Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 21

INTRODUCTION TO PATHOLOGY

Pathology  Morphologic changes


 study of the structural, biochemical, and functional changes in cells, o structural alterations in cells or tissues that are characteristic of a
tissues, and organs that underlie disease disease and hence diagnostic of an etiologic process.
 study of the structural and functional causes of human disease. o diagnostic pathology
 explain the signs and symptoms manifested by patients while providing a  uses morphology + analysis of protein expression and genetic
rational basis for clinical care and therapy alterations to determine the nature of disease and to follow its
progression.
Pathology o useful in the study of neoplasms; breast cancers that are
- study of changes that caused a disease indistinguishable morphologically may result from different genetic
- explains S/S, clinical care and therapy abnormalities that result in widely different courses, therapeutic
responses, and prognoses.
o Molecular analysis by techniques such as next-generation
sequencing has revealed genetic differences that predict the
General Pathology
behavior of tumors as well as their response to therapies, an
- common/not tissue specific rxn to injury
- *acute inflammation increasing number of which are now chosen on the basis of the
Systemic Pathology presence (or absence) of specific molecular alterations
- organ system specific

 Clinical manifestations
o The end results of genetic, biochemical, and structural changes in
cells and tissues are functional abnormalities that lead to the clinical
manifestations (symptoms and signs) of disease, as well as its
progression (clinical course and outcome).
o Rudolf Virchow
 4 aspects of a disease process that form the core of pathology: EPMC  father of modern pathology.
1. causation (etiology)  cellular basis of disease
2. biochemical and molecular mechanisms (pathogenesis)  Injury to cells and to the extracellular matrix ultimately leads to
3. structural alterations (morphologic changes) and functional tissue and organ injury, which determines the morphologic and
alterations in cells and organs clinical patterns of disease.
4. resulting clinical consequences (clinical manifestations)

 Etiology is the initiating cause of a disease. OVERVIEW OF CELLULAR RESPONSES TO STRESS


o genetic: inherited or acquired mutations, and disease-associated AND NOXIOUS STIMULI
gene variants, or polymorphisms  Normal cell’s function and structure are dictated by
o environmental: infectious, nutritional, chemical, physical o its state of metabolism, differentiation, and specialization
o constraints imposed by neighboring cells
o single gene anomalies: one etiologic agent is the cause of one o availability of metabolic substrates.
disease  Normal cell can handle physiologic demands, maintaining a healthy steady
state called homeostasis.
o multifactorial:
 atherosclerosis and cancer
 Normal cell function requires a balance between physiologic demands and
 arise from the effects of various environmental insults on a
the constraints of cell structure and metabolic capacity; the result is a
genetically susceptible individual
steady state, or homeostasis.
 Pathogenesis  Cells can alter their functional state in response to modest stress to
o refers to the sequence of molecular, biochemical, and cellular maintain the steady state.
events that lead to the development of disease.
 More excessive physiologic stresses, or adverse pathologic stimuli (injury),
o explains how the underlying etiologies produce the morphologic
result in continuum of progressive impairment of cell structure and
and clinical manifestations of the disease.
function.
o central focus of pathology.
1. Adaption
o to truly understand the disorder cystic fibrosis it is essential to know
2. Reversible Injury
the defective gene, gene product, and the biochemical and 3. Irreversible Injury and Cell Death
morphologic events that lead to clinically significant disease in the
lungs, pancreas, and other organs. Adaptation
o genomics, proteomics, metabolomics - better understanding of
 reversible
pathogenesis and identification of biomarkers that predict disease
 occurs when physiologic or pathologic stressors induce a new state that
progression and therapeutic responses.
changes the cell but otherwise preserves its viability in the face of the
exogenous stimuli.
 These changes include:
o Hypertrophy – increased cell mass
o Hyperplasia – increased cell number o most common cause of hypoxia
o Atrophy – decreased cell mass o related to decreased arterial flow or decreased
o Metaplasia – change from one mature cell type to another venous outflow
 atherosclerosis
 If the stress is eliminated, the cell can return to its original state without  thrombus
having suffered any harmful consequences.  thromboembolus
 If the limits of adaptive responses are exceeded or if cells are exposed to  inadequate oxygenation of the blood
damaging insults, deprived of critical nutrients, or compromised by  cardiorespiratory failure
mutations that affect essential cellular functions, a sequence of events  decreased oxygen-carrying capacity of the blood
follows that is termed cell injury  anemia
 carbon monoxide poisoning
Cell injury is reversible up to a point, but if the injurious stimulus is persistent or  severe blood loss.
severe, the cell suffers irreversible injury and ultimately undergoes cell death. o Depending on the severity of the hypoxic state, cells may adapt,
undergo injury, or die.
 Adaptation, reversible injury, and cell death may be stages of progressive o For example, if an artery is narrowed, the tissue supplied by that
impairment following different types of insults. vessel may initially shrink in size (atrophy), whereas more severe or
 For instance, in response to increased hemodynamic loads, the heart sudden hypoxia induces cell injury and cell death.
muscle becomes enlarged, a form of adaptation, which because of
increased metabolic demands is more susceptible to injury. If the blood  Physical Agents
supply to the myocardium is compromised or inadequate, the muscle first o mechanical trauma
suffers reversible injury, manifested by certain cytoplasmic changes. o extremes of temperature (burns and deep cold)
Unless the blood supply is quickly restored, the cells suffer irreversible o sudden changes in atmospheric pressure
injury and die (Fig. 2.2). o radiation
 The removal of damaged, unneeded, and aged cells through cell death is o electric shock
a normal and essential process in embryogenesis, the development of
organs, and the maintenance of homeostasis into adulthood. Conversely,
 Chemical Agents and Drugs
excessive cell death as a result of progressive injury is one of the most
o glucose or salt in hypertonic concentrations may cause cell injury
crucial events in the evolution of disease in any tissue or organ. It results
directly or by deranging electrolyte and fluid balance in cells.
from diverse causes, including ischemia (reduced blood flow), infection,
o oxygen at high concentrations
and toxins.
o Trace amounts of poisons: arsenic, cyanide, or mercury
 There are two principal pathways of cell death, necrosis and apoptosis.
o environmental pollutants, insecticides, and herbicides
 Nutrient deprivation triggers an adaptive cellular response called
autophagy that may also culminate in cell death. A detailed discussion of o industrial and occupational hazards: carbon monoxide and
these and some other, less common pathways of cell death follows later in asbestos
the chapter. Stresses of different types may induce changes in cells and o recreational drugs/social stimuli: alcohol, narcotics
tissues other than typical adaptations, cell injury, and death. Metabolic o therapeutic drugs
derangements and chronic injury may be associated with intracellular
accumulations of a number of substances, including proteins, lipids, and  Infectious Agents
carbohydrates. Calcium may be deposited at sites of cell death, resulting o viruses, tapeworms, rickettsiae, bacteria, fungi, parasites
in pathologic calcification. o can injure the body by direct infection of cells,
 Finally, the normal process of aging is accompanied by characteristic production of toxins, or host inflammatory response
morphologic and functional changes in cells.
 Immunologic Reactions
Causes of Cell Injury o The immune system serves an essential function in defense against
1. Oxygen Deprivation infectious pathogens, but immune reactions may also cause cell
2. Physical Agents injury.
3. Chemical Agents and Drugs o Injurious reactions to endogenous self-antigens are responsible for
4. Infectious Agents autoimmune diseases.
5. Immune reactions o Immune reactions to many external agents, such as viruses and
6. Genetic defects environmental substances
7. Nutritional shortages

 Genetic Abnormalities
o extra chromosome: Down syndrome
Causes of Cell Injury o single base pair substitution
 Oxygen Deprivation o amino acid substitution: sickle cell anemia
o Hypoxia o deficient protein function
 deficiency of oxygen  enzyme defects in inborn errors of metabolism
 reduces aerobic oxidative respiration  dec ATP  accumulation of damaged DNA or misfolded proteins
 most common cause of cell injury o polymorphisms
o Causes of hypoxia:
 reduced blood flow or loss of blood supply
 Nutritional Imbalances
 ischemia o major causes of cell injury
o undernutrition  swelling of cell
 protein-calorie deficiencies.
 marasmus – decrease in total caloric intake o Fatty change occurs in organs that are actively involved in lipid
 kwashiorkor - decrease in total protein intake metabolism (liver)  accumulation of TAG-filled lipid vacuoles.
 Deficiencies of specific vitamins
 vitamin A: night blindness, squamous metaplasia, MORPHOLOGY
immune deficiency  Cellular swelling / hydropic change / vacuolar degeneration.
 vitamin C: scurvy o earliest manifestation of almost all forms of injury to cells
 vitamin D: rickets and osteomalacia o pallor
 vitamin K: bleeding diathesis o increased turgor
 vitamin B12: megaloblastic anemia, neuropathy, and o increased weight of the affected organ.
spinal cord degeneration o small clear vacuoles within the cytoplasm – distended and pinched-
 folate megaloblastic anemia and neural tube defects off segments of the ER.
 niacin: pellagra [diarrhea, dermatitis, and dementia]  cytoplasm: red (eosinophilc) in H&E
 Nutritional shortages: anorexia nervosa, food shortages, poor o loss of RNA (binds the blue hematoxylin dye).
diet. o eosinophilia becomes more pronounced with progression toward
o overnutrition necrosis.
 Obesity  diabetes and cancer  Ultrastructural changes visible by electron microscopy:
o composition of the diet contributes to a number of diseases. o Plasma membrane alterations: blebbing, blunting, loss of microvilli
 fat-rich diets  elevated serum cholesterol  o Mitochondrial changes: swelling, appearance of small amorphous
atherosclerosis cardiovascular disease densities
o Accumulation of “myelin figures” in the cytosol composed of
phospholipids derived from damaged cellular membranes
o Dilation of the ER, with detachment of polysomes
The Progression of Cell Injury and Death
o Nuclear alterations, with disaggregation of granular and fibrillar
 molecular or biochemical level is the first affected
elements
 There is a time lag between the stress and the morphologic changes of
cell injury or death
o early changes – subtle, detected with highly sensitive methods
o Morphologic manifestations of necrosis take more time to develop CELL DEATH
than those of reversible damage.  Necrosis
 Apoptosis
 ischemia of the myocardium  Other Mechanisms of Cell Death
o cell swelling is a reversible morphologic change that may occur in a o Necroptosis
matter of minutes, and is an indicator of ongoing cellular damage o Pyroptosis
that may progress to irreversibility within 1 or 2 hours.
o Unmistakable light microscopic evidence of cell death may not be
o Ferroptosis
seen until 4 to 12 hours after onset of ischemia.  Autophagy

 Within limits, the cell can repair the alterations seen in reversible injury and
if the injurious stimulus abates, may return to normalcy. Persistent or Necrosis – accidental cell death
excessive injury, however, causes cells to pass the rather nebulous “point w/ inflammation
of no return” into irreversible injury and cell death. Different injurious stimuli Apoptosis – regulated cell death
induce death mainly by necrosis and/or apoptosis w/o inflammation

Necrosis
 “accidental” cell death
REVERSIBLE CELL INJURY  severe injury
 functional and structural alterations in early stages or mild forms of injury,  irreversible
which are correctable if the damaging stimulus is removed.  local inflammatory clears dead cells
 2 features:  Severe mitochondrial damage with depletion of ATP
o Early alterations:  rupture of lysosomal and plasma membranes
 generalized swelling of the cell and its organelles  associated with injuries due to ischemia, exposure to toxins, various
 blebbing of the plasma membrane infections, and trauma.
 detachment of ribosomes from the ER
 clumping of nuclear chromatin.

 oxygen deficiency
 dec. mitochondrial oxidative phosphorylation/ mitochondrial Apoptosis
damage by radiation or toxins
 “regulated” cell death
 dec. ATP
 without inflammation or the associated collateral damage.
 failure of the ATP- Na-K plasma membrane pump
 influx of water
Necrosis o loss of cytoplasmic RNA – no RNA to bind the blue hematoxylin dye
 pathologic process that is the consequence of severe injury. o accumulation of denatured cytoplasmic proteins – bind to red dye
o causes of necrosis: eosin
 loss of oxygen supply (ischemia)  glassy homogeneous appearance
 exposure to microbial toxins o loss of glycogen particles
 burns, chemical and physical injury  cytoplasm: vacuolated, moth-eaten appearance
 active proteases leak out of cells(pancreatitis). o enzymic digestion of cell’s organelles
 characterized by  myelin figures
o denaturation of cellular proteins o phospholipid precipitates that replaces dead cell
o leakage of cellular contents through damaged membranes o phagocytosed by other cells or further degraded into fatty acids
o local inflammation o calcification of fatty acid residues results in deposition of calcium-
o enzymatic digestion of the lethally injured cell rich precipitates.
 EM:
o severely damaged membrane o discontinuities in plasma and organelle membranes
o lysosomal enzymes enter the cytoplasm and digest the cell. o marked dilation of mitochondria with the appearance of large
o Cellular contents leak into the extracellular space, where they elicit amorphous densities
inflammation o intracytoplasmic myelin figures
o damage-associated molecular patterns (DAMPs) are released from o amorphous debris
injured cells o aggregates of fluffy material representing denatured protein
 ATP - mitochondria  Nuclear changes due to breakdown of DNA.
 uric acid - DNA o karyolysis
 decreased basophilia of the chromatin
 These molecules are recognized by receptors in macrophages  due to DNA degradation by endonucleases.
and trigger phagocytosis and production of cytokines that o pyknosis
induce inflammation  seen in apoptosis
 Inflammatory cells produce more proteolytic enzymes  nuclear shrinkage and increased basophilia
 chromatin condenses into a dense, shrunken basophilic mass.
Clearance of the necrotic cells: o karyorrhexis
phagocytosis + enzymatic digestion  fragmentation of pyknotic nucleus
 Within 1 or 2 days, the necrotic cell’s nucleus disappears

MORPHOLOGY
Coagulative necrosis
 architecture of dead tissue is preserved for some days
 tissue has a firm texture
 Necrosis-associated leakage of intracellular proteins through damaged
plasma membranes and ultimately into the circulation is the basis for  the injury denatures structural proteins and enzymes and so blocks the
blood tests that detect tissue-specific cellular injury. proteolysis of the dead cells; as a result, intensely eosinophilic cells with
o Cardiac muscle cells – troponin indistinct or reddish nuclei may persist for days or weeks
o bile duct epithelium – alkaline phosphatase  the necrotic cells are broken down by the action of lysosomal enzymes
derived from infiltrating leukocytes, which also remove the debris of the
o hepatocytes – transaminases
dead cells by phagocytosis.
 Ischemia caused by obstruction in a vessel may lead to coagulative
o these biomarkers are used clinically to assess and quantify
necrosis of the supplied tissue in all organs except the brain
tissue damage.
 A localized area of coagulative necrosis is called an infarct.
o Cardiac-specific troponins can be detected in the blood as
Liquefactive necrosis
early as 2 hours after myocardial cell necrosis, well before
 digestion of the dead cells, resulting in transformation of the tissue
histologic evidence of myocardial infarction becomes apparent.
into a viscous liquid.
 It is seen in focal bacterial or, occasionally, fungal infections,
Irreversibility because microbes stimulate the accumulation of leukocytes and the
liberation of enzymes from these cells.
 inability to reverse mitochondrial dysfunction (lack of oxidative
phosphorylation and ATP generation) even after resolution of the original  The necrotic material is frequently creamy yellow because of the
injury presence of leukocytes and is called pus. For unknown reasons,
hypoxic death of cells within the central nervous system often
 profound disturbances in membrane function
manifests as liquefactive necrosis
 injury to lysosomal membranes results in the enzymatic dissolution of the
Gangrenous necrosis
injured cell that is characteristic of necrosis.
 lower leg that has lost its blood supply and has undergone coagulative
necrosis) involving multiple tissue planes
MORPHOLOGY
 When bacterial infection is superimposed, there is more liquefactive
Necrotic cells
necrosis because of the actions of degradative enzymes in the bacteria
 increased eosinophilia in H&E stains
and the attracted leukocytes (wet gangrene)
o surface components are altered
Caseous necrosis  produce “find me” and “eat me” signals for phagocyte
 foci of tuberculous infection  dead cell and its fragments are rapidly devoured, before the
 the friable white appearance contents leak out, and therefore apoptosis does not elicit an
 structureless collection of fragmented or lysed cells and amorphous inflammatory reaction.
granular debris enclosed within a distinctive inflammatory border; this  controlled by the action of a small number of genes
appearance is characteristic of a focus of inflammation known as a  required for normal embyrogenesis.
granuloma

 Causes of Apoptosis
o Apoptosis occurs in two broad contexts:
1. normal physiologic processes
Fat necrosis 2. pathophysiologic mechanism of cell loss in many different
 refers to focal areas of fat destruction, typically resulting from release of diseases.
activated pancreatic lipases into the substance of the pancreas and the
peritoneal cavity.
 This occurs in the calamitous abdominal emergency known as acute  Apoptosis in Physiologic Situations
pancreatitis. In this disorder, pancreatic enzymes leak out of damaged o normal phenomenon
acinar cells and liquefy the membranes of fat cells in the peritoneum, o purpose:
releasing triglyceride esters that are split by pancreatic lipases.  eliminate cells that are no longer needed
 Fatty acids are generated that combine with calcium to produce grossly  maintains a constant number of various cell populations in
visible chalky-white areas (fat saponification), which enable the surgeon tissues
and the pathologist to identify the underlying disorder. o humans turn over: 1 million cells per second - central to this
 On histologic examination, the necrotic areas contain the shadowy process is death of cells by apoptosis and their removal by
outlines of necrotic fat cells, basophilic calcium deposits, and an phagocytes.
inflammatory reaction. o Apoptosis is important in the following physiologic situations:
 removal of supernumerary cells (in excess of the required
number) during development.
Fibrinoid necrosis  Cell death is critical for involution of primordial structures
 vascular damage due to immune reactions and remodeling of maturing tissues.
 fibrinoid(fibrin-like)  Apoptosis is a generic term for this pattern of cell death,
o bright pink and amorphous appearance in H&E stains regardless of the context, while programmed cell death
o Deposits of immune complexes with plasma proteins that refers only to apoptosis during development.
has leaked out of vessels  Involution of hormone-dependent tissues on hormone
 ex. immunologically mediated vasculitis syndromes withdrawal
 endometrial cell breakdown during the menstrual cycle
 ovarian follicular atresia in menopause
CELL INJURY AND NECROSIS  regression of the lactating breast after weaning.
 Exposure of cells to stress or noxious agents causes cell injury that is  Cell turnover in proliferating cell populations to maintain a
reversible to a point but may progress to death of the cells, principally by constant cell number (homeostasis).
necrosis.  immature lymphocytes in the bone marrow and thymus
 Reversible cell injury: Characterized by cellular swelling, fatty change,  B lymphocytes in germinal centers that fail to express
plasma membrane blebbing and loss of microvilli, mitochondrial swelling, useful antigen receptors
dilation of the ER, and eosinophilia (due to decreased cytoplasmic RNA)  epithelial cells in intestinal crypts,
 Necrosis: A pathologic process in which cellular membranes are  Elimination of potentially harmful self-reactive lymphocytes to
destroyed, enzymes and other constituents leak out, and local prevent immune reactions against one’s own tissues
inflammation is induced to clear the damaged cells. Morphologic features  Death of host cells that have served their useful purpose,
are: eosinophilia; nuclear shrinkage, fragmentation, and dissolution;  neutrophils in an acute inflammatory response
breakdown of plasma membrane and organellar membranes; abundant  lymphocytes at the end of an immune response.
myelin figures; and leakage and enzymatic digestion of cellular contents o In all of these situations, cells undergo apoptosis because they are
 Patterns of tissue necrosis: Under different conditions, necrosis in tissues deprived of necessary survival signals, such as growth factors and
may assume specific patterns: coagulative, liquefactive, gangrenous, interactions with the extracellular matrix, or they receive pro-
caseous, fat, and fibrinoid apoptotic signals from other cells or the surrounding environment.

Apoptosis  Apoptosis in Pathologic Conditions


 programmed cell death o Apoptosis eliminates cells that are injured beyond repair without
 cells destined to die activate intrinsic enzymes that degrade the cells’ eliciting a host reaction, thus limiting collateral tissue damage.
genomic DNA and nuclear and cytoplasmic proteins. o Death by apoptosis is responsible for loss of cells in a variety of
 apoptotic bodies pathologic states:
o plasma membrane–bound fragments of apoptotic cells which  DNA damage.
contain portions of the cytoplasm and nucleus  Radiation and cytotoxic anticancer drugs can damage
o plasma membrane remains intact DNA, either directly or via production of free radicals. If
repair mechanisms cannot correct the damage, the cell  The presence of active caspases is therefore a marker for cells
triggers intrinsic mechanisms that induce apoptosis. undergoing apoptosis
 In these situations, apoptosis has a protective effect by
preventing the survival of cells with DNA mutations that  The process of apoptosis may be divided into an
can lead to malignant transformation. o initiation phase – some caspases become catalytically active and
 Accumulation of misfolded proteins. unleash a cascade of other caspases
 Cell death triggered by improperly folded intracellular o execution phase – terminal caspases trigger cellular fragmentation.
proteins and the subsequent endoplasmic reticulum o Regulation of these enzymes depends on a finely tuned balance
(ER) stress response between the abundance and activity of pro-apoptotic and anti-
 due to inherited defects or due to free radical damage) apoptotic proteins.
 This may be the basis of cell loss in a number of  Two distinct pathways converge on caspase activation:
neurodegenerative disorders. 1. mitochondrial pathway
 Apoptosis can be induced during certain infections, particularly 2. death receptor pathway
viral infections, as a result of the virus itself (as in adenovirus o Although these pathways intersect, they are generally induced
and HIV infections) or the host immune response (as in viral under different conditions, involve different initiating molecules, and
hepatitis). serve distinct roles in physiology and disease.
 An important host response to viruses consists of cytotoxic T
lymphocytes (CTLs) specific for viral proteins, which induce  Mitochondrial (Intrinsic) Pathway of Apoptosis
apoptosis of infected cells in an attempt to eliminate reservoirs o responsible for apoptosis in most physiologic and pathologic
of infection. situations.
 During this process, there can be significant tissue damage. o It results from increased permeability of the mitochondrial outer
The same CTL-mediated mechanism is responsible for killing
membrane with consequent release of death-inducing (pro-
of tumor cells, cellular rejection of transplants, and tissue
apoptotic) molecules from the mitochondrial intermembrane space
damage in graft-versus-host disease.
into the cytoplasm
 Apoptosis may also contribute to pathologic atrophy in
o Mitochondria are organelles that contain remarkable proteins such
parenchymal organs after duct obstruction, such as occurs in
as cytochrome c, a doubleedged sword that is essential for
the pancreas, parotid gland, and kidney.
producing the energy (e.g., ATP) that sustains cell viability, but that
when released into the cytoplasm (an indication that the cell is not
healthy) initiates the suicide program of apoptosis.
o The release of pro-apoptotic proteins such as cytochrome c is
MORPHOLOGY
determined by the integrity of the outer mitochondrial membrane,
 Cell shrinkage.
which is tightly controlled by the BCL2 family of proteins.
o Cell size is reduced
o This family is named after BCL2, a gene that is frequently
o cytoplasm is dense and eosinophilic
overexpressed due to chromosomal translocations and other
o organelles are relatively normal but more tightly packed. aberrations in certain B cell lymphomas .
 Chromatin condensation. o There are more than 20 members of the BCL family, which can be
o most characteristic feature of apoptosis. divided into three groups based on their pro-apoptotic or anti-
o chromatin aggregates peripherally, under the nuclear membrane, apoptotic function and the BCL2 homology (BH) domains they
into dense masses of various shapes and sizes possess.
o nucleus itself may break up into two or more fragments.  Anti-apoptotic.
 Formation of cytoplasmic blebs and apoptotic bodies.  BCL2, BCL-XL, and MCL1
o The apoptotic cell first shows extensive surface membrane  possess four BH domains (called BH1-4). T
blebbing, which is followed by fragmentation of the dead cells into  these proteins reside in the outer mitochondrial
membrane bound apoptotic bodies composed of cytoplasm and membrane as well as in the cytosol and ER membranes.
tightly packed organelles, with or without nuclear fragments.  By keeping the mitochondrial outer membrane
 Phagocytosis of apoptotic cells or cell bodies, usually by macrophages. impermeable, they prevent leakage of cytochrome c and
o The apoptotic bodies are rapidly ingested by phagocytes and other death-inducing proteins into the cytosol
degraded by the phagocyte’s lysosomal enzymes.  Pro-apoptotic.
 In H&E-stained tissue, the apoptotic cell appears as a round or oval mass  BAX and BAK
of intensely eosinophilic cytoplasm with fragments of dense nuclear  they contain the first three BH domains (BH1-3).
chromatin. Because the cell shrinkage and formation of apoptotic bodies  On activation, BAX and/or BAK oligomerize within the
are rapid and the pieces are quickly cleared by phagocytes, considerable outer mitochondrial membrane and enhance its
apoptosis may occur in tissues before it is apparent in histologic sections. permeability
The absence of an inflammatory response can also make it difficult to  they form a channel in the outer mitochondrial
detect apoptosis by light microscopy. membrane that allows cytochrome c leakage from the
intermembranous space.
 Regulated apoptosis initiators.
Mechanisms of Apoptosis  BAD, BIM, BID, Puma, and Noxa, contain only one BH
 Apoptosis results from the activation of enzymes called caspases (so domain, the third of the four BH domains, and hence are
named because they are proteases containing a cysteine in their active sometimes called BH3-only proteins.
site and cleave proteins after aspartic residues). Like many proteases,  The activity of BH3-only proteins is modulated by
caspases exist as inactive proenzymes and must undergo enzymatic sensors of cellular stress and damage; when
cleavage to become active. upregulated and activated, they can initiate apoptosis.
 Growth factors and other survival signals stimulate the leading to autocatalytic cleavage and generation of active caspase-
production of anti-apoptotic proteins such as BCL2 thus 8.
protecting cells from apoptosis. o In turn, active caspase-8 initiates the same executioner caspase
 When cells are deprived of survival signals, suffer DNA sequence as in the mitochondrial pathway. This extrinsic apoptosis
damage, or develop ER stress due to the accumulation pathway can be inhibited by a protein called FLIP, which binds to
of misfolded proteins, BH3-only proteins are upregulated procaspase-8, thereby blocking FADD binding, but cannot activate
through increased transcription and/or post-translational the caspase. Some viruses and normal cells produce FLIP as a
modifications (e.g., phosphorylation). mechanism to protect themselves from Fasmediated apoptosis
 These BH3-only proteins in turn directly activate the two o The extrinsic and intrinsic pathways of apoptosis are initiated in
critical pro-apoptotic family members, BAX and BAK, fundamentally different ways by distinct molecules, but there may
which form oligomers that insert into the mitochondrial be interconnections between them. For instance, in hepatocytes
membrane and allow proteins from the inner and pancreatic β cells, caspase-8 produced by Fas signaling
mitochondrial membrane to leak out into the cytoplasm. cleaves and activates the BH3-only protein BID, which then feeds
 BH3-only proteins may also bind to and block the into the mitochondrial pathway. The combined activation of both
function of BCL2 and BCL-XL. At the same time, pathways delivers a fatal blow to the cells
synthesis of BCL2 and BCL-XL may decline because
their transcription relies on survival signals. Execution Phase of Apoptosis
 The net result of BAX-BAK activation coupled with loss  The intrinsic and extrinsic pathways converge to activate a caspase
of the protective functions of the anti-apoptotic BCL2 cascade that mediates the final phase of apoptosis. The intrinsic
family members is the release into the cytoplasm of mitochondrial pathway activates the initiator caspase-9
several mitochondrial proteins such as cytochrome c  extrinsic death receptor pathway activates caspase-8 and caspase-10.
that can activate the caspase cascade (see Fig. 2.14).  The active forms of these caspases trigger the rapid and sequential
 Once released into the cytosol, cytochrome c binds to a activation of the executioner caspases, such as caspase-3 and caspase-
protein called APAF-1 (apoptosis-activating factor-1), 6, which then act on many cellular components.
forming a multimeric structure called the apoptosome.  For instance, once activated these caspases cleave an inhibitor of a
 This complex binds to caspase-9, the critical initiator DNase, making the DNase enzymatically active and allowing DNA
caspase of the mitochondrial pathway, and promotes its degradation to commence. Caspases also proteolyze structural
autocatalytic cleavage, generating catalytically active components of the nuclear matrix and thus promote fragmentation of
forms of the enzyme. Active caspase-9 then triggers a nuclei. Other steps in apoptosis are less well-defined. For instance, we do
cascade of caspase activation by cleaving and thereby not know how membrane blebs and apoptotic bodies are formed.
activating other pro-caspases (such as caspase-3),
which mediate the execution phase of apoptosis Removal of Dead Cells
(discussed later). Other mitochondrial proteins with  The formation of apoptotic bodies breaks cells up into “bite-sized”
arcane names like Smac/DIABLO enter the cytoplasm, fragments that are edible for phagocytes.
where they bind to and neutralize cytoplasmic proteins  Apoptotic cells and their fragments also undergo several changes in their
that function as physiologic inhibitors of apoptosis membranes that actively promote their phagocytosis so they are most
(IAPs). The normal function of the IAPs is to block the often cleared before they lose membrane integrity and release their
inappropriate activation of caspases, including cellular contents.
executioners like caspase-3, and keep cells alive. Thus,  In healthy cells, phosphatidylserine is present on the inner leaflet of the
IAP inhibition permits initiation of the caspase cascade. plasma membrane, but in apoptotic cells this phospholipid “flips” out and
is expressed on the outer layer of the membrane, where it is recognized
 The Extrinsic (Death Receptor–Initiated) Pathway of Apoptosis by several macrophage receptors.
o This pathway is initiated by engagement of plasma membrane  Cells that are dying by apoptosis also secrete soluble factors that recruit
death receptors. Death receptors are members of the tumor phagocytes, and macrophages themselves may produce proteins that
necrosis factor (TNF) receptor family that contain a cytoplasmic bind to apoptotic cells (but not live cells), leading to their engulfment.
domain involved in protein-protein interactions. This death domain  Apoptotic bodies may also become coated with natural antibodies and
is essential for delivering apoptotic signals. proteins of the complement system, notably C1q, which are recognized by
o (Some TNF receptor family members do not contain cytoplasmic phagocytes.
death domains; their function is to activate inflammatory cascades  Thus, numerous ligands induced on apoptotic cells serve as “eat me”
[Chapter 3], and their role in triggering apoptosis is much less signals and are recognized by receptors on phagocytes that bind and
established.) engulf these cells. This process of apoptotic cell phagocytosis is called
o The best known death receptors are the type 1 TNF receptor efferocytosis; it is so efficient that dead cells disappear, often within
(TNFR1) and a related protein called Fas (CD95) minutes, without leaving a trace. In addition, production of pro-
o The ligand for Fas is called Fas ligand (FasL). inflammatory cytokines is reduced in macrophages that have ingested
o FasL is expressed on T cells that recognize self antigens (and apoptotic cells. Together with rapid clearance, this limits inflammatory
functions to eliminate self-reactive lymphocytes that also express reactions, even in the face of extensive apoptosis
the receptor Fas upon recognition of self antigens) and on some
CTLs that kill virus-infected and tumor cells. When FasL binds to APOPTOSIS
Fas, three or more molecules of Fas are brought together, and their  Regulated mechanism of cell death that serves to eliminate unwanted and
cytoplasmic death domains form a binding site for an adaptor irreparably damaged cells, with the least possible host reaction
protein called FADD (Fas-associated death domain).  Characterized by enzymatic degradation of proteins and DNA, initiated by
o Once attached to this complex, FADD binds inactivecaspase-8 (or caspases, and by recognition and removal of dead cells by phagocytes
caspase-10), bringing together multiple caspase molecules and  Initiated by two major pathways:
o Mitochondrial (intrinsic) pathway  The function of the inflammasome is to activate caspase-1 (also known as
 triggered by loss of survival signals, DNA damage, and interleukin-1β– converting enzyme), which cleaves a precursor form of
accumulation of misfolded proteins (ER stress), which interleukin-1 (IL-1) and releases its biologically active form.
leads to leakage of pro-apoptotic proteins from  IL-1 is a mediator of many aspects of inflammation, including leukocyte
mitochondrial membrane into the cytoplasm and recruitment and fever
subsequent caspase activation; can be inhibited by anti-  Caspase-1 and the closely related caspases-4 and -5 also induce death
apoptotic members of the BCL2 family, which are of the cells.
induced by survival signals including growth factors  Unlike classical apoptosis, this pathway of cell death is characterized by
o Death receptor (extrinsic) pathway release of inflammatory mediators.
 eliminates self-reactive lymphocytes and is a  Pyroptosis is thought to be the mechanism by which some microbes
mechanism of cell killing by cytotoxic T lymphocytes; is cause the death of infected cells and at the same time trigger local
initiated by engagement of death receptors (members of inflammation.
the TNF receptor family). The responsible ligands can
be soluble or expressed on the surface of adjacent cells Ferroptosis.
 distinct form of cell death that is triggered when excessive intracellular
Other Mechanisms of Cell Death levels of iron or reactive oxygen species overwhelm the glutathione-
Necroptosis. dependent antioxidant defenses to cause unchecked membrane lipid
 a hybrid that shares aspects of both necrosis and apoptosis. peroxidation.
 Morphologically, and to some extent biochemically, it resembles necrosis,  The widespread peroxidation of lipids disrupts many aspects of
as both are characterized by loss of ATP, swelling of the cell and membrane function, including fluidity, lipid-protein interactions, ion and
organelles, generation of reactive oxygen species (ROS), release of nutrient transport, and signaling pathways.
lysosomal enzymes, and ultimately rupture of the plasma membrane.  The overall effect is the loss of plasma membrane permeability, which
 Mechanistically, it is triggered by signal transduction pathways that ultimately leads to cell death resembling necrosis.
culminate in cell death, a feature similar to apoptosis. Because of these  The process is, however, regulated by specific signals (unlike necrosis)
overlapping features, necroptosis is sometimes called programmed and can be prevented by reducing iron levels (hence its name).
necrosis to distinguish it from forms of necrosis driven passively by toxic  Ultrastructurally, the most prominent features are the loss of mitochondrial
or ischemic injury to the cell. cristae and ruptured outer mitochondrial membrane.
 In sharp contrast to apoptosis, the signals leading to necroptosis do not  While its role in normal development and physiology remain controversial,
result in caspase activation, and hence it is also sometimes referred to as ferroptosis has been linked to cell death in a variety of human
“caspase-independent” programmed cell death. The process of pathologies, including cancer, neurodegenerative diseases, and stroke.
necroptosis starts in a manner similar to that of the extrinsic form of
apoptosis, that is, by ligation of a receptor by its ligand. NECROPTOSIS AND PYROPTOSIS
 Ligation of TNFR1 is the most widely studied model of necroptosis, but  Necroptosis resembles necrosis morphologically, but like apoptosis is a
many other signals, including ligation of Fas and yet to be identified genetically controlled form of cell death.
sensors of viral DNA and RNA, can also trigger necroptosis.  Necroptosis is triggered by ligation of TNFR1 and by proteins found in
 Since TNF can cause both apoptosis and necroptosis, the mechanisms RNA and DNA viruses
underlying these effects of TNF are especially illustrative (Fig. 2.16).  Necroptosis is caspase-independent and depends on the RIPK1 and
 Although the entire set of signaling molecules and their interactions are RIPK3 complex. RIPK1–RIPK3 signaling leads to the phosphorylation of
not known, necroptosis involves two kinases called receptor-interacting MLKL, which then forms pores in the plasma membrane.
protein kinase 1 and 3 (RIPK1 and RIPK3).  Release of cellular contents evokes an inflammatory reaction as in
 As indicated in Fig. 2.16, ligation of TNFR1 recruits these kinases into a necrosis.
multiprotein complex, and RIPK3 phosphorylates a cytoplasmic protein  Pyroptosis occurs in cells infected by microbes.It involves activation of
called MLKL. In response to its phosphorylation, MLKL monomers caspase-1, which cleaves the precursor form of IL-1 to generate
assemble into oligomers, translocate from the cytosol to the plasma biologically active IL-1. Caspase-1 along with other closely related
membrane, and cause the plasma membrane disruption that is caspases also cause death of the infected cell.
characteristic of necrosis. This explains the morphologic similarity of  Ferroptosis is an iron-dependent pathway of cell death induced by lipid
necroptosis with necrosis initiated by other injuries. peroxidation
 Necroptosis is postulated to be an important death pathway both in
physiologic and pathologic conditions. For example, physiologic Autophagy
necroptosis occurs during the formation of the mammalian bone growth
 a cell eats its own contents (Greek: auto, self; phagy, eating).
plate. In pathologic states, it is associated with cell death in
 It involves the delivery of cytoplasmic materials to the lysosome for
steatohepatitis, acute pancreatitis, ischemia-reperfusion injury, and
degradation.
neurodegenerative diseases such as Parkinson disease. Necroptosis also
 an evolutionarily conserved survival mechanism whereby, in states of
acts as a backup mechanism in host defense against certain viruses that
nutrient deprivation, starved cells live by cannibalizing themselves and
encode caspase inhibitors (e.g., cytomegalovirus)
recycling the digested contents.
Pyroptosis  implicated in many physiologic states (e.g., aging and exercise) and
pathologic processes.
 is a form of apoptosis that is accompanied by the release of the fever-
inducing cytokine IL-1 (pyro refers to fever).  It proceeds through several steps (Fig. 2.17):
o Nucleation and formation of an isolation membrane, also called a
 Microbial products that enter infected cells are recognized by cytoplasmic
innate immune receptors and can activate the multiprotein complex called phagophore; the isolation membrane is believed to be derived from
the inflammasome the ER, though other membrane sources such as the plasma
membrane and mitochondria may contribute
o Formation of a vesicle, called the autophagosome, from the  Autophagy is an adaptive response that is enhanced during nutrient
isolation membrane, inside which intracellular organelles and deprivation, allowing the cell to cannibalize itself to survive.
cytosolic structures are sequestered  Autophagosome formation is regulated by more than a dozen proteins
o Maturation of the autophagosome by fusion with lysosomes, to that act in a coordinated and sequential manner.
deliver digestive enzymes that degrade the contents of the  Dysregulation of autophagy occurs in many disease states, including
autophagosome cancer, inflammatory bowel diseases, and neurodegenerative disorders.
 In recent years, more than a dozen “autophagy-related genes” called Atgs  Autophagy plays a role in host defense against certain microbes
have been identified whose products are required for the creation of the
autophagosome. Environmental cues like nutrient deprivation or depletion MECHANISMS OF CELL INJURY
of growth factors activate an initiation complex of four proteins that
 Mitochondrial damage
promotes the hierarchical recruitment of Atgs to nucleate the initiation
 Membrane damage
membrane. The initiation membrane elongates further, surrounds and
 Damage to DNA
captures its cytosolic cargo, and closes to form the autophagosome. The
elongation and closure of the initiation membrane require the coordinated  Oxidative Stress: Accumulation of Oxygen-Derived Free Radicals
action of two ubiquitin-like conjugation systems that result in the covalent  Disturbance in Calcium Homeostasis
linkage of the lipid phosphatidylethanolamine (PE) to microtubule-  Endoplasmic reticulum Stress: the unfolded protein response
associated protein light chain 3 (LC3). PE-lipidated LC3 is increased
during autophagy, and it is therefore a useful marker for identifying cells in  The discussion of the pathways of cell injury and death sets the stage for
which autophagy is occurring. The newly formed autophagosome fuses a consideration of the underlying biochemical mechanisms of cell injury.
with lysosomes to form an autophagolysosome. In the terminal step, the The molecular alterations that lead to cell injury are complex, but several
inner membrane and enclosed cytosolic cargoes are degraded by principles are relevant to most forms of cell injury:
lysosomal enzymes. There is increasing evidence that autophagy is not a  The cellular response to injurious stimuli depends on the nature of the
random process that engulfs cytosolic contents indiscriminately. Rather, injury, its duration, and its severity. Small doses of a chemical toxin or
the loading of cargo into the autophagosome is selective, and one of the brief periods of ischemia may induce reversible injury, whereas large
functions of the lipidated LC3 is to target protein aggregates and effete doses of the same toxin or more prolonged ischemia might result either in
organelles rapid cell death or in slowly progressive injury that with time becomes
 Autophagy functions as a survival mechanism under various stress irreversible and leads to cell death
conditions, maintaining the integrity of cells by recycling essential  The consequences of cell injury depend on the type, state, and
metabolites and clearing intracellular debris. It is therefore prominent in adaptability of the injured cell. The cell’s nutritional and hormonal status,
atrophic cells exposed to severe nutrient deprivation. Autophagy is also metabolic demands, and functions dictate the response to injury. How
involved in the turnover of organelles like the ER, mitochondria, and vulnerable is a cell, for example, to loss of blood supply and hypoxia?
lysosomes and the clearance of intracellular aggregates that accumulate When a skeletal muscle cell in the leg is deprived of its blood supply, it
during aging, stress, and various disease states. Autophagy can trigger can be rested and preserved; cardiac muscle cells have no such option.
cell death if it is inadequate to copewith the stressor. This pathway of cell Exposure of two individuals to identical concentrations of a toxin, such as
death is distinct from necrosis and apoptosis, but the mechanism is carbon tetrachloride, may produce no effect in one and cell death in the
unknown. Furthermore, it is not clear whether cell death is caused by other. This may be due to polymorphisms in genes encoding hepatic
autophagy or by the stress that triggered autophagy. Nevertheless, enzymes that metabolize carbon tetrachloride (CCl4) to toxic by-products
autophagic vacuolization often precedes or accompanies cell death. (Chapter 9). With the complete mapping of the human genome, there is
There is accumulating evidence that autophagy plays a role in human great interest in identifying genetic polymorphisms that affect the
diseases, including the following: responses of different individuals to various injurious agents.
o Cancer: Autophagy can both promote cancer growth and act as a  Any injurious stimulus may simultaneously trigger multiple interconnected
defense against cancers. This is an area of active investigation, as mechanisms that damage cells. This is one reason why it can be difficult
discussed in Chapter 7. to ascribe cell injury in a particular situation to a single or even dominant
o Neurodegenerative disorders: Many neurodegenerative disorders biochemical derangement
are associated with dysregulation of autophagy. Alzheimer disease  We start this section with a discussion of general mechanisms that are
is characterized by impaired autophagosome maturation, and in involved in reversible injury and necrosis caused by diverse stimuli and
mouse models of the disease genetic defects in autophagy conclude with a discussion of the pathways of injury in selected clinical
accelerate neurodegeneration. In Huntington disease, mutant situations that illustrate general principles
huntingtin impairs autophagy.
o Infectious diseases: Many pathogens are degraded by autophagy; General Mechanisms of Cell Injury and Intracellular Targets of
these include mycobacteria, Shigella spp., and HSV-1. This is one Injurious Stimuli
way by which microbial proteins are digested and delivered to  Cell injury results from abnormalities in one or more essential cellular
antigen presentation pathways. Macrophage-specific deletion of components (Fig. 2.18). The principal targets of injurious stimuli are
Atg5 increases susceptibility to tuberculosis. mitochondria, cell membranes, the machinery of protein synthesis and
o Inflammatory bowel diseases: Genome-wide association studies secretion, and DNA. The consequences of injury of each of these cellular
have linked both Crohn disease and ulcerative colitis to single- components are distinct but overlapping
nucleotide polymorphisms (SNPs) in the autophagy-related gene
ATG16L1. How these polymorphisms promote intestinal Mitochondrial Damage Mitochondria are critical players in all pathways
inflammation is not known.
leading to cell injury and death.
 This should be expected because mitochondria supply life-sustaining
KEY CONCEPTS AUTOPHAGY
energy by producing ATP but are also targets of many injurious stimuli.
 Autophagy involves sequestration of cellular organelles into cytoplasmic
Thus, in many ways, they are the arbiters of life and death of cells.
autophagic vacuoles (autophagosomes) that fuse with lysosomes and
Mitochondria can be damaged by increases of cytosolic Ca2+ , ROS
digest enclosed material.
(discussed later), and oxygen deprivation, which makes them sensitive to to channel formation by pro-apoptotic BAX and BAK is the initial
virtually all types of injurious stimuli, including hypoxia and toxins. In step in apoptosis by the intrinsic pathway. This action of BAX and
addition, mutations in mitochondrial genes are the cause of some BAK is specific to mitochondrial membranes only and leads to
inherited diseases (Chapter 5). damage of other organelles indirectly.
 There are three major consequences of mitochondrial damage.
o ATP depletion
 Decreased ATP synthesis and ATP depletion are frequently Membrane Damage
associated with both hypoxic and chemical (toxic) injury (Fig.  Causes
2.19). o ROS
 ATP is produced in two ways. The major pathway in o Decreased phospholipid synthesis
mammalian cells, particularly those that are nondividing (e.g., o Increased phospholipid breakdown.
brain and liver), is oxidative phosphorylation of adenosine o Cytoskeletal abnormalities
diphosphate in a reaction that results in reduction of oxygen by
 types
the mitochondrial electron transport system.
o Mitochondrial membrane damage
 The second is the glycolytic pathway, which can generate
o Plasma membrane damage
ATP, albeit in far smaller amounts, in the absence of oxygen
using glucose derived either from body fluids or from the o Injury to lysosomal membranes
hydrolysis of glycogen.
 In addition to mitochondrial damage, the major causes of ATP  Early loss of selective membrane permeability, leading ultimately to overt
depletion are reduced supply of oxygen and nutrients membrane damage, is a consistent feature of most forms of cell injury
(because of ischemia and hypoxia), and the actions of some (except apoptosis). Membrane damage may affect the integrity and
toxins (e.g., cyanide). Mitochondrial damage often results in functions of all cellular membranes. In ischemic cells, membrane defects
the formation of a high-conductance channel in the may be the result of ATP depletion and calcium-mediated activation of
mitochondrial membrane, called the mitochondrial permeability phospholipases. The plasma membrane can also be damaged directly by
transition pore (see Fig. 2.19). The opening of bacterial toxins, viral proteins, lytic complement components, and a
thisconductance channel leads to the loss of mitochondrial variety of physical and chemical agents.
membrane potential, resulting in failure of oxidative
phosphorylation and progressive ATP depletion that  Several biochemical mechanisms may contribute to membrane damage
culminates in necrosis. (Fig. 2.20):
o High-energy phosphate in the form of ATP is required for virtually  ROS. Oxygen free radicals cause injury to cell membranes by lipid
all synthetic and degradative processes within the cell. These peroxidation, discussed later.
include membrane transport, protein synthesis, lipogenesis, and the  Decreased phospholipid synthesis. The production of phospholipids in
deacylation-reacylation reactions necessary for phospholipid cells may be reduced as a consequence of defective mitochondrial
turnover. Hence, depletion of ATP to 5% to 10% of normal levels function or hypoxia, both of which decrease ATP production and thus
has widespread effects on many critical cellular systems. affect energydependent biosynthetic pathways. Decreased phospholipid
o • The activity of the plasma membrane energy-dependent sodium synthesis may affect all cellular membranes, including those of
pump (Na+ ,K+ -ATPase) is reduced (Chapter 1). Failure of this mitochondria.
active transport system causes sodium to enter and accumulate  Increased phospholipid breakdown. Severe cell injury is associated with
inside cells and potassium concentrations to fall. The net solute increased degradation of membrane phospholipids, probably due to
gain results in osmotically driven water accumulation that leads to activation of calciumdependent phospholipases by increased cytosolic
cell swelling and ER dilation. and mitochondrial Ca2+ . Phospholipid breakdown leads to the
o • Cellular energy metabolism is altered. If the supply of oxygen to accumulation of lipid breakdown products, including unesterified free fatty
cells is reduced, as in ischemia, oxidative phosphorylation ceases, acids, acyl carnitine, and lysophospholipids, which have a detergent effect
resulting in a decrease in cellular ATP and associated increase in on membranes. They may also either insert into the lipid bilayer of the
adenosine monophosphate. These changes stimulate membrane or exchange with membrane phospholipids, potentially
phosphorylase and phosphofructokinase activities, leading to causing changes in permeability and electrophysiologic alterations.
increased rates of glycogenolysis and glycolysis, respectively, in an  Cytoskeletal abnormalities. Cytoskeletal filaments serve as anchors
effort to maintain energy supplies by generating ATP through connecting the plasma membrane to the cell interior, and proteases
metabolism of glucose derived from glycogen. As a consequence, activated by cytosolic Ca2+ may damage these tethers. When
glycogen stores are rapidly depleted. Glycolysis under anaerobic exacerbated by cell swelling, particularly in myocardial cells, this damage
conditions results in the accumulation of lactic acid and inorganic leads to detachment of the cell membrane from the cytoskeleton,
phosphates from the hydrolysis of phosphate esters. This reduces rendering it susceptible to stretching and rupture.
the intracellular pH, resulting in decreased activity of many cytosolic  Damage to different cellular membranes has diverse effects on cells.
enzymes.  Mitochondrial membrane damage. As discussed earlier, damage to
o • With prolonged or worsening depletion of ATP, structural mitochondrial membranes results in opening of the mitochondrial
disruption of the protein synthetic apparatus occurs, manifest as permeability transition pore, leading to decreased ATP generation and
detachment of ribosomes from the rough ER and dissociation of release of proteins that trigger apoptotic death.
polysomes, with a consequent reduction in protein synthesis. There  Plasma membrane damage. Plasma membrane damage results in loss of
may also be increased protein misfolding, with injurious effects that osmotic balance and influx of fluids and ions, as well as loss of cellular
are discussed later. • Ultimately, there is irreversible damage to contents. The cells may also leak metabolites (e.g., glycolytic
mitochondrial and lysosomal membranes, and the cell undergoes intermediates) that are vital for the reconstitution of ATP, thus further
necrosis. • Incomplete oxidative phosphorylation also leads to the depleting energy stores. •
formation of ROS, which have many deleterious effects, described  Injury to lysosomal membranes results in leakage of their enzymes into
later. • As discussed earlier, leakage of mitochondrial proteins due the cytoplasm and activation of acid hydrolases in the acidic intracellular
pH of the injured cell. These lysosomal hydrolases include RNases, been implicated in a wide variety of pathologic processes, including cell
DNases, proteases, phosphatases, and glucosidases, which degrade injury, cancer, aging, and some degenerative diseases, such as
RNA, DNA, proteins, phosphoproteins, and glycogen, respectively, and Alzheimer disease. ROS are also produced in large amounts by activated
push cells into necrosis. leukocytes, particularly neutrophils and macrophages, during
inflammatory reactions aimed at destroying microbes and cleaning up
dead cells and other unwanted substances (Chapter 3). The following
Damage to DNA section discusses the generation and removal of ROS, and how they
 Damage to nuclear DNA activates sensors that trigger p53-dependent contribute to cell injury. The properties of some of the most important free
pathways (Chapter 7). DNA damage may be caused by exposure to radicals are summarized in Table 2.2.
radiation, chemotherapeutic (anticancer) drugs, and ROS, or may occur
spontaneously as a part of aging, due largely to deamination of cytosine
residues to uracil residues. DNA damage activates p53, which arrests Generation of Free Radicals
cells in the G1 phase of the cell cycle and activates DNA repair  Free radicals may be generated within cells in several ways (Fig. 2.21):
mechanisms. If these mechanisms fail to correct the DNA damage, p53  The reduction-oxidation reactions that occur during normal metabolic
triggers apoptosis by the mitochondrial pathway. Thus, the cell chooses to processes. As a part of normal respiration, molecular O2 is reduced by
die rather than survive with abnormal DNA that has the potential to induce the transfer of four electrons to H2 to generate two water molecules. This
malignant transformation. Predictably, mutations in p53 that interfere with conversion is catalyzed by oxidative enzymes in the ER, cytosol,
its ability to arrest cell cycling or to induce apoptosis are associated with mitochondria, peroxisomes, and lysosomes. During this process, small
numerous cancers (Chapter 7). In addition to damage to various amounts of partially reduced intermediates are produced in which
organelles, some biochemical alterations are involved in many situations different numbers of electrons have been transferred from O2; these
that lead to cell injury. Two of these general pathways are discussed next. include superoxide anion (O2 • , one electron), hydrogen peroxide (H2O2,
two electrons), and hydroxyl radicals (˙OH, three electrons).
 Absorption of radiant energy (e.g., ultraviolet light, x-rays). For example,
ionizing radiation can hydrolyze water into ˙OH and hydrogen (H) free
Oxidative Stress: Accumulation of Oxygen-Derived Free Radicals radicals.
 Generation of Free Radicals  Rapid bursts of ROS are produced in activated leukocytes during
o reduction-oxidation reactions inflammation. This occurs in a precisely controlled reaction carried out by
o Absorption of radiant energy a plasma membrane multiprotein complex that uses NADPH oxidase for
the redox reaction (Chapter 3). In addition, some intracellular oxidases
o in activated leukocytes during inflammation
(e.g., xanthine oxidase) generate O2 • . Defects in leukocytic superoxide
o Enzymatic metabolism of exogenous chemicals or drugs
production lead to chronic granulomatous disease
o Transition metals
 Enzymatic metabolism of exogenous chemicals or drugs can generate
o Nitric Oxide free radicals that are not ROS but have similar effects (e.g., CCl4 can
 Removal of Free Radicals generate ˙CCl3, described later in the chapter).
o antioxidants  Transition metals such as iron and copper donate or accept free electrons
o iron, copper during intracellular reactions and catalyze free radical formation, as in the
o enzyme Fenton reaction (H2O2 + Fe2+ → Fe3+ + ˙OH + OH− ). Because most of
 catalase the intracellular free iron is in the ferric (Fe3+ ) state, it must be reduced
 superoxidase dismutase to the ferrous (Fe2+ ) form to participate in the Fenton reaction. This
 glutathione peroxidase reduction can be enhanced by O2 • , and hus sources of iron and O2 •
 Pathologic Effects of Free Radicals may cooperate in oxidative cell damage.
o Lipid peroxidation in membranes  Nitric oxide (NO), an important chemical mediator generated by
o Oxidative modification of proteins endothelial cells, macrophages, neurons, and other cell types (Chapter 3),
o Lesions in DNA. can act as a free radical and may also be converted to highly reactive
peroxynitrite anion (ONOO− ) as well as NO2 and NO3
 Cell injury induced by free radicals, particularly ROS, is an important
mechanism of cell damage in many pathologic conditions, such as
chemical and radiation injury, ischemiareperfusion injury (induced by Removal of Free Radicals
restoration of blood flow in ischemic tissue), cellular aging, and microbial  Free radicals are inherently unstable and generally decay spontaneously.
killing by phagocytes. Free radicals are chemical species that have a O2 • , for example, is unstable and decays (dismutates) spontaneously to
single unpaired electron in an outer orbit. Unpaired electrons are highly O2 and H2O2 in the presence of water. In addition, cells have developed
reactive and “attack” and modify adjacent molecules, such as inorganic or multiple nonenzymatic and enzymatic mechanisms to remove free
organic chemicals—proteins, lipids, carbohydrates, nucleic acids—many radicals and thereby minimize injury (see Fig. 2.21). These include the
of which are key components of cell membranes and nuclei. Some of following: • Antioxidants either block free radical formation or inactivate
these reactions are autocatalytic, whereby molecules that react with free (e.g., scavenge) free radicals. Examples are the lipid-soluble vitamins E
radicals are themselves converted into free radicals, thus propagating the and A as well as ascorbic acid and glutathione in the cytosol. • As we
chain of damage. ROS are a type of oxygen-derived free radical whose have seen, free iron and copper can catalyze the formation of ROS.
role in cell injury is well established. ROS are produced normally in cells Under normal circumstances, the reactivity of these metals is minimized
during mitochondrial respiration and energy generation, but they are by their binding to storage and transport proteins (e.g., transferrin, ferritin,
degraded and removed by intracellular ROS scavengers. These defense lactoferrin, and ceruloplasmin), which prevents these metals from
systems allow cells to maintain a steady state in which free radicals may participating in reactions that generate ROS. • Several enzymes act as
be present at low concentrations but do not cause damage. Increased free radical–scavenging systems and break down H2O2 and O2 • . These
production or decreased scavenging of ROS may lead to an excess of enzymes are located near the sites of generation of the oxidants and
free radicals, a condition called oxidative stress. Oxidative stress has include the following: 1. Catalase, present in peroxisomes, decomposes
H2O2 (2H2O2 → O2 + 2H2O). 2. Superoxidase dismutases (SODs) are responsible for DNA and chromatin fragmentation), and ATPases
found in many cell types and convert O2 • to H2O2 ( 2O2 • + 2H → H2O2 (thereby hastening ATP depletion)
+ O2). This group of enzymes includes both manganeseSOD, which is
localized in mitochondria, and copperzinc-SOD, which is found in the Endoplasmic Reticulum Stress: the Unfolded Protein Response
cytoplasm. 3. Glutathione peroxidase also protects against injury by  The accumulation of misfolded proteins in the ER can stress adaptive
catalyzing free radical breakdown (H2O2 + 2GSH → GSSG [glutathione mechanisms and trigger apoptosis. Chaperones in the ER control the
homodimer] + 2H2O, or 2˙OH + 2GSH → GSSG + 2H2O). The proper folding of newly synthesized proteins, and misfolded polypeptides
intracellular ratio of oxidized glutathione (GSSG) to reduced glutathione are shuttled into the cytoplasm where they are ubiquitinated and targeted
(GSH) reflects the oxidative state of the cell and is an important indicator for proteolysis in proteasomes (Chapter 1). If, however, unfolded or
of the cell’s ability to detoxify ROS. misfolded proteins accumulate in the ER, they trigger a number of
alterations that are collectively called the unfolded protein response. The
Pathologic Effects of Free Radicals unfolded protein response activates signaling pathways that increase the
 The effects of ROS and other free radicals are wide-ranging, but three production of chaperones, enhance proteasomal degradation of abnormal
reactions are particularly relevant to cell injury (see Fig. 2.21): proteins, and slow protein translation, thus reducing the load of misfolded
 Lipid peroxidation in membranes. In the presence of O2, free radicals proteins in the cell (Fig. 2.23). However, if this cytoprotective response is
may cause peroxidation of lipids within plasma and organellar unable to cope with the accumulation of misfolded proteins, the cell
membranes. Oxidative damage is initiated when the double bonds in activates caspases and induces apoptosis. This process is called ER
unsaturated fatty acids of membrane lipids are attacked by O2-derived stress. Intracellular accumulation of misfolded proteins may be caused by
free radicals, particularly by OH. The lipid–free radical interactions yield an increased rate of misfolding or a reduction in the cell’s ability to repair
peroxides, which are themselves unstable and reactive, and an or eliminate them. Increased misfolding may be a consequence of
autocatalytic chain reaction ensues (called propagation) that can result in deleterious mutations or decreased capacity to correct misfolded proteins,
extensive membrane damage. as occurs in aging. Protein misfolding may also be increased in viral
 Oxidative modification of proteins. Free radicals promote oxidation of infections when proteins encoded by the viral genome are synthesized in
amino acid side chains, formation of covalent protein-protein cross-links such large quantities that they overwhelm the quality control system that
(e.g., disulfide bonds), and oxidation of the protein backbone. Oxidative normally ensures proper protein folding. Increased demand for secretory
modifications may also damage the active sites of enzymes, disrupt the proteins such as insulin in insulin-resistant states, and changes in
conformation of structural proteins, and enhance proteasomal intracellular pH and redox state are other stressors that result in misfolded
degradation of unfolded or misfolded proteins, thereby raising havoc protein accumulation. Protein misfolding is thought to be the causative
throughout the cell cellular abnormality in several neurodegenerative diseases (Chapter 28).
 Lesions in DNA. Free radicals are capable of causing single- and double- Given that many “foldases” require ATP to function, deprivation of glucose
strand breaks in DNA, cross-linking DNA strands, and forming adducts. and oxygen, as in ischemia and hypoxia, also may increase the burden of
Oxidative DNA damage has been implicated in cell aging (discussed later misfolded proteins. Diseases caused by misfolded proteins are listed in
in the chapter) and in malignant transformation of cells (Chapter 7). The Table 2.3.
traditional thinking about free radicals was that they cause cell injury and
death by necrosis, and, in fact, the production of ROS is often a prelude MECHANISMS OF CELL INJURY
to necrosis. However, it is now clear that free radicals can also trigger  ATP depletion: failure of energy-dependent functions → reversible injury
apoptosis. It is also possible that these potentially deadly molecules, → necrosis
when produced under controlled conditions in the “right” dose, serve  Mitochondrial damage: ATP depletion → failure of energydependent
important physiologic functions in signaling by cellular receptors and other cellular functions → ultimately, necrosis; under some conditions, leakage
pathways. of mitochondrial proteins that cause apoptosis
 Increased permeability of cellular membranes: may affect plasma
membrane, lysosomal membranes, mitochondrial membranes; typically
Disturbance in Calcium Homeostasis culminates in necrosis
 Calcium ions normally serve as second messengers in several signaling  Accumulation of damaged DNA and misfolded proteins:triggers apoptosis
pathways, but if released into the cytoplasm of cells in excessive o Accumulation of ROS: covalent modification of cellular proteins,
amounts, are also an important source of cell injury. In keeping with this, lipids, nucleic acids
calcium depletion protects cultured cells from injury induced by a variety o Influx of calcium: activation of enzymes that damage cellular
of harmful stimuli. Cytosolic free Ca2+ is normally maintained at very low components and may also trigger apoptosis
concentrations (~0.1 µmol) compared with extracellular levels of 1.3 o Unfolded protein response and ER stress: Accumulation of
mmol, and most intracellular Ca2+ is sequestered in mitochondria and the misfolded proteins in the ER activates adaptive mechanisms that
ER. Ischemia and certain toxins cause an excessive increase in cytosolic help the cell to survive, but if their repair capacity is exceeded they
Ca2+ , initially because of release from intracellular stores, and later due trigger apoptosis
to increased influx across the plasma membrane (Fig. 2.22). Excessive
intracellular Ca2+ may cause cell injury by several mechanisms, although
CLINICOPATHOLOGIC CORRELATIONS: SELECTED
the significance of these mechanisms in cell injury in vivo is not
established. EXAMPLES OF CELL INJURY AND DEATH
 The accumulation of Ca2+ in mitochondria results in opening of the  Hypoxia and Ischemia
mitochondrial permeability transition pore and, as described earlier, failure  Ischemia-Perfusion Injury
of ATP generation  Chemical (Toxic) Injury
 Increased cytosolic Ca2+ activates a number of enzymes with potentially Having briefly reviewed the causes, morphology, and general mechanisms of
deleterious effects on cells. These enzymes include phospholipases cell injury and death, we now describe some common and clinically significant
(which cause membrane damage), proteases (which break down both forms of cell injury. These examples illustrate many of the mechanisms and
membrane and cytoskeletal proteins), endonucleases (which are sequence of events in cell injury described earlier.
Hypoxia and Ischemia reperfused tissue. The inflammation causes additional tissue injury
 Ischemia, the most common cause of cell injury in clinical medicine, (Chapter 3). The importance of neutrophil influx in reperfusion injury has
results from hypoxia induced by reduced blood flow, most often due to a been demonstrated experimentally by the salutary effects of treatment
mechanical arterial obstruction. It can also occur due to reduced venous with antibodies that block cytokines or adhesion molecules and thereby
drainage. In contrast to hypoxia, in which blood flow is maintained and reduce neutrophil extravasation. • Activation of the complement system
during which energy production by anaerobic glycolysis can continue, may contribute to ischemia-reperfusion injury. For unknown reasons,
ischemia compromises the delivery of substrates for glycolysis. Thus, in some IgM antibodies have a propensity to deposit in ischemic tissues.
ischemic tissues, not only does aerobic metabolism cease but anaerobic When blood flow is resumed, complement proteins bind to the deposited
energy generation also fails after glycolytic substrates are exhausted or antibodies, are activated, and exacerbate cell injury and inflammation
glycolysis is inhibited by the accumulation of metabolites, which otherwise
would be washed out by flowing blood. For this reason, ischemia causes Chemical (Toxic) Injury
more rapid and severe cell and tissue injury than hypoxia. Mechanisms of  Chemical injury remains a frequent problem in clinical medicine and is a
Ischemic Cell Injury The sequence of events following hypoxia or major limitation to drug therapy. Because many drugs are metabolized in
ischemia reflects many of the biochemical alterations in cell injury, the liver, this organ is a major target of drug toxicity. In fact, toxic liver
summarized here in Fig. 2.24 and shown earlier in Figs. 2.5 and 2.6. As injury is often the reason for terminating the therapeutic use or
intracellular oxygen tension falls, oxidative phosphorylation fails and ATP development of a drug. The mechanisms by which chemicals, certain
generation decreases. The consequences of ATP depletion were drugs, and toxins produce injury are described in greater detail in Chapter
described earlier in the Mitochondrial Damage section. In brief, loss of 9 in the discussion of environmental diseases. Here the major pathways
ATP results initially in reversible cell injury (cell and organelle swelling) of chemically induced injury with selected examples are described.
and later in cell death by necrosis. Mammalian cells have developed  Chemicals induce cell injury by one of two general mechanisms
protective responses to deal with hypoxic stress. The best defined of  Direct toxicity. Some chemicals injure cells directly by combining with
these is induction of a transcription factor called hypoxia-inducible factor-1 critical molecular components. For example, in mercuric chloride
(HIF-1), which promotes new blood vessel formation, stimulates cell poisoning, mercury binds to the sulfhydryl groups of cell membrane
survival pathways, and enhances glycolysis. Several promising proteins, causing increased membrane permeability and inhibition of ion
investigational compounds that promote HIF-1 signaling are being transport. In such instances, the greatest damage is usually to the cells
developed. Nevertheless, there are still no reliable therapeutic that use, absorb, excrete, or concentrate the chemicals—in the case of
approaches for reducing the injurious consequences of ischemia in mercuric chloride, the cells of the gastrointestinal tract and kidney.
clinical situations. The strategy that is perhaps the most useful in ischemic Cyanide poisons mitochondrial cytochrome oxidase and thus inhibits
(and traumatic) brain and spinal cord injury is the transient induction of oxidative phosphorylation. Many antineoplastic chemotherapeutic agents
hypothermia (lowering the core body temperature to 92°F). This treatment and antibiotics also induce cell damage by direct cytotoxic effects.
reduces the metabolic demands of the stressed cells, decreases cell  Conversion to toxic metabolites. Most toxic chemicals are not biologically
swelling, suppresses the formation of free radicals, and inhibits the host active in their native form but must be converted to reactive toxic
inflammatory response. All of these may contribute to decreased cell and metabolites, which then act on target molecules. This modification is
tissue injury. usually accomplished by the cytochrome P-450 mixed-function oxidases
in the smooth ER of the liver and other organs. The toxic metabolites
Ischemia-Reperfusion Injury cause membrane damage and cell injury mainly by formation of free
 Restoration of blood flow to ischemic tissues can promote recovery of radicals and subsequent lipid peroxidation; direct covalent binding to
cells if they are reversibly injured but can also paradoxically exacerbate membrane proteins and lipids may also contribute. For instance, CCl4,
cell injury and cause cell death. As a consequence, reperfused tissues which was once widely used in the dry cleaning industry, is converted by
may sustain loss of viable cells in addition to those that are irreversibly cytochrome P-450 to the highly reactive free radical ˙CCl3, which causes
damaged by the ischemia. This process, called ischemia-reperfusion lipid peroxidation and damages many cellular structures. The analgesic
injury, is clinically important because it contributes to tissue damage drug acetaminophen is also converted to a toxic product during
during myocardial and cerebral infarction following therapies that restore detoxification in the liver, leading to cell injury. These and other examples
blood flow (Chapters 12 and 28). How does reperfusion injury occur? The of chemical injury are described in Chapter 9.
likely answer is that new damaging processes are set in motion during
reperfusion, causing the death of cells that might have recovered
otherwise. Several mechanisms have been proposed: • Oxidative stress. EXAMPLES OF CELL INJURY
New damage may be initiated during reoxygenation by increased  Mild ischemia: Reduced oxidative phosphorylation → ATP depletion
generation of reactive oxygen and nitrogen species. These free radicals → failure of Na pump → influx of sodium and water → organelle
may be produced in reperfused tissue as a result of incomplete reduction and cellular swelling (reversible)
of oxygen in leukocytes, and in damaged endothelial cells and  Severe/prolonged ischemia: Severe swelling of mitochondria,
parenchymal cells. Compromise of cellular antioxidant defense calcium influx into mitochondria and into the cell with rupture of
mechanisms during ischemia may sensitize cells to free radical damage. • lysosomes and plasma membrane. Death by necrosis and apoptosis
Intracellular calcium overload. As mentioned earlier, intracellular and due to release of cytochrome c from mitochondria. • Reperfusion
mitochondrial calcium overload begins during acute ischemia; it is injury follows restoration of blood flow to ischemic tissues and is
exacerbated during reperfusion due to influx of calcium resulting from cell caused by oxidative stress due to release of free radicals from
membrane damage and ROS-mediated injury to sarcoplasmic reticulum. leukocytes and endothelial cells. Blood brings calcium that overloads
Calcium overload favors opening of the mitochondrial permeability reversibly injured cells with consequent mitochondrial injury, as well
transition pore with resultant ATP depletion. This, in turn, causes further as oxygen and leukocytes, which generate free radicals and
cell injury. • Inflammation. Ischemic injury is associated with inflammation cytokines. Complement may be activated locally by IgM antibodies
as a result of “danger signals” released from dead cells, cytokines deposited in ischemic tissues.
secreted by resident immune cells such as macrophages, and increased  Chemicals may cause injury directly or by conversion into toxic
expression of adhesion molecules by hypoxic parenchymal and metabolites. The organs chiefly affected are those involved in
endothelial cells, all of which act to recruit circulating neutrophils to
absorption or excretion of chemicals or others such as liver, where because beyond a certain point, it becomes maladaptive. Hypertrophy
the chemicals are converted to toxic metabolites. Direct injury to results from the action of growth factors and direct effects on cellular
critical organelles such as mitochondria or indirect injury from free proteins (Fig. 2.26):
radicals generated from the chemicals/toxins is involved.  Mechanical sensors in the cell detect the increased load.
 These sensors activate a complex downstream web of signaling
ADAPTATIONS OF CELLULAR GROWTH AND pathways, including the phosphoinositide 3-kinase (PI3K)/AKT pathway
DIFFERENTIATION (postulated to be most important in physiologic, e.g., exercise-induced,
 Hypertrophy hypertrophy) and G-protein–coupled receptor–initiated pathways
o Pathologic (activated by many growth factors and vasoactive agents, and thought to
be more important in pathologic hypertrophy).
o Physiologic
 Some of the signaling pathways stimulate increased production of growth
 Hyperplasia
factors (e.g. TGF-β, insulin-like growth factor 1 [IGF1], fibroblast growth
 Atrophy
factor) and vasoactive agents (e.g., α-adrenergic agonists, endothelin-1,
 Metaplasia and angiotensin II). These and other pathways activate transcription
factors, including GATA4, nuclear factor of activated T cells (NFAT), and
Adaptations are reversible changes in the size, number, phenotype, metabolic myocyte enhancer factor 2 (MEF2), which increase the expression of
activity, or functions of cells in response to changes in their environment. genes that encode muscle proteins. Cardiac hypertrophy is also
 Cells capable of division: hyperplasia and hypertrophy associated with a switch in gene expression from genes that encode
 Nondividing cells (e.g., myocardial fibers): hypertrophy. adult-type contractile proteins to genes that encode functionally distinct
 Hypertrophy and hyperplasia may coexist, with both contributing to fetal isoforms of the same proteins. For example, the α isoform of myosin
increased organ size. heavy chain is replaced by the β isoform, which has a slower, more
energetically economical contraction. Other proteins that are altered in
hypertrophic myocardial cells are the products of genes that participate in
Hypertrophy the cellular response to stress. For example, cardiac hypertrophy is
 Hypertrophy is an increase in the size of cells that results in an increase associated with increased atrial natriuretic factor gene expression. Atrial
in the size of the affected organ. natriuretic factor is a peptide hormone that causes salt secretion by the
 The hypertrophied organ has no new cells, just larger cells. kidney, decreases blood volume and pressure, and therefore serves to
 The increased size of the cells is due to the synthesis and assembly of reduce hemodynamic load. Whatever the exact cause and mechanism of
additional intracellular structural components. cardiac hypertrophy, it eventually reaches a limit beyond which
 Pathologic hypertrophy. enlargement of muscle mass is no longer able to cope with the increased
o The striated muscle cells in the heart and skeletal muscles have burden. At this stage, several regressive changes occur in the myocardial
fibers, of which the most important are degradation and loss of myofibrillar
only a limited capacity for division, and respond to increased
contractile elements. In extreme cases, myocyte death can occur. The net
metabolic demands mainly by undergoing hypertrophy.
result of these changes is cardiac failure, a sequence of events that
o most common stimulus for hypertrophy of skeletal and cardiac
illustrates how an adaptation to stress can progress to functionally
muscle: increased workload.
significant cell injury if the stress is not relieved.
o In both tissue types, muscle cells respond by synthesizing more
protein and increasing the number of myofilaments per cell. This in
turn increases the amount of force each myocyte can generate and Hyperplasia
thus the strength and work capacity of the muscle as a whole.  Hyperplasia is an increase in the number of cells in an organ or tissue in
o example response to a stimulus. Although hyperplasia and hypertrophy are distinct
 hypertension or valvular disease processes, they frequently occur together, and may be triggered by the
 pressure overload same external stimuli. Hyperplasia can only take place if the tissue
 enlargement of the heart contains cells capable of dividing, thus increasing the number of cells. It
 Initially, it improves heart’s function can be physiologic or pathologic.
 over time this adaptation often fails  heart failure  Physiologic hyperplasia. Physiologic hyperplasia due to the action of
hormones or growth factors occurs when there is a need to increase
 Physiologic hypertrophy functional capacity of hormone sensitive organs, or when there is need for
o caused by increased functional demand or stimulation by hormones compensatory increase after damage or resection. Hormonal hyperplasia
is well illustrated by the proliferation of the glandular epithelium of the
and growth factors
female breast at puberty and during pregnancy, usually accompanied by
o The massive physiologic growth of the uterus during pregnancy is a
enlargement (hypertrophy) of the glandular epithelial cells. The classic
good example of hormone-induced enlargement of an organ that
illustration of compensatory hyperplasia comes from the study of liver
results mainly from hypertrophy of smooth muscle fibers (Fig. 2.25).
regeneration. In individuals who donate one lobe of the liver for
Uterine hypertrophy during pregnancy is stimulated by estrogenic
transplantation, the remaining cells proliferate so that the organ soon
hormone signaling through estrogen receptors that eventually result
grows back to its original size. Experimental models of partial
in increased synthesis of smooth muscle proteins and an increased
hepatectomy have been very useful for defining the mechanisms that
cell size. The bulging muscles of bodybuilders engaged in “pumping
stimulate regeneration of the liver (Chapter 3). The bone marrow is also
iron” result from enlargement of individual skeletal muscle fibers in
remarkable in its capacity to undergo rapid hyperplasia in response to a
response to increased demand.
deficiency of mature blood cells. For example, in the setting of an acute
bleeding or premature breakdown of red blood cells (hemolysis),
Mechanisms of Hypertrophy
feedback loops involving the growth factor erythropoietin are activated
 Hypertrophy is a result of increased cellular protein production. Much of
that stimulate the growth of red blood cell progenitors, allowing red blood
our understanding of hypertrophy is based on studies of the heart. There
cell production to increase as much as eightfold. The regulation of
is great interest in defining the molecular basis of myocardial hypertrophy
hematopoiesis is discussed further in Chapter 13.
 Pathologic hyperplasia. Most forms of pathologic hyperplasia are caused disease results in tissue atrophy. In late adult life, the brain may undergo
by excessive or inappropriate actions of hormones or growth factors progressive atrophy, mainly because of reduced blood supply as a result
acting on target cells. Endometrial hyperplasia is an example of abnormal of atherosclerosis (Fig. 2.27). This is called senile atrophy.
hormone-induced hyperplasia. Normally, after a menstrual period there is  Inadequate nutrition. Profound protein-calorie malnutrition (marasmus) is
a rapid burst of proliferative activity in the endometrium that is stimulated associated with the utilization of skeletal muscle proteins as a source of
by pituitary hormones and ovarian estrogen. It is brought to a halt by the energy after other reserves such as adipose stores have been depleted.
rising levels of progesterone, usually about 10 to 14 days before the end This results in marked muscle wasting (cachexia; Chapter 9). Cachexia is
of the menstrual period. In some instances, however, the balance also seen in patients with chronic inflammatory diseases and cancer. In
between estrogen and progesterone is disturbed, resulting in absolute or some cachectic states, chronic overproduction of the inflammatory
relative increases in the amount of estrogen, with consequent hyperplasia cytokine TNF is thought to be responsible for appetite suppression and
of the endometrial glands. This form of pathologic hyperplasia is a lipid depletion, culminating in muscle atrophy.
common cause of abnormal uterine bleeding. Benign prostatic  Loss of endocrine stimulation. Many hormone-responsive tissues, such as
hyperplasia is another common example of pathologic hyperplasia, in this the breast and reproductive organs, are dependent on endocrine
case as a response to hormonal stimulation by androgens. Although stimulation for normal metabolism and function. The loss of estrogen
these forms of pathologic hyperplasias are abnormal, the process stimulation after menopause results in atrophy of the endometrium,
remains controlled and the hyperplasia can either regress or stabilize if vaginal epithelium, and breast. Similarly, the prostrate atrophies following
the hormonal stimulation is eliminated. As is discussed in Chapter 7, the chemical or surgical castration (e.g., for treatment of prostate cancer).
increased cell division associated with hyperplasia elevates the risk of  Pressure. Tissue compression for any length of time can cause atrophy.
acquiring genetic aberrations that drive unrestrained proliferation and An enlarging benign tumor can cause atrophy in the surrounding
cancer. Thus, although hyperplasia is distinct from cancer, pathologic uninvolved tissues. Atrophy in this setting is probably the result of
hyperplasia constitutes a fertile soil in which cancerous proliferations may ischemic changes caused by compromise of the blood supply by the
eventually arise. For instance, patients with hyperplasia of the pressure exerted by the expanding mass. The fundamental cellular
endometrium are at increased risk for developing endometrial cancer changes associated with atrophy are similar in all of these settings. The
(Chapter 22). Hyperplasia is a characteristic response to certain viral initial response is a decrease in cell size and organelles, which may
infections, such as papillomaviruses, which cause skin warts and several reduce the metabolic needs of the cell sufficiently to permit its survival. In
mucosal lesions composed of masses of hyperplastic epithelium. Here, atrophic muscle, the cells contain fewer mitochondria and myofilaments
the viruses make factors that interfere with host proteins that regulate cell and a reduced amount of rough ER. By bringing into balance the cell’s
proliferation. Like other forms of hyperplasia, some of these virally metabolic demands and the lower levels of blood supply, nutrition, or
induced proliferations are also precursors to cancer (Chapter 7). trophic stimulation, a new equilibrium is achieved. Early in the process,
atrophic cells and tissues have diminished function, but cell death is
Mechanisms of Hyperplasia minimal. However, atrophy caused by gradually reduced blood supply
 Hyperplasia is the result of growth factor–driven proliferation of mature may progress to the point at which cells are irreversibly injured and die,
cells and, in some cases, by increased output of new cells from tissue often by apoptosis. Cell death by apoptosis also contributes to the atrophy
stem cells. For instance, after partial hepatectomy, growth factors are of endocrine organs after hormone withdrawal.
produced in the liver that engage receptors on the surviving cells and
activate signaling pathways that stimulate cell proliferation. But if the
proliferative capacity of the liver cells is compromised, as in some forms Mechanisms of Atrophy
of hepatitis causing cell injury, hepatocytes can instead regenerate from  Atrophy results from decreased protein synthesis and increased protein
intrahepatic stem cells. The roles of growth factors and stem cells in degradation in cells. Protein synthesis decreases because of reduced
cellular replication and tissue regeneration are discussed in more detail in trophic signals (e.g., those produced by growth receptors), which enhance
Chapter 3 uptake of nutrients and increase mRNA translation. The degradation of
cellular proteins occurs mainly by the ubiquitin-proteasome pathway.
Atrophy Nutrient deficiency and disuse may activate ubiquitin ligases, which attach
 Atrophy is a reduction in the size of an organ or tissue due to a decrease the small peptide ubiquitin to cellular proteins and target these proteins for
in cell size and number. Atrophy can be physiologic or pathologic. degradation in proteasomes. This pathway is also thought to be
Physiologic atrophy is common during normal development. Some responsible for the accelerated proteolysis seen in a variety of catabolic
embryonic structures, such as the notochord and thyroglossal duct, conditions, including cancer cachexia. In many situations, atrophy is also
undergo atrophy during fetal development. The decrease in the size of the accompanied by increased autophagy, marked by the appearance of
uterus that occurs shortly after parturition is another form of physiologic increased numbers of autophagic vacuoles. Some of the cell debris within
atrophy. Pathologic atrophy has several causes, and it can be local or the autophagic vacuoles may resist digestion and persist in the cytoplasm
generalized. Common causes of atrophy include the following: as membrane-bound residual bodies. An example of residual bodies is
 Decreased workload (disuse atrophy). When a fractured bone is lipofuscin granules, discussed later in the chapter. When present in
immobilized in a plaster cast or when a patient is restricted to complete sufficient amounts, they impart a brown discoloration to the tissue (brown
bed rest, skeletal muscle atrophy rapidly ensues. The initial decrease in atrophy). Autophagy is associated with various types of cell injury, as
cell size is reversible once activity is resumed. With more prolonged discussed earlier.
disuse, skeletal muscle fibers decrease in number (due to apoptosis) as
well as in size; muscle atrophy can be accompanied by increased bone
resorption, leading to osteoporosis of disuse. Metaplasia
 Loss of innervation (denervation atrophy). The normal metabolism and  Metaplasia is a reversible change in which one differentiated cell type
function of skeletal muscle are dependent on its nerve supply. Damage to (epithelial or mesenchymal) is replaced by another cell type. It often
the nerves leads to atrophy of the muscle fibers supplied by those nerves represents an adaptive response in which one cell type that is sensitive to
(Chapter 27). a particular stress is replaced by another cell type that is better able to
 Diminished blood supply. A gradual decrease in blood supply (chronic withstand the adverse environment. The most common epithelial
ischemia) to a tissue as a result of slowly developing arterial occlusive metaplasia is columnar to squamous (Fig. 2.28), as occurs in the
respiratory tract in response to chronic irritation. In the habitual cigarette  Atrophy: decreased cell and organ size, as a result of decreased nutrient
smoker, the normal ciliated columnar epithelial cells of the trachea and supply or disuse; associated with decreased synthesis of cellular building
bronchi are often replaced by stratified squamous epithelial cells. Vitamin blocks and increased breakdown of cellular organelles by increased
A (retinoic acid) deficiency can also induce squamous metaplasia in the autophagy
respiratory epithelium and in the cornea, the latter with highly deleterious  Metaplasia: change in phenotype of differentiated cells, often in response
effects on vision. Stones in the excretory ducts of the salivary glands, to chronic irritation, that makes cells better able to withstand the stress;
pancreas, or bile ducts, which are normally lined by secretory columnar usually induced by altered differentiation pathway of tissue stem cells;
epithelium, may also lead to squamous metaplasia. In all these instances, may result in reduced functions or increased propensity for malignant
the more rugged stratified squamous epithelium is able to survive under transformation
circumstances in which the more fragile specialized columnar epithelium
might have succumbed. However, the change to metaplastic squamous
cells comes with a price. In the respiratory tract, for example, although the INTRACELLULAR ACCUMULATIONS
epithelial lining becomes more durable, important mechanisms of  Lipids
protection against infection—mucus secretion and the ciliary action of the  Proteins
columnar epithelium—are lost. Thus, epithelial metaplasia, in most  Hyaline Change
circumstances, represents an undesirable change. Moreover, the  Gycogen
influences that predispose to metaplasia, if persistent, can initiate  Pigments
malignant transformation in metaplastic epithelium. The development of
squamous cell carcinoma in areas of the lungs where the normal
 One of the manifestations of metabolic derangements in cells is the
columnar epithelium has been replaced by squamous epithelium is one
intracellular accumulation of substances that may be harmless or cause
example. Metaplasia from squamous to columnar type may also occur, as
further injury.
in Barrett esophagus, in which the esophageal squamous epithelium is
 These accumulations may be located in the cytoplasm, within organelles
replaced by intestinal-like columnar cells under the influence of refluxed
(typically lysosomes), or in the nucleus, and they may be composed of
gastric acid. As might be expected, the cancers that arise in these areas
substances that are synthesized by the affected cells or are produced
are typically glandular (adenocarcinomas). Connective tissue metaplasia
elsewhere.
is the formation of cartilage, bone, or adipose cells (mesenchymal
 There are four main mechanisms leading to abnormal intracellular
tissues) in tissues that normally do not contain these elements. For
accumulations
example, bone formation in muscle, designated myositis ossificans,
o Inadequate removal of a normal substance secondary to defects in
occasionally occurs after intramuscular hemorrhage. This type of
metaplasia is less clearly seen as an adaptive response, and may be a packaging and transport – fatty change (steatosis) in the liver
result of cell or tissue injury. Unlike epithelial metaplasia, this type of o Accumulation of an endogenous substance as a result of genetic or
metaplasia is not associated with increased cancer risk. acquired defects in its folding, packaging, transport, or secretion –
mutated forms of α1-antitrypsin
o Failure to degrade a metabolite due to inherited enzyme
Mechanisms of Metaplasia deficiencies, typically lysosomal enzymes – lysosomal storage
 Metaplasia does not result from a change in the phenotype of an already diseases
differentiated cell type; rather, it results from either reprogramming of local o Deposition and accumulation of an abnormal exogenous substance
tissue stem cells or, alternatively, colonization by differentiated cell when the cell has neither the enzymatic machinery to degrade the
populations from adjacent sites. In either case, the metaplastic change is substance nor the ability to transport it to other sites – Accumulation
stimulated by signals generated by cytokines, growth factors, and of carbon or silica particles
extracellular matrix components in the cells’ environment. In the case of  In many cases, if the overload can be controlled or stopped, the
stem cell reprogramming, these external stimuli promote the expression accumulation is reversible. In inherited storage diseases, accumulation is
of genes that drive cells toward a specific differentiation pathway. A direct progressive and may cause cellular injury, leading in some instances to
link between transcription factor dysregulation and metaplasia is seen death of the tissue and the patient.
with vitamin A (retinoic acid) deficiency or excess, both of which may
cause metaplasia. Retinoic acid regulates gene transcription directly
through nuclear retinoid receptors, which can influence the differentiation
Lipids
of progenitors derived from tissue stem cells.
 All major classes of lipids can accumulate in cells: triglycerides,
CELLULAR ADAPTATIONS TO STRESS cholesterol/cholesterol esters, and phospholipids.
 Phospholipids are components of the myelin figures found in necrotic
 Hypertrophy: increased cell and organ size, often in response to
cells. In addition, abnormal complexes of lipids and carbohydrates
increased workload; induced by growth factors produced in response to
accumulate in the lysosomal storage diseases
mechanical stress or other stimuli; occurs in tissues incapable of cell
division. It may be physiologic (e.g., enlargement of the uterus in  Steatosis (Fatty Change)
pregnancy) or pathologic (e.g., enlargement of the heart in hypertension o abnormal accumulations of triglycerides within parenchymal cells.
of valvular disease). o Fatty change is seen in the liver (major organ involved in fat
 Hyperplasia: increased cell numbers in response to hormones and other metabolism), heart, muscle, and kidney.
growth factors; occurs in tissues whose cells are able to divide or contain o The causes of steatosis include toxins, protein malnutrition,
abundant tissue stem cells. It may be physiologic in response to diabetes mellitus, obesity, and anoxia.
increased need (e.g., breast acini during lactation) or pathologic in o In higher-income nations, the most common causes of significant
response to inappropriate secretion of hormones (e.g., endometrial fatty change in the liver (fatty liver) are alcohol abuse and
hyperplasia due to excessive estrogenic stimulation). nonalcoholic fatty liver disease, which is often associated with
diabetes and obesity.
 Cholesterol and Cholesterol Esters but also ER stress caused by the misfolded proteins, which initiates
o cellular metabolism of cholesterol is tightly regulated such that most the unfolded protein response and culminates in cell death by
cells use cholesterol for the synthesis of cell membranes without apoptosis (discussed earlier).
intracellular accumulation of cholesterol or cholesterol esters. o Accumulation of cytoskeletal proteins. There are several types of
o Accumulations manifested histologically by intracellular vacuoles cytoskeletal proteins, including microtubules (20 to 25 nm in
are seen in several pathologic processes. diameter), thin actin filaments (6 to 8 nm), thick myosin filaments (15
o Atherosclerosis. In atherosclerotic plaques, smooth muscle cells nm), and intermediate filaments (10 nm). Intermediate filaments,
and macrophages within the intimal layer of the aorta and large which provide a flexible intracellular scaffold that organizes the
arteries are filled with lipid vacuoles, most of which contain cytoplasm and resists forces applied to the cell, are divided into five
cholesterol and cholesterol esters. Such cells have a foamy classes: keratin filaments (characteristic of epithelial cells),
appearance (foam cells), and aggregates of them in the intima neurofilaments (neurons), desmin filaments (muscle cells), vimentin
produce the yellow cholesterol-laden atheromas characteristic of filaments (connective tissue cells), and glial filaments (astrocytes).
this serious disorder. Some of these fat-laden cells may rupture, Accumulations of keratin filaments and neurofilaments are
releasing cholesterol and cholesterol esters into the extracellular associated with certain types of cell injury. Alcoholic hyaline is an
space, where they may form crystals. eosinophilic cytoplasmic inclusion in liver cells that is characteristic
o Some form long needles that produce distinct clefts in tissue of alcoholic liver disease and is composed predominantly of keratin
sections, while other small crystals are phagocytosed by intermediate filaments. The neurofibrillary tangle found in the brain in
macrophages and activate the inflammasome, contributing to local Alzheimer disease contains neurofilaments and other proteins.
inflammation. o Aggregation of abnormal proteins. Abnormal or misfolded proteins
o Xanthomas. Intracellular accumulation of cholesterol within may deposit in tissues and interfere with normal functions. The
macrophages is also characteristic of acquired and hereditary deposits can be intracellular, extracellular, or both, and the
hyperlipidemic states. Clusters of foamy cells are found in the aggregates may either directly or indirectly cause the pathologic
subepithelial connective tissue of the skin and in tendons, changes. Certain forms of amyloidosis fall in this category of
producing tumorous masses known as xanthomas. diseases. These disorders are sometimes called proteinopathies or
o Cholesterolosis. This refers to the focal accumulations of protein-aggregation diseases.
cholesterol-laden macrophages in the lamina propria of the
gallbladder (Fig. 2.31). The mechanism of accumulation is
unknown.
o Niemann-Pick disease, type C. This lysosomal storage disease is Hyaline Change
caused by mutations affecting an enzyme involved in cholesterol  The term hyaline usually refers to an alteration within cells or in the
trafficking, resulting in cholesterol accumulation in multiple organs. extracellular space that gives a homogeneous, glassy, pink appearance in
routine histologic sections stained with H&E.
 It is widely used as a descriptive histologic term rather than a specific
Proteins marker for cell injury.
 This morphologic change is produced by a variety of alterations and does
 Intracellular accumulations of proteins usually appear as rounded, not represent a specific pattern of accumulation.
eosinophilic droplets, vacuoles, or aggregates in the cytoplasm.  Intracellular hyaline accumulations of protein include reabsorption
 By electron microscopy, they can be amorphous, fibrillar, or crystalline in droplets,
appearance.  Russell bodies, and alcoholic hyaline. Extracellular hyaline has been
 In some disorders, such as certain forms of amyloidosis, abnormal more difficult to analyze. Collagenous fibers in old scars may appear
proteins deposit primarily in extracellular spaces hyalinized, but the biochemical basis of this change is not clear. In long-
 Excesses of proteins within the cells sufficient to cause morphologically standing hypertension and diabetes mellitus, the walls of arterioles,
visible accumulation have diverse causes. especially in the kidney, become hyalinized, resulting from extravasated
o Reabsorption droplets in proximal renal tubules are seen in renal plasma protein and deposition of basement membrane material.
diseases associated with protein loss in the urine (proteinuria). In the
kidney, small amounts of protein filtered through the glomerulus are Glycogen
normally reabsorbed by pinocytosis in the proximal tubule. In  Excessive intracellular deposits of glycogen are seen in patients with an
disorders with heavy protein leakage across the glomerular filter, abnormality in either glucose or glycogen metabolism.
there is increased reabsorption of the protein into vesicles, and the  Glycogen is a readily available source of glucose stored in the cytoplasm
protein appears as pink hyaline droplets within the cytoplasm of the of healthy cells.
tubular cell (Fig. 2.32). The process is reversible; if the proteinuria  Whatever the clinical setting, the glycogen masses appear as clear
diminishes, the protein droplets are metabolized and disappear. vacuoles within the cytoplasm because glycogen dissolves in aqueous
o The proteins that accumulate may be normal secreted proteins that fixatives; thus, it is most readily identified when tissues are fixed in
are produced in excessive amounts and accumulate within the ER, absolute alcohol. Staining with Best carmine or the PAS reaction imparts
as occurs in certain plasma cells engaged in active synthesis of a rose-to-violet color to the glycogen, but can also stain protein-bound
immunoglobulins. The ER becomes hugely distended, producing carbohydrates.
large, homogeneous eosinophilic inclusions called Russell bodies. •  Diastase digestion of a parallel section that demonstrates loss of staining
Defective intracellular transport and secretion of critical proteins. In due to glycogen hydrolysis is therefore an important validation.
α1-antitrypsin deficiency, mutations in the protein significantly slow  Diabetes mellitus is the prime example of a disorder of glucose
folding, resulting in the buildup of partially folded intermediates, metabolism. In this disease, glycogen is found in renal tubular epithelial
which aggregate in the ER of the hepatocyte and are not secreted. cells, as well as within liver cells, β cells of the islets of Langerhans within
The resultant deficiency of the circulating enzyme where it is needed the pancreas, and heart muscle cells. Glycogen accumulates within select
in the lung causes emphysema. In many of these protein-folding cells in a group of related genetic disorders that are collectively referred to
diseases, the pathology results not only from loss of protein function
as the glycogen storage diseases, or glycogenoses. In these diseases, forms hemosiderin granules, which are easily seen with the light
enzymatic defects in the synthesis or breakdown of glycogen result in microscope. Hemosiderin pigment represents aggregates of ferritin
massive accumulation, causing cell injury and cell death. micelles. Under normal conditions, small amounts of hemosiderin
can be seen in the mononuclear phagocytes of the bone marrow,
spleen, and liver, which are responsible for recycling of iron derived
Pigments from hemoglobin during the breakdown of effete red blood cells.
 Pigments are colored substances, some of which are normal constituents Local or systemic excesses of iron cause hemosiderin to
of cells (e.g., melanin), whereas others are abnormal and accumulate in accumulate within cells. Local excesses result from hemorrhages in
cells under special circumstances. tissues. The best example of localized hemosiderosis is the
 exogenous – coming from outside the body common bruise. Extravasated red blood cells at the site of injury
 endogenous – synthesized within the body itself. are phagocytosed over several days by macrophages, which break
down the hemoglobin and recover the iron. After removal of iron,
 Exogenous Pigments the heme moiety is converted first to biliverdin (“green bile”) and
o most common exogenous pigment: carbon (coal dust), then to bilirubin (“red bile”). In parallel, the iron released from heme
o carbon is an ubiquitous air pollutant in urban areas. When inhaled, is incorporated into ferritin and eventually hemosiderin. These
conversions account for the often dramatic play of colors seen in a
it is picked up by macrophages within the alveoli and then
healing bruise, which typically changes from red-blue to green-blue
transported through lymphatic channels to lymph nodes in the
to golden-yellow before it is resolved. When there is systemic iron
tracheobronchial region.
overload, hemosiderin may be deposited in many organs and
o Accumulations of this pigment blacken the tissues of the lungs
tissues, a condition called hemosiderosis.
(anthracosis) and the involved lymph nodes.
o In coal miners, the aggregates of carbon dust may induce a
main causes of hemosiderosis
fibroblastic reaction or even emphysema, and thus cause a serious
 increased absorption of dietary iron due to an inborn error of metabolism
lung disease known as coal worker’s pneumoconiosis.
called hemochromatosis
o Tattooing is a form of localized, exogenous pigmentation of the
 hemolytic anemias, in which excessive lysis of red blood cells leads to
skin. The pigments inoculated are phagocytosed by dermal
release of abnormal quantities of iron,
macrophages, in which they reside for the remainder of the life of
 repeated blood transfusions, because transfused red blood cells
the embellished. The pigments do not usually evoke any
constitute an exogenous iron load
inflammatory response.

 Endogenous Pigments PATHOLOGIC CALCIFICATION


o Lipofuscin  Dystrophic Calcification
 AKA: lipochrome or wear-and-tear pigment  Metastatic Calcification
 insoluble pigment
 composed of polymers of lipids and phospholipids in complex PATHOLOGIC CALCIFICATION
with protein, suggesting that it is derived through lipid  abnormal tissue deposition of calcium salts, together with smaller
peroxidation of polyunsaturated lipids of intracellular amounts of iron, magnesium, and other mineral salts. There are two
membranes. forms of pathologic calcification
 Lipofuscin is not injurious to the cell or its functions.  dystrophic calcification
 Its importance lies in its being a telltale sign of free radical o deposition occurs locally in dying tissues
injury and lipid peroxidation.
o it occurs despite normal serum levels of calcium and in the absence
 The term is derived from the Latin (fuscus, brown), referring to
of derangements in calcium metabolism
brown lipid.
 metastatic calcification
 In tissue sections, it appears as a yellow-brown, finely granular
o deposition of calcium salts in normal tissues
cytoplasmic, often perinuclear, pigment (Fig. 2.33).
 It is seen in cells undergoing slow, regressive changes and is o results from hypercalcemia secondary to some disturbance in
particularly prominent in the liver and heart of aging patients or calcium metabolism.
patients with severe malnutrition and cancer cachexia.
 Melanin, derived from the Greek (melas, black), is an Dystrophic Calcification
endogenous, brown-black, pigment formed when the enzyme  encountered in areas of necrosis, whether they are of coagulative,
tyrosinase catalyzes the oxidation of tyrosine to caseous, or liquefactive type, and in foci of enzymatic necrosis of fat.
dihydroxyphenylalanine in melanocytes. Calcification is almost always present in the atheromas of advanced
o For practical purposes, melanin is the only endogenous brown- atherosclerosis.
black pigment. The only other that could be considered in this  It also commonly develops in aging or damaged heart valves, further
category is homogentisic acid, a black pigment that occurs in hampering their function.
patients with alkaptonuria, a rare metabolic disease.  Whatever the site of deposition, the calcium salts appear macroscopically
o Here the pigment is deposited in the skin, connective tissue, and as fine, white granules or clumps, often felt as gritty deposits.
cartilage, and the pigmentation is known as ochronosis.  Sometimes a tuberculous lymph node is virtually converted to stone.
o Hemosiderin, a hemoglobin-derived, golden yellowto-brown,  may simply be a telltale sign of previous cell injury but it is often a cause
granular, or crystalline pigment is one of the major storage forms of of organ dysfunction. Such is the case in calcific valvular disease and
iron. Iron is normally carried by a specific transport protein called atherosclerosis
transferrin. In cells, it is stored in association with a protein,  Serum calcium is normal
apoferritin, to form ferritin micelles. Ferritin is a constituent of most
cell types. When there is a local or systemic excess of iron, ferritin
MORPHOLOGY  Cholesterol deposition: Result of defective catabolism and excessive
 Histologically, with the usual H&E stain, calcium salts have a basophilic, intake; in macrophages and smooth muscle cells of vessel walls in
amorphous granular, sometimes clumped appearance. atherosclerosis
 They can be intracellular, extracellular, or in both locations.  Deposition of proteins: Reabsorbed proteins in kidney tubules;
 In the course of time, heterotopic bone may form in the focus of immunoglobulins in plasma cells
calcification.  Deposition of glycogen: In macrophages of patients with defects in
 On occasion, single necrotic cells may constitute seed crystals that lysosomal enzymes that break down glycogen (glycogen storage
become encrusted by the mineral deposits. The progressive acquisition of diseases) and in certain disorders of glycogen metabolism
outer layers may create lamellated configurations, called psammoma  Deposition of pigments: Typically indigestible pigments,such as carbon,
bodies. lipofuscin (breakdown product of lipid peroxidation), or iron (usually due to
 Some types of papillary cancers (e.g., thyroid) are apt to develop overload, as in hemosiderosis)
psammoma bodies. In asbestosis, calcium and iron salts gather about  Pathologic calcifications
long slender spicules of asbestos in the lung, creating exotic, beaded o Dystrophic calcification: Deposition of calcium at sites of cell injury
dumbbell forms known as asbestos bodies. and necrosis
o Metastatic calcification:Deposition of calcium in normal tissues,
caused by hypercalcemia (usually a consequence of parathyroid
Metastatic Calcification hormone excess)
 may occur in normal tissues whenever there is hypercalcemia.
Hypercalcemia also accentuates dystrophic calcification. CELLULAR AGING
 causes of hypercalcemia:  DNA Damage
o increased secretion of PTH with subsequent bone resorption
 Cellular Senescence
 hyperparathyroidism due to parathyroid tumors
 ectopic secretion of PTH-related protein by malignant tumors;  Defective Protein Homeostasis
o resorption of bone tissue o Telomere attrition
 primary tumors of bone marrow: multiple myeloma, leukemia o Activation of tumor suppressor genes
 diffuse skeletal metastasis: breast cancer  Dysregulated Nutrient Sensing
 accelerated bone turnover: Paget disease
 immobilization CELLULAR AGING
o vitamin D–related disorders, including vitamin D intoxication,  Individuals age because their cells age
sarcoidosis (in which macrophages activate a vitamin D precursor),  Age is one of the strongest independent risk factors for many chronic
and idiopathic hypercalcemia of infancy (Williams syndrome), diseases, such as cancer, Alzheimer disease, and ischemic heart disease
characterized by abnormal sensitivity to vitamin D; and  Cellular aging is not simply a consequence of cells “running out of steam,”
o renal failure, which causes retention of phosphate, leading to but in fact is regulated by genes that are evolutionarily conserved from
secondary hyperparathyroidism. yeast to worms to mammals.
o Less common causes include aluminum intoxication, which occurs  Cellular aging is the result of a progressive decline in cellular function and
in patients on chronic renal dialysis, and milk-alkali syndrome, viability caused by genetic abnormalities and the accumulation of cellular
which is due to excessive ingestion of calcium and absorbable and molecular damage due to the effects of exposure to exogenous
antacids such as milk or calcium carbonate. influences
 Metastatic calcification may occur widely throughout the body but  Aging is influenced by a limited number of genes, and genetic anomalies
principally affects the interstitial tissues of the gastric mucosa, kidneys, underlie syndromes resembling premature aging in humans as well
lungs, systemic arteries, and pulmonary veins.  Aging is associated with definable mechanistic alterations
 Although quite different in location, all of these tissues excrete acid and  Several mechanisms, some cell intrinsic and others environmentally
therefore have an internal alkaline compartment that predisposes them to induced, are believed to play a role in aging
metastatic calcification.
 In all of these sites, the calcium salts morphologically resemble those
described in dystrophic calcification. Thus, they may occur as
noncrystalline amorphous deposits or, at other times, as hydroxyapatite DNA Damage.
crystals.  nuclear and mitochondrial DNA damage
 Usually the mineral salts cause no clinical dysfunction, but on occasion  cause:
massive involvement of the lungs produces remarkable x-ray images and o exogenous agents: physical, chemical, and biologic
respiratory compromise. Massive deposits in the kidney o endogenous factors: ROS
(nephrocalcinosis) may in time cause renal damage.  most are repaired by DNA repair enzymes, some persists and
accumulates as cells age
o DNA repair slow down aging process
ABNORMAL INTRACELLULAR DEPOSITIONS AND CALCIFICATIONS
o average hematopoietic stem cell suffers 14 new mutations per year,
Abnormal deposits of materials in cells and tissues are the result of excessive
and it is likely that this accumulating damage explains why, like
intake or defective transport or catabolism.
most cancers, the most common hematologic malignancies are
Deposition of lipids
diseases of the aged
 Fatty change: Accumulation of free triglycerides in cells, resulting from
 Werner syndrome
excessive intake or defective transport (often because of defects in
o premature aging
synthesis of transport proteins); manifestation of reversible cell injury
o defective DNA helicase
 for DNA replication and repair and DNA unwinding.
 rapid accumulation of chromosomal damage that may mimic chaperones of the heat shock protein family age rapidly, and conversely,
some aspects of the injury that normally accumulates during those that overexpress such chaperones are long-lived. Similar data exist
cellular aging. for the role of autophagy and proteasomal degradation of proteins. Of
 Bloom syndrome and ataxia-telangiectasia interest, administration of rapamycin, which inhibits the mTOR (molecular
o aging at an increased rate target of rapamycin) pathway, increases the life span of middle-aged
o Genetic instability in somatic cells mice. Rapamycin has multiple effects, including promotion of autophagy.
o mutated genes encode proteins involved in repairing double-strand Abnormal protein homeostasis can have many effects on cell survival,
breaks in DNA replication, and functions. In addition, it may lead to accumulation of
misfolded proteins, which can trigger apoptosis

Cellular Senescence.
 All normal cells have a limited capacity for replication, and after a fixed Dysregulated Nutrient Sensing.
number of divisions cells become arrested in a terminally nondividing  Caloric restriction increases life span
state, known as replicative senescence.  Insulin and insulin-like growth factor 1 (IGF-1) signaling pathway.
 Aging is associated with progressive replicative senescence of cells. o IGF-1 is produced in many cell types in response to growth
 Cells from children have the capacity to undergo more rounds of hormone secretion by the pituitary gland.
replication than do cells from older people. o IGF-1 mimics intracellular signaling by insulin and thereby informs
 Two mechanisms: cells of the availability of glucose, promoting an anabolic state as
o Telomere attrition well as cell growth and replication.
 progressive shortening of telomeres, which ultimately results o IGF-1 signaling has multiple downstream targets; relevant to this
in cell cycle arrest discussion are two kinases: AKT and its downstream target, mTOR,
 Telomeres are short repeated sequences of DNA present at which, as the name implies, is inhibited by rapamycin.
the ends of linear chromosomes that are important for  Sirtuins.
ensuring the complete replication of chromosome ends and for o Sirtuins are a family of NAD-dependent protein deacetylases. There
protecting the ends from fusion and degradation. are at least seven types of sirtuins in mammals that are distributed
 When somatic cells replicate, a small section of the telomere in different cellular compartments and have nonredundant functions
is not duplicated and telomeres become progressively designed to adapt bodily functions to various environmental
shortened. As the telomeres become shorter, the ends of stresses, including food deprivation and DNA damage.
chromosomes cannot be protected and are seen as broken o Sirtuins are thought to promote the expression of several genes
DNA, which signals cell cycle arrest. whose products increase longevity. These include proteins that
 Telomere length is maintained by nucleotide addition mediated inhibit metabolic activity, reduce apoptosis, stimulate protein
by an enzyme called telomerase. folding, and counteract the harmful effects of oxygen free radicals.
 Telomerase is a specialized RNA-protein complex that uses its o Sirtuins also increase insulin sensitivity and glucose metabolism,
own RNA as a template for adding nucleotides to the ends of and may be targets for the treatment of diabetes.
chromosomes. o It is thought that caloric restriction increases longevity both by
 Telomerase is expressed in germ cells and is present at low reducing the signaling intensity of the IGF-1 pathway and by
levels in stem cells, but it is absent in most somatic tissues increasing sirtuins. Attenuation of IGF-1 signaling leads to lower
(Fig. 2.36) rates of cell growth and metabolism and possibly reduced cellular
 Therefore, as most somatic cells age, their telomeres become damage.
shorter and they exit the cell cycle, resulting in an inability to o This effect can be mimicked by rapamycin. An increase in sirtuins,
generate new cells to replace damaged ones. particularly sirtuin-6, serves dual functions: the sirtuins
 In immortalized cancer cells, telomerase is usually reactivated  contribute to metabolic adaptations of caloric restriction and
and telomere length is stabilized, allowing the cells to  promote genomic integrity by activating DNA repair enzymes
proliferate indefinitely. through deacylation.
 telomere shortening is also associated with premature o Although the anti-aging effects of sirtuins have been widely
development of diseases, such as pulmonary fibrosis and publicized, much remains to be known before sirtuin-activating pills
aplastic anemia. will be available to increase longevity. Nevertheless, optimistic wine
lovers have been delighted to hear that a constituent of red wine
o Activation of tumor suppressor genes. may activate sirtuins and thus increase life span!
 CDKN2A locus encodes two tumor suppressor proteins,
expression of one of which, known as p16 or INK4a, is  The various forms of cellular derangements and adaptations described in
correlated with chronologic age this chapter cover a wide spectrum, which includes adaptations in cell
 By controlling G1- to S-phase progression during the cell size, growth, and function; reversible and irreversible forms of acute cell
cycle, p16 protects cells from uncontrolled mitogenic signals injury; regulated cell death (e.g., in apoptosis); pathologic alterations in
and pushes cells along the senescence pathway. cell organelles; and less ominous forms of intracellular accumulations,
including pigmentations. Reference is made to all of these alterations
Defective Protein Homeostasis throughout this book, because all organ injury and ultimately all clinical
two mechanisms: disease arise from derangements in cell structure and function.
 maintenance of proteins in their correctly folded conformations (mediated
by chaperones) and degradation of misfolded, damaged, or unneeded
proteins by the autophagy-lysosome system and ubiquitin-proteasome CELLULAR AGING
system. There is evidence that both normal folding and degradation of  Cellular aging results from a combination of accumulating cellular damage
misfolded proteins are impaired with aging. Mutant mice deficient in (e.g., by free radicals), reduced capacity to divide (replicative
senescence), reduced ability to repair damaged DNA, and defective
protein homeostasis.
 Accumulation of DNA damage: Defective DNA repair mechanisms;
conversely, caloric restriction may activate DNA repair and slow aging in
model organisms
 Replicative senescence: Reduced capacity of cells to divide secondary to
progressive shortening of chromosomal ends (telomeres)
 Defective protein homeostasis: Resulting from impaired chaperone and
proteasome functions.
 Nutrient sensing system: Caloric restriction increases longevity. Mediators
may be reduced IGF-1 signaling and increased sirtuins.

You might also like