of the granulomas and sometimes a central fat vacuole. Fibrin-ring granulomas are documented in many conditions, , including Boutonneuse fever, allopurinol hypersensitivity, cytomegalovirus (CMV) infection, leishmaniasis, hepatitis A, staphylococcal infection, Epstein–Barr virus (EBV) infection, systemic lupus erythematosus (SLE) and giant cell arteritis. ,

The following account is confined to a discussion of epithelioid granulomas . These are characterized by clustered macrophages that have undergone phenotypic modulation to a proinflammatory microbicidal and secretory state. The macrophages show abundant pale eosinophilic cytoplasm and may fuse to form multinucleate giant cells. The periodic acid-Schiff (PAS) stain highlights the distribution and density of parenchymal granulomas. Kupffer cells can slowly redistribute to form granulomas if directly involved, for example by intracellular infection, but newly recruited macrophages otherwise come from circulating bone marrow-derived monocytes. Activated antigen-specific CD4 T cells are central to the complete ‘epithelioid’ phenotypic transformation of macrophages, which is effected in tuberculosis (TB) by tumour necrosis factor and other Th1 inflammatory cytokines, interferon-γ and interleukins (IL)-1 and -12; the granulomatous Th2 response to Schistosome eggs is promoted by T h2 cytokines such as IL-4 and -13. , T lymphocytes traffic continuously within the granuloma, including areas of necrosis. Other inflammatory cells populating granulomas include dendritic cells, B lymphocytes, granulocytes and innate lymphoid cells.

The prevalence and causes of liver granulomas depend heavily on case-mix, geographic location and reporting era but are reported in 2% to over 10% of native liver biopsies. The causes are many and most are uncommon ( Tables 15.2 and 15.3 ); few show distinguishing morphological features. , ,

Table 15.2
Hepatic granulomas—reported infectious causes and major features
Route/pathogenesis (geographical distribution) Morphology Staining/test
Bacterial
Actinomycosis Spread from caecum, appendix Abscess, granuloma basophilic bacilli in grains Gram, Grocott
Nocardiosis Immunocompromised Abscess granuloma basophilic bacilli in grains Like actinomyces but also weakly acid-fast positive. Wade-Fite
Bartonellosis Cat scratch, skin and local lymphadenopathy Stellate microabscess, granulomatous edge Warthin Starry
Borrelliosis (Lyme disease) Tick-borne, relapsing fever Granuloma, Kupffer cell hyperplasia, hepatocyte necrosis/mitosis Warthin Starry or Dieterle (10–20 µm) in sinusoids
Brucellosis Ruminants, dairy products. Great mimic Micro/epithelioid cell granuloma, ± necrosis Serology
Listeriosis Pregnancy.
Neonatal septicaemia (transplacental). Immunocompromised
Microabscess, necrotic granuloma Gram-positive rods
Melioidosis Soil, water (India, Southeast Asia) Abscess. Necrotic granuloma Gram, Giemsa-positive rods
Staphylococcus Chronic granulomatous disease Portal macrophages, parenchymal necrotizing granuloma Gram-positive cocci
Syphilis Sexual/vertical Granuloma in secondary, gumma in tertiary Warthin Starry
Tularaemia Tick bite, mosquitoes, mammalian reservoir. Ulceroglandular at site of entry (Northern hemisphere: North America, Russia, Scandinavia) Liver involvement in bacteraemic phase (can replicate in hepatocytes) Gram-negative
Typhoid Faecal–oral transmission Non-necrotizing granuloma, becoming necrotic in fastigium phase Gram-negative
Whipple disease Rare. Possibly abnormal response to the bacterium (North America, Western Europe) Noncaseating epithelioid granulomas. Foamy macrophages PAS/PAS-D+ve macrophages. Gram-positive
Yersiniosis ( Yersinia enterocolitica ) Contaminated food (pork). Mesenteric adenitis. Paediatric (North America/Europe) Suppurative granuloma. Abscess formation Gram-negative. Requires iron to survive (siderosis)
Mycobacterial
Tuberculosis Pulmonary/intestinal entry Granuloma with/without necrosis depending on reactive or anergic state Ziehl–Neelsen
Atypical mycobacteria (e.g. M. avium intracellulare ) Immunocompromised host Granuloma Ziehl–Neelsen, foamy macrophages stuffed with mycobacteria
BCG immunization and immunotherapy Immunization/immunotherapy Granuloma Ziehl–Neelsen
Leprosy (lepromatous and tuberculoid) Nasal oral secretion. Skin and peripheral nervous system involvement (Tropic/subtropic) Tuberculoid granuloma with no AFB; lepromatous foamy cells with AFB. Amyloid Wade-Fite
Rickettsial
Boutonneuse fever Tick-borne (Mediterraneum, Africa, India) Granuloma or focal hepatocyte necrosis Immunohistochemistry
Q fever Inhalation. Pneumonia, hepatitis, fever NOS (North America typically) Fibrin ring granuloma, but other types can occur Serology/PCR on peripheral blood
Rickettsia conorii infection Tick-borne (America). Granuloma NOS, vasculitis Immunohistochemistry
Chlamydial
Lymphopathia venereum Sexual transmission (Tropics) Granuloma. Perihepatitis Cell culture, direct immunofuorescence. PCR
Psittacosis Air-borne Microgranuloma Serology, direct Immunofluorescence and PCR
Fungal
Aspergillosis Opportunistic Abscess/granuloma
Hyphae invading vessels
Grocott, PASd
Blastomycosis Inhalation of conidia (North and South American) Abscess/granuloma Grocott, PASd
Paracoccidio-mycosis Spread from lungs (South American blastomycosis) Abscess/granuloma Steering wheel-like yeasts
Candidiasis Opportunistic Abscess/granuloma Grocott, PASd
Coccidioido-mycosis Inhalation of dust with arthrospore (America) Abscess/granuloma Grocott, PASd
Cryptococcosis Opportunistic Sometimes epithelioid granuloma Grocott, PASd, mucicarmine
Histoplasmosis Inhalation. Opportunistic (Worldwide/Africa depending on organism) Kupffer cells, granuloma, histoplasmoma, abscess Grocott
Mucormycosis (zygomycosis) Opportunistic Abscess, vascular invasion Grocott, PASd
Viral
Cytomegalovirus infection Immunocompromised Mononucleosis like, epithelioid granuloma In immunocompetent: No inclusions. immunohistochemistry−ve, serum
Epstein–Barr virus—infectious mononucleosis Adolescence Sinusoidal lymphocytosis, atypical. Epithelioid granuloma Immunohistochemistry/ in situ hybridization. Serum
Hepatitis A See Chapter 6 Fibrin ring granuloma See Chapter 6
Hepatitis B See Chapter 6 ∼1.5% of HBV shows granuloma See Chapter 6
Hepatitis C See Chapter 6 ∼1.3% of HCV shows granuloma. Relationship with IFN questionable See Chapter 6
Herpes zoster Chickenpox/shingles (reactivation). Liver involvement in immunocompromised host Similar to herpes
Parasitic
Amoebiasis Faecal–oral Abscess PAS
Immunohistochemistry
Ancylostomiasis Tropics. Larva from faeces penetrate skin, spread to lung and to GI tract Ova/parasite in stools
Capillariasis Food contaminated by eggs Granuloma, eosinophils, typical double shelled eggs
Enterobiasis Temperate zones Granuloma around degenerate worm
Fascioliasis Sheep, goat, cattle. Worm in bile duct (Europe) Cholangitis. Granulomas around eggs
Giardiasis Faecal–oral. GI symptoms Granuloma/abscess Stools, serology, duodenal biopsy
Paragonimiasis Raw crustaceans, upper GI, liver and lung (Southeast Asia, South America, Africa) Granuloma around eggs/worms Demonstration of ova/parasites in fluid/tissue. Serology
Opisthorchiasis Freshwater fish. Worm in biliary tract (Far East) Cholangitis, abscess
Pentastomiasis Larva stage (nymphs) of Linguatula serrata (Africa, Asia) Calcification, granuloma
Schistosomiasis Tropical worm in portal/mesenteric veins. Eggs to liver Granuloma, eosinophils, eggs, haemozoin, clay-pipe stem fibrosis Ova in urine, stools. Serology
Strongyloidiasis Persistent, unmasked by immunosuppression.
(tropical–subtropical)
Associates with Gram-negative sepsis. Small portal veins, giant cells, eosinophils
Toxocariasis Faecal–oral. Dogs Eosinophilic necrosis lined by eosinophil-rich granulomas (tracks of larvae)
Visceral leishmaniasis (kala-azar) Bite of sand fly (Southern Europe, Middle east, Asia, Africa) Plasma cells, sinusoidal lymphocytosis, granuloma Parasites in portal macrophages and Kupffer cells
This table is based on several excellent reviews , , , ; additional references are given as appropriate. AFB , Acid-fast bacilli; BCG , bacille Calmette-Guerin; GI , gastrointestinal; HBV , hepatitis B virus; HCV , hepatitis C virus; IFN , interferon; NOS , not otherwise specified; PAS , periodic acid-Schiff; PASd , periodic acid-Schiff-diastase; PCR , polymerase chain reaction.

Table 15.3
Hepatic granulomas—reported noninfectious causes and major features
Hypersensitivity
Drugs (see Chapter 12 ) Long list of drugs may be associated with cholestatic or hepatitic pattern. Eosinophils are variably present
Metals (see Chapter 12 )
Beryllium Industrial exposure (metallurgy, beryllium melting)
Copper Occupational (copper sulphate in vineyard spray), domestic
Gold Treatment of rheumatoid arthritis
Immunological diseases
Common variable immunodeficiency , Defect of B-cell differentiation, hypogammaglobulinaemia, second/third decade, autoimmune disease, lymphoid tumours
Chronic granulomatous disease of childhood <1 year of age, lymphadenopathy, hepatosplenomegaly. Bacterial, fungal. Lipofuscin deposition
Polymyalgia rheumatica Proximal muscles, over 50 years, associated with giant cell arteritis. May coexist with PBC
Primary biliary cholangitis (PBC) See Chapter 9
Primary sclerosing cholangitis See Chapter 9
Rheumatic fever Heart, joint, skin, brain involvement predominates; in children usually
Systemic lupus erythematosus
Vascular diseases
Allergic granulomatosis (Churg–Strauss syndrome) Angitis with allergic rhinitis, asthma and peripheral eosinophilia. Heart disease. Hepatic infarction
Necrotizing angiitis in drug abuse See reference
Polyarteritis nodosa Nodular regenerative hyperplasia. Bile duct damage
Giant cell arteritis Large arteries, temporal, vertebral, ophthalmic
Wegener granulomatosis Necrotizing granuloma of respiratory tract, necrotizing vasculitis, renal disease
Foreign materials
Anthracotic pigments See Lung disease
Barium
Cement and mica dust See reference
Mineral oil—radiocontrast media, food additives Portal tracts, perivenular, nonfatty liver, spleen and lymph node involvement
Silica Birefringent silica particles in liver of patients with anthracosilicosis, sand blasters, dental technicians
Silicone rubber—renal dialysis tubing See reference
Starch
Suture material
Talc Drug abusers, portal and centrilobular talc-laden macrophages
Thorotrast Contrast medium (abandoned in 1955); dark brown refractile pigment (phase contrast). See Chapter 12
Neoplasms
Extrahepatic malignancy See reference
Hepatocellular adenoma and liver adenomatosis See reference
Hodgkin disease (HD) Epithelioid granuloma in ∼10% of patients, usually portal. Does not mean liver involvement by HD
Non-Hodgkin lymphoma Same as HD
Miscellaneous
Biliary tract obstruction—bile granulomas See Chapter 9
Chronic inflammatory bowel disease
Eosinophilic enteritis
Jejuno-ileal bypass surgery See Chapter 5
Porphyria cutanea tarda Lobular aggregates of iron, ceroid-laden Kupffer cells and fat globules
Sarcoidosis
Lipiodolized neocarzinostatin Hepatocellular carcinoma
This table is based on several excellent reviews , , , , , ; additional references are given as appropriate.

Liver function tests are usually cholestatic (raised alkaline phosphatase). In practical terms, the cause may be evident from the histopathology, or known but not evident on biopsy, or suspected only, or remain unknown after full clinicopathological evaluation, including appropriate skin or interferon-γ release assay, microbiological, serological, molecular and biochemical screening. Long-term follow-up is important and reveals a cause in an additional minority, , , but failure to establish a cause in 25% or more patients is well documented. In broad terms, most established causes can be grouped within the categories of infection, immune disorder, drug-related or reaction to neoplasia. In an early study of over 6000 biopsies, 74% were associated with generalized granulomatous disease, 4% with primary hepatic disease, and the remaining 22% were indeterminate for cause. Gaya and colleagues found 63 of 1662 consecutive liver biopsies to show hepatic granulomas; the commonest underlying diagnoses were primary biliary cholangitis (PBC) (23.8%), sarcoidosis (11.1%), drugs (9.5%), HCV (9.5%), autoimmune overlap syndrome (6.3%) and Hodgkin disease (6.3%). Drebber et al. had similar findings, with 48% of 442 biopsies of granulomatous hepatitis being related to PBC, 8% to sarcoidosis and 2% to drugs. Polymerase chain reaction (PCR) identified pathogens in 15 biopsy samples ( Bartonella henselae , Listeria , Mycobacterium tuberculosis , Yersinia pseudotuberculosis , CMV and Epstein–Barr virus). PBC and sarcoid were likewise the two commonest causes in the 35 (1.3%) from 2662 liver biopsies with granulomas in a recent Turkish study.

On microscopy, granulomatous duct injury characterizes PBC but can occur with drug injury or sarcoidosis; periductal bile granulomas may occur in large-duct obstruction, often with acute cholangitis. Ill-defined hyalinized nodules in portal tracts may mark the site of past granulomas, for example, in sarcoidosis. Particulate material such as schistosome ova may be seen on routine stains or with phase-contrast or polarizing microscopy; serial sectioning might be needed to show that the lesion is primarily vascular. Eosinophil-rich necrotizing granulomas raise the possibility of toxocariasis (visceral larva migrans). Special histochemical or immunohistochemical stains are helpful when particular infectious agents are suspected (see Chapter 9 ). In TB, a nonportal distribution of granulomas is said to be characteristic, but on needle biopsy caseation is uncommon and acid-fast bacilli demonstrable in <10% of proven cases (although more often in autopsy material). Instead, real-time PCR analysis is a more sensitive means of diagnosis in paraffin-embedded tissue. In miliary TB, well-formed granulomas are infrequent and the characteristic features are Kupffer cell hyperplasia with ill-formed macrophage ‘microgranulomas’.

Patients may be regarded as having idiopathic granulomatous hepatitis only after exhaustive investigation does not reach a specific diagnosis. , The term is inaccurate in that there is seldom significant hepatocellular damage, and it has been viewed as a form of sarcoidosis confined to the liver. Some of these patients have a prolonged or recurrent pyrexial illness with weight loss, myalgia, arthralgia and vague abdominal pain. They fail to benefit from a trial of antituberculous drugs but may respond to immunosuppression ; in some, the condition resolves spontaneously. In other patients, the granulomas are an incidental finding and appear clinically and biochemically without consequence, and follow-up may be sufficient.

Epithelioid granulomas and microgranulomas were found after liver transplantation in 42 of 563 patients (7.5%), predominantly in the early months ; most cases (71%) had a possible cause, including reaction to hepatocyte necrosis, steatosis and, in portal tracts, acute rejection or, later on, recurrent PBC. Subsequent studies addressed the prevalence of allograft granulomas in patients transplanted for HCV-related cirrhosis. Fiel et al. reviewed allograft liver biopsies from 820 patients transplanted for HCV; they found noncaseating epithelioid granulomas in 25 biopsies (0.24%), more often in patients receiving pegylated interferon therapy. An additional survey identified granulomas in liver allograft of 4 (8%) of 53 patients transplanted for HCV-related cirrhosis but found no prognostic significance. A study by Collins et al. of 23 cases of hepatic granulomas in children showed that the yield of specific diagnoses is increased when molecular approaches are included. They identified an aetiology in 87%; histoplasma was incriminated in 65% of their cases by PCR.

There is one report of familial granulomatous hepatitis in which two parents and three of their seven children were affected. There are also occasional reports of hepatocellular carcinoma developing in patients with chronic granulomatous hepatitis. ,

Sarcoidosis and the liver

Sarcoidosis is a common cause of noninfectious hepatic granulomas. Most patients are between 25 and 45 years, with a second peak among women over 50 years in Europe and Japan. , The cause is unknown, but thought to involve a genetic predisposition for exaggerated granulomatous response to pathogen-associated molecular patterns, including persistent products from killed mycobacteria or propionibacteria. Indeed, there are multiple reports of interferon-induced pulmonary and cutaneous sarcoidosis during treatment of viral hepatitis C. The development of hepatic and other organ sarcoid-like granulomatosis is also reported during therapeutic tumour necrosis factor-α (TNF-α) blockade for various chronic inflammatory diseases, , potentially reflecting interference with T h1 responses.

The liver follows lymph nodes and lung in frequency of involvement. Hepatic sarcoidosis is often asymptomatic, but about 20–30% patients have cholestatic liver function tests. In one study of 837 sarcoidosis patients, 204 (24%) had abnormal liver biochemistry, among which 127 (15%) were attributed to hepatic involvement, correlating in degree with granulomatous extent and fibrosis in the minority biopsied. In some patients a diagnosis of sarcoidosis has been established on liver biopsy with no radiological evidence of pulmonary involvement. Imaging modalities such as whole body mapping with positron emission tomography (PET)/computed tomography (CT) of uptake of the glucose analogue 2-[(18)F]-fluoro-2-deoxy- d -glucose (FDG) help to evaluate the extent and distribution of inflammatory foci in sarcoidosis between different organs, including liver involvement. Serum angiotensin-converting enzyme levels are not accurate for the diagnosis of sarcoidosis.

The histopathology of hepatic sarcoidosis was reviewed by Ishak. Sarcoid granulomas are more frequent in portal tracts or in the periportal area. They consist of a compact aggregate of large epithelioid macrophages, sometimes with multinucleated giant cells, and with a surrounding rim of CD4- and CD8-positive T-lymphocytes and macrophages; occasional eosinophils may be present ( Fig. 15.6A ). Schaumann and asteroid bodies are infrequent. Central granular eosinophilic fibrinoid necrosis may occur (liver biopsy has been rarely reported in necrotizing sarcoid ) but caseation is never found. Reticulin fibres are abundant within the granulomas, particularly in older lesions, when a surrounding cuff of fibrous tissue becomes prominent. Giant cells may persist for some time in the fibrous scars and dense amyloid-like scars may replace the granulomas. Confluent granulomas may produce extensive irregular scarring. A nonspecific reactive hepatitis often accompanies the granulomas, and lobular hepatitis may be prominent during active clinical disease. There may be focal damage to bile ducts ( Fig. 15.6B ), which can resemble that of PBC or primary sclerosing cholangitis (PSC) (see later). , ,

Figure 15.6, Liver in sarcoidosis. (A) Typical parenchymal granuloma with a surrounding cuff of lymphocytes. (H&E.) (B) Portal tract granuloma causing damage to the bile duct. (H&E.) (C) Diffuse hepatic involvement in sarcoidosis with a confluent aggregate of giant cell granulomas. (H&E.) (D) Sarcoid granulomas are typically sclerotic. (Reticulin stain.)

Hepatic sarcoidosis infrequently progresses to clinical chronic liver disease with hepatomegaly, portal hypertension, ascites and hepatic encephalopathy , , ; sometimes, sarcoidosis may only be recognized as the cause of liver failure on examination of the explanted liver after transplantation. The fibrosis can be directly related to granulomas, but extensive portal and parenchymal fibrosis unrelated to granulomas is also described to contribute to cirrhosis ( Fig. 15.6C, D) . Signs of portal hypertension often develop in the absence of cirrhosis. , , , Valla et al. described 32 patients with sarcoidosis in whom portal hypertension was the predominant clinical feature. The portal hypertension was presinusoidal due to pressure effect by portal tract granulomas, sometimes with concurrent sinusoidal block due to fibrosis. Nodular regenerative hyperplasia due to portal venular attrition is evident in some cases. , Obstruction of hepatic vein branches by sarcoid granulomas is a rare cause of Budd–Chiari syndrome, with recurrence after liver transplantation documented in one case.

Rarely, hepatic sarcoidosis manifests with a progressive ductopenic cholestatic syndrome leading to biliary cirrhosis, and so it merits consideration in the differential diagnosis of chronic cholestatic disease. Rudzki and colleagues reviewed 21 such cases and reported five of their own; clinically and biochemically these patients expressed many of the features of PBC but antimitochondrial antibody was not found and their five additional cases were males. Murphy et al. emphasized the progressive bile duct loss that was a feature in five patients. The similarities between sarcoidosis and PBC have been reviewed in a number of studies. , , In the ‘overlap’ patients, the principal initial manifestations were pulmonary symptoms. In general, antimitochondrial antibody is not present in sarcoidosis. There are rare cases in which it remains speculative that both disorders coexist. , Cutaneous and pulmonary manifestations of sarcoidosis have also occurred months after liver transplantation for PBC.

Occasional cases of cholestasis in sarcoidosis are the result of a mass effect of sarcoid nodules at the hilum of the liver with bile duct obstruction. , In one patient hepatobiliary sarcoidosis mimicked a Klatskin tumour. Hepatic sarcoidosis does not appear to respond well to therapeutic intervention. Although frequently used, there is no evidence that corticosteroids prevent long-term hepatic disease progression in asymptomatic patients. A variety of steroid-sparing immunosuppressive agents including methotrexate have been used in cases with advanced liver disease. Sarcoidosis is a rare indication for liver transplant (0.12% in one analysis) ; the disease can recur in the allograft and is usually mild, but in about 3% patients causes graft failure. ,

Nutritional liver disease

Malnutrition can contribute to or cause some liver diseases. Conversely, disturbances of nutrition that occur in liver disease can be significant factors in the accompanying clinical presentation. ,

The liver in protein-energy malnutrition

Protein-energy malnutrition includes the various disease states arising from inadequate intake of protein and/or calories. The extreme manifestations are kwashiorkor (protein malnutrition with adequate calories) or marasmus (the childhood analogue of starvation in adults); marasmic kwashiorkor describes states with features of both (wasting and pitting oedema). In established kwashiorkor, asymptomatic massive hepatomegaly due to steatosis is almost invariable. The steatosis begins as small droplets in periportal hepatocytes, with subsequent large droplet macrosteatosis that becomes panlobular. Mallory–Denk body-like material can be seen, but there is little hepatocyte necrosis or inflammation and true steatohepatitis does not develop. Biochemical tests of liver function can show mildly raised transaminases and low albumin ; occasional cases of severe cholestasis have been described. The steatosis of kwashiorkor per se does not lead to cirrhosis , ; fibrosis and/or necroinflammation in patients with kwashiorkor is probably due to different disease such as chronic hepatitis B virus (HBV) infection, malaria or TB. , After refeeding, lipid first clears within days from the perivenular hepatocytes then resolves fully.

The pathogenesis of steatosis in kwashiorkor probably includes several factors, such as increased mobilization of fat for carbohydrate synthesis, altered hepatocyte β-oxidation and deficiency of apolipoproteins mediating lipid transport from liver. , In some areas, hepatotoxicity from aflatoxin ingestion might contribute.

In marasmus , the hepatocytes tend to be atrophic and the sinusoids are dilated but there is no parenchymal inflammation or fibrosis. Fatty liver is not a feature of marasmus , ; steatosis, if present, is mild and focal with no particular zonal distribution. Peliosis hepatis has been reported. Marasmic kwashiorkor in developed countries can result from child abuse by neglect.

Anorexia nervosa is a common psychiatric eating disorder characterized by behaviour that maintains excessively low weight, including excessive dieting and/or purging. Increases of liver transaminases are common (over 40% patients ), correlating negatively with body mass index, and the severest cases can present with hypoglycaemia and acute liver failure, including ascites. , Liver biopsy is uncommonly required, but biopsies in 12 consecutive cases of acute liver failure attributable to anorexia nervosa, taken 1–9 days after admission, showed consistent glycogen depletion and more variable perivenular hepatocyte atrophy with mild perisinusoidal fibrosis ( Fig. 15.7 ). Despite marked alanine transaminase (ALT) elevation there was no significant lobular inflammation, necrosis or, in many, increased apoptosis despite more than 50-fold increases in transaminase values. Instead, organelle depletion and increased autophagosomes on electron microscopy suggested starvation-induced autophagy to be the primary mechanism of liver damage. , Scattered single cell hepatocyte death is also occasionally evident and probably contributes. A case report with a later liver biopsy, after 18 days of treatment, described the accumulation of glycogen in the recovering liver. Mild increases in stainable iron have occasionally been observed and speculatively linked to unused iron due to reduced haemoglobin synthesis. , , Liver enzyme abnormalities normally resolve during refeeding unless complicated by a refeeding syndrome, in which rising metabolic capacity depletes mineral and cofactor micronutrients, causing electrolyte and fluid shifts. ,

Figure 15.7, Anorexia nervosa. (A) Perivenular glycogen depletion and plate atrophy in severe disease (periodic acid-Schiff [PAS] stain), with (B) perivenular perisinusoidal fibrosis in the same area on a serial section (picrosirius red stain).

Bariatric surgery

Bariatric surgery to treat severe obesity includes restrictive and malabsorptive procedures, some of which may be combined. Roux-en Y gastric bypass is a common restrictive and malabsorptive procedure. Nutritional complications include protein malnutrition (‘secondary kwashiorkor’) and a variety of micronutrient deficiencies. A study of the effects of bariatric surgery on liver injury with biopsy at 1 and 5 years showed reduced steatosis and ballooning that correlated with improved insulin resistance, and a slight increase in postoperative hepatic fibrosis at 5 years, although early stage in 95% of patients. Liver failure has occurred in obese patients after jejuno-ileal bypass, a procedure that is no longer performed.

Intestinal failure-associated liver disease

Intestinal failure is diagnosed when absorption of macronutrients and/or water and electrolytes cannot maintain health or growth without intravenous supplementation. Intestinal failure-associated liver disease (IFALD) has replaced parenteral nutrition (PN)-associated liver disease as the preferred terminology, reflecting the current understanding that multiple factors can contribute to liver injury in this setting. These factors include mucosal barrier disruption with dysbiosis and inflammation, parenteral nutrient composition (lipid emulsions and non-lipid elements), with important roles for sepsis, malnutrition, antibiotic usage, transfusions and recent surgery. In practice, it is therefore difficult to apportion the contribution that PN might make to liver dysfunction in individual patients. Those dependent on parenteral nutrition (PN) due to intestinal failure, especially infants, are susceptible to cholestatic liver disease. Patients with one of the many conditions causing short bowel syndrome are at particular risk. Cholestatic liver biochemistry is a hallmark of IFALD, but in older children and adults steatosis is often also present. , Biliary sludging, cholelithiasis and acalculous cholecystitis can also complicate intestinal failure and are predisposed to by ileal resection or ileal Crohn disease. Ductopenia was identified by Naini and colleagues in 25% of both infants and adults in a large review of 53 infants and 36 older children and adults dependent on PN. The authors observed that the combination in some patients of biliary periportal fibrosis with perivenular fibrosis was usefully characteristic of PN-associated liver injury in adults and infants, compared with other causes of biliary fibrosis. Liver fibrosis of some degree develops in the great majority of infants and older patients during prolonged PN (longer than 6 weeks), including periportal and perivenular fibrosis. , Risk factors include duration of PN and short bowel length , ; progression to severe fibrosis and cirrhosis may be more likely in infants, in particular those with short bowel syndrome. Patients with IFALD-associated liver disease can be evaluated for intestinal or combined liver–intestinal transplantation, for which staging of hepatic fibrosis may be required. Regression of IFALD-associated native liver fibrosis can occur in those for whom isolated intestinal transplantation has allowed weaning from PN. ,

Intestinal failure-associated hepatic disease in young infants

IFALD-associated cholestasis in neonates is more likely with longer treatment, with younger gestational age and low birth weight. The differentials include ‘physiological’ cholestasis, sepsis and the numerous other causes of neonatal cholestasis (see Chapter 3 ). Morphologically, the individual changes are not specific and extremely variable. , Bilirubinostasis is consistently present and may develop in a matter of days; cholestatic rosettes are often present; bile plugs may also be present in interlobular bile ducts. Steatosis is infrequent. Hepatocellular ballooning is more often severe than in older children or adults. The portal tracts show a variable mixed inflammatory cell infiltrate and, with prolonged therapy, a periportal ductular reaction; progressive fibrosis can produce biliary cirrhosis ( Fig. 15.8 ). During prolonged treatment, serial liver biopsy may be needed to assess fibrosis, which is common but not usefully predicted or monitored by serum liver function tests , , ; fibrous progression varies widely between individuals. ,

Figure 15.8, Paediatric intestinal failure-associated liver disease. Severe chronic liver injury with cholestasis, ductular reaction and bridging fibrosis in a 10-year-old child. (H&E stain.)

The pathogenesis of IFALD-associated cholestasis is uncertain. Suggestions have included altered bile composition with (in neonates) immature hepatic bile acid metabolism and transport, the lack of enteral nutrition with disturbed enterohepatic circulation of bile acids, suppression of trophic or secretion-stimulating hormones and the composition of the infusate (including nutritional deficiencies or direct epithelial toxicity). ,

Intestinal failure-associated hepatic disease in older children and adults

Hepatic abnormalities are common in older children and adults with IFALD, in particular bilirubinostasis and periportal steatosis, but the incidence is difficult to establish and, in most cases, biochemical evidence of dysfunction is transient. High-calorie dextrose-based PN has been superseded by lower calorie infusions containing lipid emulsions, with a reduced incidence of hepatic dysfunction. Increases in serum bilirubin are milder and less common than in infants, but increased serum transaminases, alkaline phosphatase and γ-glutamyltranspeptidase affect 20–60% of patients, may develop after 5–20 days of treatment and persist in 15–25% of patients who receive long-term PN. , Perivenular bilirubinostasis develops after 2–3 weeks on PN and may be accompanied by periportal inflammation and portal fibrosis that persists after stopping PN ( Fig. 15.9 ). , , Chronic cholestasis in IFALD-associated liver disease is associated with significant secondary copper overload. The steatosis may reflect unbalanced lipid turnover; carnitine and choline deficiency have each been postulated. , Buchman and colleagues emphasized the pathologic differences between IFALD-associated steatosis and that of nonalcoholic fatty liver disease (NAFLD), with the former distinguished by its periportal distribution, often admixed microvesicular steatosis, rarity of steatohepatitis, together with the concurrence of bilirubinostasis and portal biliary features (oedema, ductular reaction, ductopenia), and a ‘jigsaw’ pattern of fibrosis versus perisinusoidal fibrosis. If conservative management of IFALD-related liver disease fails, isolated intestinal or intestine/liver transplantation may be indicated.

Figure 15.9, Adult intestinal failure-associated liver disease. Periportal fibrosis and cholate stasis in an adult on parenteral nutrition for short bowel syndrome.

The liver in gastrointestinal diseases

Hepatic involvement in diseases of the gastrointestinal tract is common, the portal vein affording direct access for toxins, microorganisms and tumour emboli to cause nonspecific reactive hepatitis, intrahepatic sepsis and intrahepatic metastases, respectively. There is also increasing understanding of the influence on different liver diseases of the intestinal microbiota and intestinal mucosal barrier health. There are specific hepatobiliary disorders associated with chronic inflammatory bowel disease (IBD); other miscellaneous associations are outlined later.

Chronic inflammatory bowel disease

IBD is thought to represent an unbalanced mucosal immune response to gut contents in genetically predisposed individuals, central to which are altered microbiota and failing mucosal firewalls which permit pathobiont penetration into mucosa. Hepatobiliary disease in IBD is among the commonest of extraintestinal manifestations. With some exceptions, the spectrum of such disease is similar between Crohn disease and ulcerative colitis (UC) (see also Chapter 9 ). Colonic Crohn disease is more often associated with hepatic dysfunction than noncolonic disease and is usually coincident with other extraintestinal systemic complications ; this distinction accords with genetic association data that IBD is best categorized into three equally distinct genotype–phenotype groups, taking account of disease location: ileal Crohn disease, colonic Crohn disease and UC.

Abnormalities of liver function tests affect approximately 50% of patients with chronic IBD. , The frequency of biopsy-proven significant liver disease is considered to be lower, from 5% to 17% in UC and 10% to 30% in Crohn disease. If the liver function tests are normal, Broomé et al. found that <3% develop biopsy-proven liver disease on follow-up. PSC, drug hepatotoxicity, steatosis and cholelithiasis are common morbidities, but other important associations are summarized here and in Table 15.4 . PBC is rare but appears to be more prevalent in IBD, particularly UC, than in the general population and has a much reduced female preponderance. , IBD may also occur de novo after liver transplantation; risk factors include CMV infection and immunosuppression with tacrolimus.

Table 15.4
Liver involvement in chronic inflammatory bowel disease
Approximate frequency
Steatosis 25%
Primary sclerosing cholangitis 1–5% (especially ulcerative colitis)
Chronic hepatitis 1–2%
Cirrhosis 2–5%
Bile duct carcinoma 150-fold in PSC (13%)
Gallstones 5–10-fold (ileal Crohn disease)
Epithelioid granulomas 5%
Amyloidosis 0.5% (Crohn disease)
Pylephlebitis and abscess Rare (mainly Crohn disease)
PSC , Primary sclerosing cholangitis.

Primary sclerosing cholangitis

The majority (∼80%) of patients with PSC also have IBD at some point in their life, often preceding diagnosis of PSC. The development of PSC in IBD is thought to reflect common susceptibilities to the development of unbalanced mucosal immune responses to environmental stimuli. PSC is more prevalent in patients with UC (1–5%) than with Crohn disease (1–3%) (see Chapter 9 ) but the colitis associated with PSC often has a relatively distinct phenotype (pancolonic or predominantly right sided, but relatively quiescent with increased incidence of colonic dysplasia). PSC patients have an estimated 150-fold increased risk of cholangiocarcinoma, which affected 13% patients in one large follow-up study, was commonly apparent at or soon after diagnosis of PSC, with a subsequent incidence of ∼1.5% per year, the risk being confined to those PSC patients with IBD. , A detectable gallbladder mass in patients with PSC is sufficiently likely to be carcinoma to merit cholecystectomy. Small duct PSC, in which the cholangiogram is normal, is relatively uncommon but requires liver biopsy for diagnosis and is typically associated with IBD; some patients progress to manifest large-duct PSC. Approximately 70% of children with PSC have IBD, usually UC or, less often, colonic Crohn disease, the liver disease preceding, coinciding or following IBD. The term ‘autoimmune sclerosing cholangitis’ (ASC) in children refers to the presence of cholangiopathy (as detected by cholangiography) in patients with serological and histological features of autoimmune liver disease (see Chapter 9 ). IBD is present in approximately 40–50% of patients with ASC, as opposed to ∼20% of children with typical autoimmune hepatitis.

Steatosis

Macrovesicular steatosis is the commonest histological liver abnormality in IBD, affecting 25–40% of patients. While steatosis may be directly related to IBD-associated inflammation and dysbiosis, concurrent metabolic syndrome is also an important contributor. Indeed, steatosis can persist after colectomy. Hepatomegaly is unusual.

Chronic hepatitis

A diagnosis of chronic hepatitis should not be made unless cholangiography is normal: historical reported prevalence rates up to 10% were likely overestimates due to the difficulties in discriminating autoimmune hepatitis from PSC and an inability to account for post-transfusional chronic hepatitis C viral infection. An estimate of 1–2% seems likely for UC, but there is little evidence of an association with Crohn disease.

Cirrhosis

Edwards and Truelove reported that cirrhosis was present in 2.5% of patients with UC and accounted for 10% of deaths over 20 years follow-up. The overall prevalence of cirrhosis is about 2–5%, , , about 12–50-fold more common than controls without chronic IBD. Patients with extensive colonic disease are most at risk. Cirrhosis may develop as a sequel to PSC where it will show a biliary pattern, or less commonly due to chronic hepatitis (viral or auto­­immune). Cirrhotic patients with colectomy may suffer variceal bleeding at the ileostomy stoma or ileo-rectal anastomosis. , Colonic disease may be exacerbated after liver transplantation for PSC.

Cholangiocarcinoma

Approximately 8–20% of patients with cholangiocarcinoma of the proximal bile ducts have UC, and such patients are on average 20 years younger than those without IBD. , The risk of cholangiocarcinoma is increased 10–30-fold in patients with UC, but in Crohn disease the association appears to be uncommon. The carcinoma develops as a complication of PSC, which is considered a premalignant condition from which carcinoma develops in 10–15% of cases. Biliary intraepithelial neoplasia (BilIN) can be seen adjacent to invasive cholangiocarcinoma ( Fig. 15.10 ). Carcinoma usually develops in patients with longstanding (≥15 years), extensive and severe UC, occasionally some years after total colectomy. , Radiological discrimination from duct involvement by PSC is challenging and cholangiocarcinoma may not be diagnosed until liver resection at transplantation or at autopsy. The prognosis is dismal, with a mean survival time of approximately 6–18 months. Liver transplantation is of limited value and carcinoma diagnosed incidentally at the time of operation may recur in the allograft.

Figure 15.10, Biliary intraepithelial neoplasia and invasive cholangiocarcinoma in primary sclerosing cholangitis associated with ulcerative colitis. (H&E.)

Other changes

Patients with Crohn disease have a 2-fold increased incidence of gallstones but this increases to 5–10-fold with disease of the terminal ileum, related to the extent and duration of the ileal disease or ileal resection. This complication is thought to be due to bile acid malabsorption and possibly changes in bile acid composition, causing cholesterol saturation in the bile. The gallbladder itself can be affected by Crohn disease.

Amyloidosis AA affects about 0.5% IBD patients. , It is more common in Crohn disease than UC and merits consideration in Crohn disease patients presenting with hepatomegaly. Regression of amyloidosis has been reported after colectomy.

Hepatic granulomas are found in approximately 5% of IBD patients, predominantly in Crohn disease, , and may resolve soon after colectomy. Granulomas may also be associated with drug hepatotoxicity related to treatment of IBD, including TNF-α antagonists and sulfasalazine or mesalazine. Approximately 3% of patients with granulomatous hepatitis have Crohn disease.

Pylephlebitis and pyogenic abscess are rare complications of IBD, although can be the presenting feature, encountered mainly in Crohn disease. Abscesses are often multiple, in the right lobe, yield a positive culture and there is often bacteraemia. Unusual differentials such as amoebic liver abscess due to unrecognized amoebic colitis merit consideration, particularly if a patient is on immunosuppressive anticolitic therapy. Another differential diagnosis is of so-called aseptic abscesses, an apparently noninfectious condition that predominantly presents in patients with IBD. Typically, there are lesions in multiple organs including usually the spleen and lymph nodes (unlike infective abscess), the liver (40% of patients) and less frequently other organs. Microscopically, there is granulomatous inflammation surrounding abundant central neutrophil infiltration. The condition usually responds to corticosteroids, although may relapse. Inflammatory pseudotumour has also been reported in isolated patients with Crohn disease.

Patients with IBD have an overall threefold increased risk of venous thromboembolism, but this is much greater during disease flares, although an unique cause has not been found. Portal vein thrombosis occurs in both UC and Crohn disease ; it is rare overall but is common after abdominal surgery: thrombosis affecting main or segmental branches of the portal vein affects about 40% of patients undergoing proctocolectomy, usually nonocclusive in the more proximal branches. , There is a reported association with postoperative pouchitis. Mesenteric thrombosis and Budd–Chiari syndrome due to hepatic vein thrombosis are rare complications in IBD. Nodular regenerative hyperplasia was present in 6% of thiopurine-naïve IBD patients.

PN is often used in severe Crohn disease (see earlier discussion and Fig. 15.8 ). Many of the therapeutic agents used in the treatment of IBD also have hepatotoxicity (see Chapter 12 ), including thiopurines (such as azathioprine), aminosalicylates (sulfasalazine or mesalazine), TNF inhibitors (infliximab or adalimumab), methotrexate and corticosteroids. There is a possible association of hepatosplenic T-cell lymphoma in young patients with IBD receiving TNF-α inhibitor or thiopurine therapy. Immunosuppressive treatment of IBD can cause reactivation of chronic viral hepatitis, in particular hepatitis B. , Liver dysfunction may occur as part of multiorgan failure when there is toxic megacolon.

Hepatic angiomyolipoma has been described in a patient with UC.

Miscellaneous bowel diseases with liver involvement

Coeliac disease

Coeliac disease can affect individuals of any age and affect any organ, with extraintestinal symptoms recognized to be more prevalent in the clinical presentation. About 20–40% of coeliac disease patients have abnormal serum transaminases (and less often raised alkaline phosphatase) at diagnosis. Conversely, coeliac disease is a potential cause for raised transaminases of unidentified cause or is an unsuspected comorbidity revealed, for example, during treatment for hepatitis C. The raised transaminases normalize in many patients within a year on a gluten-free diet; in such individuals they are thought to be secondary to the intestinal mucosal injury and altered permeability of coeliac disease, associated with altered microbiota and microbial products draining to liver. , Changes on biopsy include steatosis and nonspecific reactive hepatitis. , However, coeliac disease also confers a 4–10-fold increased risk of specific autoimmune liver disease, before or after coeliac diagnosis, , most often PBC (about 6%), , , followed by autoimmune hepatitis, then PSC. , These conditions are not improved on a gluten-free diet and there is an increased risk of liver fibrosis or cirrhosis (meriting consideration of coeliac disease as a comorbidity in the differential diagnosis of cryptogenic cirrhosis) , and liver failure. Nodular regenerative hyperplasia and biopsy-proven noncirrhotic portal hypertension is also reported. Rarely, hepatic T-cell lymphoma may occur.

Whipple disease

Whipple disease ( Tropheryma whipplei infection) often shows nonintestinal involvement, which may be the only manifestation of the disease. Inappropriate immunosuppressive therapy, for example of undiagnosed T. whipplei arthropathy with TNF inhibitors, can trigger severe localized or disseminated forms of T. whipplei , including sepsis. Infected PAS-positive-diastase-resistant foamy macrophages which characterize the intestinal involvement may also be found in the liver ( Fig. 15.11 ). , The ‘sickle-form’ bacilli seen in these macrophages have been described in Kupffer cells. Noncaseating epithelioid granulomas also occur in the liver in Whipple disease and may precede the onset of intestinal symptoms ; these granulomas, however, do not contain identifiable bacilli. PCR for T. whipplei is useful to demonstrate the presence of organisms. Massive steatosis has been described in one patient.

Figure 15.11, Liver biopsy in a patient with Whipple disease. (A) There is Kupffer cell hyperplasia with some large intrasinusoidal cells. (H&E.) (B) These are strongly PAS-positive. (PAS-diastase.)

Eosinophilic gastroenteritis

Hepatic granulomas with a prominent eosinophil infiltrate have been reported in two patients, while two case reports document mild and intense eosinophil infiltration of portal tracts. , Sclerosing cholangitis has been described in the hypereosinophilic syndrome in which there was intestinal involvement. ,

The liver in pancreatic diseases

Exocrine pancreas

Cystic fibrosis is discussed in Chapter 3 .

Extrahepatic obstruction due to an annular pancreas is rare and a diagnosis of exclusion. Acute pancreatitis is complicated by jaundice, often transient, in 15–75% of patients; this may be due to inflammatory bile duct obstruction or a common cause such as alcohol excess or gallstones. Chronic pancreatitis is complicated by intrapancreatic common bile duct obstruction in 3–10% cases, which may be transient, recurrent or persistent and accompanied or not by jaundice. Transient jaundice usually manifests during acute inflammatory exacerbations; pressure due to a pancreatic pseudocyst can be a contributing factor , and there is an increased risk of secondary biliary cirrhosis. Steatosis, portal tract inflammation, ductular reaction with fibrosis and cirrhosis may be seen in patients with chronic pancreatitis but probably reflect the common underlying causes of alcohol liver injury or cholelithiasis. Type I autoimmune pancreatitis (the pancreatic manifestation of immunoglobulin G4 [IgG4]-related disease ) often presents with obstructive jaundice due to concurrent IgG4-related sclerosing cholangitis that itself can infrequently be associated with lymphoplasmacytic hilar inflammatory pseudotumours (see Chapter 9 ). Segmental or localized portal hypertension occurs in some patients with chronic hepatitis on a background of pancreatitis, most probably as a consequence of splenic vein occlusion or stenosis. Vascular complications such as hepatic artery injury and portal vein thrombosis after pancreatic surgery are relatively uncommon. Anastomotic bilio-digestive stenosis and biloma can be observed as late complications after pancreatic surgery. Pancreatic pseudocysts have been described within the liver. Pancreatic malignancy presents with obstructive jaundice in about 30% of patients.

Acute pancreatitis has been found in a third of patients with panacinar liver cell necrosis ; it may also occur in acute fatty liver of pregnancy and with acute hepatitis A and C.

Endocrine pancreas

The liver has a key role in carbohydrate metabolism. The ‘hepatotropic’ effects of pancreatic hormones were first investigated by Starzl and colleagues and it is now clear that insulin, glucagon and insulin-like growth factors, together with several other hormones and hepatic growth factors, modulate hepatic function in normal circumstances and regulate hepatic regeneration after liver injury. Only in diabetes mellitus, however, is there evidence of significant liver disease in association with Islet-cell dysfunction.

Diabetes mellitus

The role of type II diabetes in the development of NAFLD is discussed in Chapter 5 . Here we consider the liver abnormalities in insulin-dependent type I diabetes. The liver is central to carbohydrate and lipid metabolism. Insulin deficiency shifts hepatic metabolism towards gluconeogenesis, increasing glucose release from the liver. Declining glucokinase levels (insulin-regulated) also become rate limiting and limit hepatic trapping of glucose from sinusoidal blood. Together with reduced peripheral tissue uptake of glucose, these changes cause hyperglycaemia. Conversely, in diabetics on insulin, glycogen can accumulate within the cytoplasm and nuclei of hepatocytes. The cytoplasmic glycogen appears ultrastructurally predominantly as rosetted composites of particles (alpha granules), nuclear glycogen as smaller dispersed particles. Nuclear glycogenation is seen more often in periportal than perivenular hepatocytes and is very nonspecific but considered to correlate closer with diabetes than obesity. , Abnormal hepatic glycogen loading has been attributed to high insulin dosing in hyperglycaemia, which promotes glycogen storage that can accumulate with time or sometimes very acutely: typical clinical scenarios include vigorous treatment of acutely presenting diabetes with high-dose insulin , ; also long-term poor diabetic control involving repeated excessive insulin dosing that is then self-corrected with high quantity glucose intake; or chronic hyperglycaemia with intermittent insulin dosing. Consequent hepatomegaly due to glycogen loading of the liver in type I diabetics on insulin was described by Mauriac in 1930 as part of a childhood syndrome characterized by poor glycaemic control, growth retardation, pubertal delay and cushingoid features. However, such acquired glycogenosis can also affect adults with type I diabetes. , , The characteristic presentation is with hepatomegaly that may be painful and with raised serum transaminases (sometimes dramatically so) in a context of poor diabetic control. The changes resolve with improved glycaemic control and are not thought to lead to chronic liver injury. , , , The liver histology of 14 affected adults and children was reviewed in detail, for which the term ‘glycogenic hepatopathy’ was coined and has subsequently become prevalent to describe the condition ( Fig. 15.12A ). Hepatocytes are enlarged, with rarefied, pale staining cytoplasm replete with glycogen (demonstrable with PAS), frequent nuclear glycogenation, giant mitochondria and prominent cell borders contrasting with the pale cytoplasm. Sinusoids appear compressed. Steatosis was absent or sparse in most cases, and fibrosis mild in two only, one showing mild steatohepatitis with mild fibrosis. Rebiopsy in one patient after adequate glycaemic control and resolution of signs showed normal liver. A recent series of 31 diabetic children (19 with liver biopsy) showed similar histological features of glycogenosis, but more prevalent steatosis, inflammation and fibrosis. Acquired metabolic glycogenosis can occur without type 1 diabetes, in the setting of metabolic syndrome with reduced glucose tolerance or sugar-rich diet such as from sugar-rich drinks. Adipokines related to metaflammation impair skeletal muscle uptake of glucose, normally the route for most insulin-stimulated glucose disposal. High endogenous insulin levels in such a setting of peripheral insulin resistance would favour hepatic glycogenosis. The glycogenosis may be focal but in some cases is diffuse and striking ( Fig. 15.12B–D ). A recent study documented frequent occurrence in biopsies with NAFLD, both from children and adults, associated with milder degrees of fibrosis and steatosis.

Figure 15.12, Acquired metabolic glycogenosis. (A) Glycogenosis in type I diabetes mellitus. Hepatocytes are enlarged due to glycogenosis; the appearances resemble those seen in inherited glycogen storage diseases. (H&E.) (B) Glycogenosis in a nondiabetic adult with abnormal lipid profile, obesity and echogenic liver on ultrasound. (H&E.) (C) Same case as (B), showing hepatocyte distended with cytoplasmic glycogen that on higher magnification (D) resolves into abundant alpha granule rosette-like aggregates and individual small spherical beta granules. (Electron microscopy: C, × 5160; D, × 104,000.)

The altered lipid metabolism in type 1 diabetes includes the mobilization of free fatty acids from adipose to be converted to triglyceride and very-low-density lipoprotein (VLDL) in liver, where they accumulate. Decreased lipoprotein lipase (insulin-regulated) perpetuates this hyperlipidaemic accumulation of triglyceride and VLDL. Hepatocellular fatty acid oxidation can generate acetyl coenzyme A (CoA) sufficient to saturate the oxidative enzymes and cause ketone formation. However, type 1 diabetes is not itself associated with significant hepatic steatosis, unlike type II diabetes, confirmed with magnetic resonance imaging in a large comparative study and in a biopsy-based survey of 155 children with steatosis on liver biopsy.

Collagenization of the space of Disse, together with deposition of basement membrane components, has been reported in diabetes mellitus , (latterly termed ‘diabetic hepatosclerosis’ when present without steatosis) and thought to represent liver involvement as part of diabetic microangiopathy. , An association with cholestasis has also been suggested. However, no specific association with perisinusoidal fibrosis was found in a cross-sectional comparative study of liver biopsies from 89 diabetics (half insulin-dependent) and matched controls. Instead, the authors identified hepatic arteriolosclerosis as significantly increased in hypertensive diabetics and raised again a possible association with cholestatic liver biochemistry and biliary abnormalities.

Iron-induced beta cell injury was initially proposed as the cause of diabetes in haemochromatosis, although insulin resistance appears also to be involved. The reciprocal influence of iron and insulin and the relationship between steatohepatitis, diabetes and iron overload are complex and discussed further in Chapter 4 . There is an association between type I diabetes and autoimmune hepatitis. ,

Diabetics are at increased risk of liver neoplasms. Subjects with an inherited monoallelic mutation of the transcription factor 1 (TCF1) gene encoding for the hepatocyte nuclear factor 1α (HNF1α) may develop maturity-onset diabetes of the young type 3 and liver adenomatosis when the second allele is inactivated in hepatocytes (see Chapter 13 ). , There are case reports of nodular regenerative hyperplasia, of xanthomatous neuropathy affecting unmyelinated nerve fibres in the hilum and in large portal tracts, of intrahepatic and perihepatic abscess and of PSC occurring in patients with diabetes. Liver injury attributable to drugs used in the treatment of diabetes is described in Chapter 12 .

The liver in endocrine diseases

Polycystic ovary syndrome is the commonest endocrine abnormality in premenopausal women and is associated with insulin resistance and metabolic syndrome, including fatty liver disease (see Chapter 5 ). The hepatotoxic effects of lipid-lowering drugs, oral hypoglycaemic and other drugs used in the therapy of endocrine and metabolic syndrome-related abnormalities are dealt with in Chapter 12 ; this includes the effects of gonadal steroids on the liver, which occur principally in relation to their therapeutic administration.

Thyroid

The liver is the principal site for conversion of tetraiodothyronine (T4) to triiodothyronine (T3). Liver is also the main source of the major thyroid hormone-binding proteins and for conjugation and biliary excretion of the thyroid hormones. As a result, liver disease can be associated with changes in thyroid hormone metabolism and common thyroid function test values, for example, increased thyroid-binding globulin in acute hepatitis. Conversely, thyroid hormones affect hepatocyte metabolism, including synthetic activity, bilirubin and bile acid metabolism, and thyroid diseases can alter liver function.

There is a well-recognized association between autoimmune thyroid disease and PBC (see Chapter 9 ); occasional cases of Graves disease with autoimmune hepatitis and PSC are also described. Post-infantile giant cell hepatitis has been documented in a patient with Graves disease. Increased serum aminotransferase levels are common in patients on propylthiouracil for the treatment of hyperthyroidism. Thyroid and liver abnormalities may both be produced by some drugs including carbamazepine, mefloquine and amiodarone. Sorafenib, used to treat hepatocellular carcinoma, can cause hypothyroidism.

Hyperthyroidism

Abnormal liver function tests are reported in 15–75% of patients with hyperthyroidism and usually resolve with treatment. Increased serum alkaline phosphatase is the most common abnormality (including the bone isoenzyme), but there may be elevated aminotransferases and bilirubin. Jaundice attributable purely to hyperthyroidism is uncommon, usually seen during a thyroid storm where canalicular and sometimes hepatocellular cholestasis with associated swelling injury on liver biopsy are described. Other changes are mild and nonspecific. Jaundice in hyperthyroidism can also be precipitated in the presence of heart failure or concurrent liver disease, including associated PBC. In such patients, the role of the hyperthyroidism in causing jaundice may be overlooked.

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