Robbins Pathology - Chapter 2 Trans

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

CHAPTER 2: CELLULAR RESPONSES TO STRESS AND TOXIC Functional Derangements and Clinical Manifestations

INSULTS: ADAPTATION, INJURY, AND DEATH  functional consequences of these changes


 Functional abnormalities – the end results of genetic, biochemical, and structural
INTRODUCTION TO PATHOLOGY changes in cells and tissues are functional abnormalities
o Lead to clinical manifestations (symptoms and signs) of disease, as well
as its progress (clinical course and outcome)
 Pathology – devoted to the study of the structural, biochemical, and functional
 Clinicopathologic correlations are very important in the study of the disease
changes in cells, tissues, and organs that underlie disease  All forms of disease start with molecular or structural alterations in cells
o Explain whys and whereof of the signs and symptoms manifested by  Rudolph Virchow – father of modern pathology; developed the concept of cellular
patients; provide rational basis for clinical care and therapy basis of disease
o Serves as the bridge between the basic sciences and clinical medicine  Cell and ECM injury leads to tissue and organ injury which determines the
o Scientific foundation for all of medicine morphologic and clinical patterns of disease
 General pathology – concerned with the common reactions of cells and tissues to
injurious stimuli
OVERVIEW: CELLULAR RESPONSE TO STRESS AND NOXIOUS STIMULI
o Such reactions are not tissue specific (thus acute inflammation is response
to bacterial infections produces same reactions in most tissues)
 Normal cell is confined to narrow range of function and structure by its state of
 Systemic pathology – examines the alterations and underlying mechanisms in
metabolism, differentiation, and specialization; by constraints of neighboring cells; and
organ specific diseases such as ischemic heart disease
by the availability of metabolic substrates
 Four aspects of disease process that form the core of pathology:
o Homeostasis – steady state
 Adaptations – reversible functional and structural responses to changes in
Etiology or Cause
 Cause of the disease physiologic states (e.g., pregnancy) and some pathologic stimuli (new and altered
 Can be grouped into two classes: steady states are achieved to allow the cell survival and functioning)
 Genetic – inherited mutations, disease-associated gene variants, polymorphisms  Adaptive response may consist of:
 Acquired – infectious, nutritional, chemical, physical o Hypertrophy – increase in size and function
 The idea that one etiologic agent is the cause of a disease is from the study of o Hyperplasia – increase in number of cells
infections and inherited disorders caused by single genes o Atrophy – decrease in size and metabolic activity
o Idea is not applicable to majority of diseases (e.g., atherosclerosis and
o Metaplasia – change in the phenotype of cells
cancer  multifactorial and cause by various external triggers)
 When the stress is eliminated, the cell can recover to its original state without
consequences
Pathogenesis
 biochemical and molecular mechanisms of its development If the limits of adaptive responses are exceeded or if cells are exposed to
 Refers to the sequence of cellular, biochemical, and molecular events that injurious agents or stress, deprived of essential nutrients, or become
follow the exposure of cells or tissues to an injurious agent compromised by mutations that affect essential cellular constituents, a
o How mutations induced disease sequence of events follows that is termed cell injury.
 Even when initial cause is known, it is many steps removed from expression of disease
o Example: to understand cystic fibrosis it is essential to know the defective  Cell injury is reversible upto certain point  but if the stimulus persists or is severe
gene product and the biochemical and morphological events leading to enough from the beginning  may lead to irreversible injury and ultimately cell
formation of cysts and fibrosis in lungs, pancreas, etc.
death
 Stages of progressive impairment following different types of insults:
o Adaptation
Morphologic Changes
 structural alterations induced in the cells and organs of the body o Reversible injury
 Refer to the structural alterations in cells or tissues that are either o Cell death
characteristic of a disease or diagnostic of an etiologic process  Example: ↑ hemodynamics  enlargement of heart muscle (adaptation)  may then
 Morphology – determine the nature of the disease and to follow its progression; become injured (if blood supply to myocardium is not enough, heart muscle suffers
diagnostic cornerstone reversible injury and eventually irreversible injury and cell death)
 Limitations: morphologically identical lesions may arise by distinct molecular  Cell death – the end result of progressive cell injury; one of the most crucial events
mechanisms
in the evolution of disease in any tissue or organ
o E.g., tumors  breast cancers that are indistinguishable morphologically
may have widely different courses, therapeutic responses, and prognosis o Results from diverse causes (ischemia, infection, toxins)
 Molecular analysis (e.g., next generation sequencing) – reveal genetic differences  Cell death is also a normal and essential process in embryogenesis, the development
that predict the behaviour of tumor and responses of organs, and the maintenance of homeostasis
abmrmtmd
 Two principal pathways of cell death:  Striated cells (heart and skeletal muscles) have limited capacity for division 
o Necrosis respond to increased metabolic demands mainly by undergoing hypertrophy
o Apoptosis o Stimulus for hypertrophy in heart is usually chronic hemodynamic overload
 Autophagy – adaptive cellular response triggered by nutrient deprivation; may also (may result from hypertension or faulty valves)
culminate in death o Cells synthesize more proteins and number of myofilaments increases 
 Metabolic derangements in cells and sublethal, chronic injury may be associated with increase strength and work capacity
intracellular accumulations of a number of substances, including proteins, lipids, and  Most common stimulus for hypertrophy of muscle is increased workload (e.g., body
carbohydrates builders)
 Calcium – often deposited at sites of cell death, resulting in pathologic calcification  Pregnancy  massive physiologic growth of uterus
 Aging is also accompanied by characteristic morphologic and functional changes in o Hormone-induced enlargement of organ mainly from hypertrophy of muscle
cells o Uterine hypertrophy is stimulated by estrogenic hormones acting on smooth
 Three other processes that affect cells and tissues: muscle  ↑ synthesis of smooth muscle proteins and an increase in cell size
o Intracellular accumulations
o Pathologic calcifications Mechanisms of Hypertrophy
o Cell aging
 Hypertrophy is the result of increased production of cellular proteins
 Hypertrophy of the heart may become mal-adaptive and can lead to heart failure,
arrhythmias, and sudden death.

Three basic steps in the molecular pathogenesis of cardiac hypertrophy:


 The actions of mechanical sensors (triggered by ↑ workload), growth factors
(TGF-B. IGF-1, FGF) and vasoactive agents (a-adrenergic agonist, endothelin
1, ANG II). Mechanical sensors induce production of growth factors and agonist.
 Signals from cell membrane activate a complex of signal transduction
pathways. Two biochemical pathways are involved in muscle hypertrophy
o PI3K/AKT pathway – most important in physiologic hypertrophy
o Signaling downstream of GPCRs – induced by many growth factors
and vasoactive agents; though to be more important in pathologic
hypertrophy
 Signalling pathways activate transcription factors such as GATA4, NFAT, and
MEF2. These transcription factors work co-ordinately to increase the synthesis of
muscle proteins that are responsible for hypertrophy

ADAPTATIONS OF CELLULAR GROWTH AND DIFFERENTIATION


 Hypertrophy is also asso. w/ a switch of contractile proteins from adult to fetal
 Adaptations – reversible changes in the size, number, phenotype metabolic neonatal forms
activity, or functions of cells in response to changes in their environment
o During muscle hypertrophy, the alpha isoform of heavy chain is replaced by
the beta isoform  has slower, more energetically economical contraction
HYPERTROPHY
 Some genes expressed only during early development are reexpressed in hypertrophic
cells, and the products participate in the cellular response to stress
 Hypertrophy refers to an increase in the size of the cells, that results in an
o E.g.,gene for ANF is expressed in both the atrium and the ventricle in the
increase in the size of the affected organ
embryonic heart. Cardiac hypertrophy is associated with ↑ ANF gene
o Organ has no new cells, only larger cells
o ANF – peptide hormone that causes salt secretion by the kidney, decreases
 Due to synthesis and assembly of additional intracellular structural components
blood volume and pressure  serves to reduce hemodynamic load
 Cells capable of division may respond to stress by undergoing both hyperplasia and
 Cardiac hypertrophy reaches a limit  enlargement is unable to cope with
hypertrophy
enlargement  regressive changes (lysis and loss of myofribrillar contractile
o Non dividing (e.g., myocardial fibers) increased tissue mass is due to
elements)  myocyte death  net result: cardiac failure (if stress is not relieved)
hypertrophy
o To prevent such consequences, several drugs that inhibit key signaling
o Hypertrophy and hyperplasia may coexist and contribute to increase size
pathways involving NFAT, GATA4, and MEF2 genes are in clinical trials
 Hypertrophy can be physiologic or pathologic
o Physiologic – caused by increased functional demand or by stimulation by
hormones and growth factors

abmrmtmd
HYPERPLASIA ATROPHY

 Hyperplasia is defined as an increase in the number of cells in an organ or  Atrophy is defined as a reduction in the size of an organ or tissue due to a
tissue in response to a stimulus decrease in cell size and number; physiologic or pathologic
 Hyperplasia can only take place if the tissue contains cells capable of
dividing  increase number of cells; can be physiologic or pathologic Physiologic Atrophy
 Common during normal development
Physiologic Hyperplasia  Example: notochord and thryoglossal duct  atrophy during fetal development
 Physiologic hyperplasia due to the action of hormones or growth factors occurs in  Uterus  decrease in size after parturition
several circumstances: Pathologic Atrophy
o When there is a need to increase functional capacity of hormone  Has several causes and can be: local or generalized
sensitive organs
o When there is need for compensatory increase after damage or
resection Common Causes of Atrophy
 Hormonal hyperplasia – eg, proliferation of glandular epithelium of female breast Decreased - Atrophy of disuse
and enlargement (hypertrophy) of glandular epithelial cells workload - Example: immobilized fractured bone in a cast or complete bed rest
 Compensatory hyperplasia – eg, liver regeneration (proliferation to its normal size) - Initial decrease in cell size is reversible once activity is resumed
 Marrow undergo rapid hyperplasia in response to a deficiency of terminally - Prolonged disuse  skeletal muscle fibers ↓ in size
differentiated blood cells - Muscle atrophy can be accompanied by ↑ bone resorption  lead to
o Example: acute bleed or hemolysis  EPO are activated to stimulate RBC osteoporosis of disuse
progenitors  ↑ RBC production upto 8x Loss of - Denervation atrophy
inervation - Normal metabolism and function is dependent on nerve supply
- Damage to nerves  atrophy of the muscle fibers supplied
Pathologic Hyperplasia Diminished - Ischemia (↓ blood supply; e.g., occlusion)
 Most forms of pathologic hyperplasia are caused by excessive or blood supply - Brain may go progressive atrophy in late adult life  due to ↓ blood
inappropriate actions of hormones or growth factors acting on target cells supply from atherosclerosis  senile atrophy (also affect heart)
 Endometrial hyperplasia – example of abnormal hormone-induced hyperplasia Inadequate - Marasmus (profound protein calorie malnutrition) – asso. w/
o Disturbed balance between estrogen and progesterone  ↑ estrogen  nutrition utilization of skeletal muscle proteins as a source of energy (other
hyperplasia of endometrial glands reserves like adipose tissue has already been depleted)  results in
o Common cause of abnormal menstrual bleeding cachexia (marked muscle wasting)
 Benign prostatic hyperplasia – pathologic hyperplasia induced by androgens - Cachexia – also seen in patients w/ chronic inflammatory disease
 These pathologic hyperplasia remains controlled; hyperplasia regresses if hormonal (CID) and cancer
stimulation is eliminated - In CID, TNF is overproduced and is responsible for appetite
 Pathologic hyperplasia constitutes a fertile soil for cancer suppression and lipid depletion  ends in muscle atrophy
 Hyperplasia is a characteristic response to certain viral infections Loss of - Hormone responsive tissue (e.g., breast and reproductive
o HPV – may cause skin warts & several mucosal lesions composed of endocrine organs) are dependent on endocrine stimulation for normal function
masses of hyperplastic epithelium stimulation - Loss of estrogen stimulation after menopause results in physiologic
atrophy of endometrium, vaginal epithelium, and breast
Pressure - Tissue compression can cause atrophy
Mechanisms of Hyperplasia - Enlarging benign tumor  atrophy in surrounding uninvolved tissue
- This atrophy is due to ischemic changes caused by compromise of
 Hyperplasia is the result of growth factor-driven proliferation of mature the blood supply by the pressure exerted by the expanding mass
cells and, in some cases, by increased output of new cells from tissue stem
 Fundamental cellular changes asso. w/ atrophy are identical in all of these settings
cells
o Initial response  decrease in cell size and organelles, which ↓ the
 Example: after partial hepatectomy growth factors are produced in the liver that
metabolic needs of the cell for its survival
engage receptors on surviving cells and activate cell proliferation
 In atropic muscle, cells contain fewer mitochondria and myofilaments and a reduced
o If proliferative capacity of liver is compromised (e.g., hepatitis) hepatocytes
amount of RER
can instead regenerate from intrahepatic stem cells
o New equilibrium is achieved  balance of metabolic demands, low levels of
blood supply, nutrition or trophic stimulation
 Atrophy caused by gradually reduced blood supply  progress to irreversible injury
and death of cells (apoptosis)
o cell death by apoptosis contributes to the atrophy of endocrine organs after
hormonal withdrawal
abmrmtmd
Mechanism of Atrophy Mechanisms of Metaplasia

 Atrophy results from decreased protein synthesis and increased protein  Metaplasia does not result from a change in the phenotype of an already
degradation in cells differentiated cell type; instead it is the result of a reprogramming of stem
o Protein synthesis decreases because of reduced metabolic activity cells that are known to exist in normal tissues, or of undifferentiated
 The degradation of cellular proteins occurs mainly by the ubiquitin- mesenchymal xells present in connective tissue
proteasome pathway o Precursor cells differentiate in a new pathway or lineage by signals from
o Nutrient deficiency and disuse  activate ubiquitin ligase  attach ubiquitin cytokines, growth factors, and ECM components in the cell’s environment
to cellular proteins  target proteins for degradation in proteasome  Direct link between transcription factor dysregulation and metaplasia is seen with
o This pathway is responsible for the accelerated proteolysis in catabolic vitamin A (retinoic acid) deficiency or excess (both may cause metaplasia)
conditions (e.g., cancer cachexia) o Retinoic acid regulates gene transcription directly through nuclear retinoid
 Atrophy is also accompany by autophagy, marked by appearance of ↑ #s of receptors  influence differentiation of progenitors derived from tissue
autophagic vacuoles stem cells
o Autophagy (“self-eating”) – process where starved cells eat its own
components to reduced nutrient demand to match the supply KEY CONCEPTS: Cellular Adaptation to Stress
o Some cell debris resist digestion and seen in cytoplasm as membrane-bound Hypertrophy - Increased cell and organ size in response to increased workload
residual bodies (e.g., lipofuscin granules  brown atrophy) - Induced by growth factors produced in response to mechanical
stress or other stimuli
- Occurs in tissues incapable of cell division
METAPLASIA
Hyperplasia - Increased cell numbers in response to hormones ad ther growth
factors
 Metaplasia is reversible change in which one differentiated cell type - Occurs in tissues whose cells are able to divide or contain
(epithelial or mesenchymal) is replaced by another cell type abundant tissue stem cells
 Often represents as an adaptive response  “one cell type that is sensitive to a Atrophy - Decreased cell and organ size as a result of decreased nutrient
particular stress is replaced by another cell type that is better able to withstand the supply or disuse
adverse environment” - Asso. w/ decreased synthesis of cellular building blocks and
 Most common epithelial metaplasia is columnar to squamous (occurs in respiratory increased breakdown of cellular organelles
Metaplasia - Change in phenotype of differentiated cells, often in response to
tract in response to chronic irritation)
chronic irritation, that makes cells better able to withstand the
o Habitual cigar smoker’s trachea and bronci: ciliated columnar is replaced by stress
stratified squamous cells - Usually induced by altered differentiation pathway of tissue stem
o Secretory columnar epithelium (salivary gland, pancreas, bile duct) is cells
replaced by stratified squamous epithelium (squamous metaplasia) - May result in reduced functions or increased propensity for
o Vitamin A deficiency induces squamous metaplasia in the respiratory malignant transformation
epithelium
 Stratified squamous is able to survive under circumstances than columnar cells
o However, important mechanisms of protection against infection are lost
(e.g., mucus secretion and ciliary action)
 Epithelial metaplasia is “double-edged sword”
 The influences that predispose to metaplasia, if persistent, can initiate malignant
transformation in metaplastic epithelium (common cause of cancer in respiratory tract
is compose of squamous cells)
 Metaplasia from squamous to columnar type also occur in Barrett esophagus 
squamous is replaced by columnar cells under the influence of refluxed gastric acid
o Cancer may arise in this areas and are typically glandular
 Connective tissue metaplasia – formation of cartilage, bone, or adipose tissue
(mesenchymal tissues) in tissues that do not contain these elements
o Example: myositis ossificans – bone formation in muscle; occur after
intramuscular hemorrhage; seen as adaptive response from cell or tissue
injury

abmrmtmd
OVERVIEW OF CELL INJURY AND DEATH CAUSES OF CELL INJURY

 Cell injury results when cells are severely stressed and no longer able to adapt or  May range from physical trauma to subtle cellular abnormalities such as mutation
when cells are exposed to inherently damaging agents or from intrinsic abnormalities (e.g., lack of enzyme that impairs normal function)
 Injury may progress through a reversible stage and culminate in cell death  Most injurious stimuli can be grouped into the following broad categories:

Reversible Cell Injury - Hypoxia – deficiency of oxygen; causes cell injury by


 Changes are reversible in early or mild forms if damaging stimulus are removed reducing aerobic oxidative respiration
 Hallmarks of reversible injury: - Hypoxia is an extremely important and common cause of
o Reduced oxidative phosphorylation with resultant depletion of energy stored cell injury and cell death
in the form of ATP and; Oxygen Deprivation - Causes of hypoxia:
o Cellular swelling caused by changes in ion concentration and water influx o Ischemia (reduced blood flow)
 Intracellular organelles (mitochondria and cytoskeleton) may show alterations o Inadequate oxygenation of the blood due to
cardiorespiratory failure
o Decreased oxygen-carrying capacity of blood (as
in anemia or carbon monoxide poisoning or after
severe blood loss)
- Trauma, extremes of temperature (burns and deep cold),
Cell Death
Physical agents sudden changes in atmospheric pressure, radiation,
 Injury becomes irreversible with continuous damage  cell cannot recover and dies
electric shock
 Two principal types of cell death: necrosis and apoptosis
- Simple chemicals (glucose or salt) in hypertonic
concentrations may cause cell injury directly or by
- “accidental” and unregulated form of cell death due to damage
deranging electrolyte balance in cells
to cell membranes and loss of ion hemostasis
- Oxygen at high concentration is toxic
- When membrane damage is severe  lysosomal enzymes
Chemical agents and - Poisons (arsenic, cyanide or mercuric salts) in trace
enter the cytoplasma and digest the cell  give rise to
drugs amounts may damage cells to cause death
Necrosis morphologic changes or necrosis
- Daily companions: environmental and air pollutants,
- Cellular contents leak through the damaged plasma membrane
insecticides, herbicides
into extracellular space where they elicit reaction 
- Industrial/Occupational hazards: carbon monoxide,
inflammation
asbestos
- Necrosis is the pathway of cell death in many commonly
- Recreational drugs (alcohol) and ever-increasing variety of
encountered injuries (e.g., resulting from ischemia, exposure
therapeutic drugs
to toxins, various infections, trauma)
Infectious agents - Range from viruses to tapeworms
- When the cell’s DNA or proteins are damaged beyond repair,
- Rickettsiae, bacteria, fungi, and parasites
cell kill itself by apoptosis
Immunologic - Immune system serves an essential function in defense
- Characterized by:
reactions against pathogens but may also cause cell injury
Apoptosis o Nuclear dissolution
- Autoimmune diseases
o Fragmentation of cell w/o complete loss of
- Genetic defects may cause cell injury due to deficiency of
membrane integrity
functional proteins
o Rapid removal of the cellular debris
o Enzyme defects in inborn errors of metabolis
- Cellular contents do not leak out  no inflammatory
Genetic o Accumulation of damaged DNA or misfolded
reaction
derangements proteins
- Highly regulated process driven by a series of genetic
o Both trigger cell death when damage is beyond
pathways
repair
- “programmed cell death”
- Polymorphism (DNA sequence variants) can also influence
the susceptibility of cell to injury by chemicals and
 Whereas necrosis is always pathologic process, apoptosis serves many normal
environmental insults
functions and is not necessarily associated with cell injury
- Major cause of cell injury
 Necrosis in some cases is also a form of programmed cell death  necroptosis
Nutritional - Protein-calorie deficiencies cause number of deaths
imbalances (among under privileged populations)
- Can be self-imposed (anorexia nervosa)
- Nutritional excess also cause cell injury (atherosclerosis
and obesity)

abmrmtmd
MORPHOLOGIC ALTERATIONS IN CELL INJURY REVERSIBLE INJURY

 All stresses and noxious influences exert their effects first at the molecular or  Two features of reversible cell injury can be recognized under the light
biochemcical level microscope:
 There is a time lag between stress and morphologic changes of cell injury or death o Cellular swelling
o The duration of this delay may vary with the sensitivity of methods used to o Fatty change
detect changes  Cellular swelling – appears when cells are incapable of maintaining ionic
 Histochemical or Ultrastructural techniques  changes may be seen in minutes to homeostasis; result of failure of energy-dependent ion pumps in the plasma
hours after injury membrane
 Light microscopy or gross examination – changes take longer time (hours to days) to  Fatty change – occurs in hypoxic injury and various forms of toxic or metabolic
be seen injury
 Morphologic manifestations of necrosis take more time to develop than those or o Manifested by the appearance of lipid vacuoles in the cytoplasm
reversible damage (Example: ischemia  cell swelling occur in minutes  progress to o Seen mainly in cells involved in and dependent on fat metabolism, such as
irreversibility within an hour or two) hepatocytes and myocardial cells
 Light microscopic changes of cell death may not be seen until 4 to 12 hours after
onset of ischemia MORPHOLOGY
 FIGURE 2-8 (sequential changes in cell injury to cell death) Cellular swelling – first manifestation of almost all forms of injury to cells
 Difficult to see in LM; more apparent at the level of whole organ
Features of Necrosis and Apoptosis  When it affects many cells, it causes pallor, increased turgor, increased weight of the
organ; Swelling of cells is reversible
Feature Necrosis Apoptosis
 On microscopic exam: small clear vacuoles may be seen in cytoplasm  represent
Cell size Enlarged (swelling) Reduced (shrinkage)
distended and pinched-off segments of the ER
Nucleus Pyknosis  karyorrhexis  Fragmentation into nucleosome-size
o This pattern of non lethal injury is called hydropic change or vacuolar
karyolysis fragments
degeneration
Plasma Disrupted Intact; altered structure (orientation of  Cells may also increased eosinophilic staining  becomes more pronounced with
membrane lipids) progression to necrosis
 Ultractructural changes of reversible injury include:
Cellular Enzymatic digestion; may leak out Intact; may be released in apoptotic 1. Plasma membrane alterations  blebbing, blunting, and loss of microvilli
contents of cell bodies 2. Mitochondrial changes  including swelling anf the appearance of small
Adjacent Frequent No amorphous densities
inflammation 3. Dilation of the ER with detachment of polysomes; intracytoplasmic myelin
Physiologic or Invariably pathologic (culmination Often physiologic, means of figures may be present
pathologic of irreversible cell injury) eliminating unwanted cells; may be 4. Nuclear alterations  disaggregation of granular and fibrillar elements
role pathologic after injury (DNA damage)

 Reversible injury is characterized by: NECROSIS


o Generalized swelling of cell and organelles
o Blebbing of the plasma membrane  The morphologic appearance of necrosis as well as necroptosis is the result
o Detachment of ribosomes from the ER of denaturation of intracellular proteins and enzymatic digestion of the
o Clumping of nuclear chromatin lethally injured cell
 These morphologic changes are associated with:  Necrotic cells are unable to maintain membrane integrity and their contents leak out
o Decreased generation of ATP  cause inflammation
o Loss of cell membrane integrity  Enzymes that digest necrotic cells are derived from the lysosomes of the dying cells
o Defects in protein synthesis themselves and from the lysosomes of leukocytes
o Cytoskeletal damage  Digestion of cellular contents and response take hours to develop
o DNA damage o No detectable changes if myocardial infarct caused sudden death
 Persistent of excessive injury  “point of no return” ; irreversible injury and death o Earliest histologic evidence of myocardial necrosis does not become
 Severe mitochondrial damage with depletion of ATP and rupture of lysosomal and apparent until 4-12 hours later
plasma membranes are associated with necrosis  Because of the loss of plasma membrane integrity, cardiac-specific enzymes and
 Necrosis also occur in: proteins are rapidly released from necrotic muscle and can be detected in the blood as
o Ischemia; Exposure to toxins early as 2 hours after myocardial necrosis
o Various infections; Trauma

abmrmtmd
pancreatitis  pancreatic enzymes leak out of acinar cells and
liquefy the membranes of fat cells in the peritoneum  released
lipase split the triglyceride esters within fat cells
- Fatty acids combine with calcium to produce chalky-white areas
(fat saponification)
- Histologic exam: necrosis takes the form of foci shadowy outlines of
necrotic fat cells, with basophilic calcium deposits, surrounded
by an inflammatory reaction
Fibrinoid - Special form of necrosis usually seen in immune reactions involving
necrosis blood vessels
- Occurs when complexes of antigens and antibodies are deposited in
the wallsof arteries
- Deposits of “immune complexes” together with fibrin result in a
bright pink and amorphous appearance in H&E  called “fibrinoid”
Patterns of Tissue Necrosis (fibrin-like)
 Most necrotic cells disappear due to enzymatic digestion and phagocytosis by WBC
 When large number of cells die the tissue or organ is said to be necrotic  If not destroyed  they provide a nidus for deposition of calcium and become
 Necrosis of tissue has severe distinct morphological patterns  important to recognize calcified  dystrophic calcification
because they provide clues about the underlying cause
KEY CONCEPTS: Morphologic Alterations in Injured Cells and Tissues
Coagulative - Architecture of dead tissues is preserved for a span of some days  Reversible cell injury: cellular swelling, fatty change, PM blebbing and loss of
necrosis - Affected tissue exhibit a firm texture microvilli, mitochondrial swelling, ER dilation, eosinophilia (↓ cytoplasmic RNA)
- The injury denatures not only structural proteins but also enzymes  Necrosis: ↑ eosinophilia, nuclear shrinkage, fragmentation, dissolution; breakdown of
and so blocks the proteolysis of the dead cells  as a result, PM and organelle membranes; abundant myelin figures; leakage and enzymatic
eosinophilic, anucleate cells may persist for days or weeks digestion of cellular contents
- Ultimately the necrotic cells are removed by phagocytois of the  Pattern of tissue necrosis: under different conditions, necrosis in tissues may assume
cellular debris by infiltrating leukocytes and by digestion of dead specific patterns: coagulative, liquefactive, gangrenous, casseous, fat, fibrinoid
cells by lysosomal enzymes of leukocytes
- Ischemia caused by obstruction in a vessel may lead to coagulative
necrosis of the supplied tissue in all organs except the brain MECHANISMS OF CELL INJURY
- A localized area of coagulative necrosis is called an infarct
Liquefactive - Characterized by digestion of the dead cells  results in  Several principles that are relevant to most forms of cells injury:
necrosis transformation of the tissue into a liquid viscous mass
- Seen in focal bacterial or fungal infections because microbes
stimulate the accumulation of leukocytes and the liberation of  The cellular response to injurious stimuli depends on the nature of the
enzymes from these cells injury, its duration, and its severity
o Small doses of chemical toxin or brief period of ischemia may induce
Gangrenous - Not a specific pattern of cell death reversible injury
necrosis - Usually applied to a limb that has lost its blood supply and has  The consequence of cell injury depend on the type, state, and adaptability
undergone necrosis (coagulative necrosis) involving multiple tissue of the injured cell
planes
o Nutritional and hormonal status and metabolic needs  important in
- When bacterial infection is superimposed there is more liquefactive
necrosis because of the actions of degradative enzymes in the response to injury
bacteria and the attracted leukocytes  give rise to wet gangrene o Striated muscle in leg can be placed at rest and preserved when deprived of
Caseous - Encountered most often in foci of tuberculosis infection its blood supply; not so the striated muscle of the heart
necrosis - “caseous” (cheese-like) derived from the friable white appearance o Exposure of two individual cells to a toxin may have no effect in one cell
of the area of necrosis and cell death in another  due to polymorphism
- Microscopic exam: necrotic area appears as a structureless collection  Cell injury results from different biochemical mechanisms acting on several
of fragmented or lysed cells and amorphous granular debris
essential cellular components (Figure 2-16)
enclosed within a distinctive inflammatory border  this appearance
is characteristic of a focus of inflammation known as granuloma o Cellular components that are most frequently damaged by injurious stimuli
Fat necrosis - Does not denote a specific pattern of necrosis include: mitochondria, cell membranes, the machinery of protein synthesis
- Refers to focal areas of fat destruction, typically resulting from and packaging, and DNA
release of activated pancreatic lipase into pancreas and peritoneal o Any injurious stimulus may simultaneously trigger multiple interconnected
cavity mechanisms that damage cells  difficult to ascribe cell injury to a single
- Occurs in the calamitous abdominal emergency known as acute biochemical derangement
abmrmtmd
DEPLETION OF ATP MITOCHONDRIAL DAMAGE

 Reduction in ATP levels is fundamental cause of necrotic cell death  Mitochondria are critical players in cell injury and cell death by all pathways
 ATP depletion and decreased ATP synthesis  asso. w/ hypoxic and chemical (toxic) o They supply life-sustaining energy by producing ATP
injury  Mitochondria can be damaged by : (they are sensitive to hypoxia and toxins)
 ATP is produced in two ways: o Increases of cytosolic Ca2+
o Major pathway is oxidative phosphorylation of ADP  results in o Reactive oxygen species
reduction of oxygen by the electron transfer system of mitochondria o Oxygen deprivation
o Glycolytic pathway  can generate ATP in the absence of oxygen using  Mutations in mitochondrial genes are the cause of some inherited diseases
glucose derived from body fluids or from hydrolysis of glycogen  Three major consequence of mitochondrial damage:
 Major causes of ATP depletion are:
o Reduced supply of oxygen and nutrients  Formation of a high-conductance channel in the mitochondrial membrane called
o Mitochondrial damage mitochondrial permeability transition pore
o Actions of some toxins (e.g., cyanide) o Opening of channel leads to the loss of mitochondrial membrane potential
 High energy phosphate in the form of ATP is required for virtually all synthetis and  leads to failure of oxidative phosphorylation and progressive ↓ of ATP 
degradative processes within the cell (e.g., membrane transport, protein synthesis, ends in cell death
lipogenesis, deacylation-reacylation reaction for phospholipid turnover) o Cyclophilin D – one of the structural components of mitochondrial
 Depletion of ATP to 5%-10% of normal levels has widespread effects on many critical permeability transition pore (MPTP)
cellular systems:  Targeted by cyclosporine (immunsuppressive drug)
 Activity of the plasma membrane energy-dependent Na-pump is reduced  Cyclosporine reduces injury by preventing opening of the MPTP
o Failure of this active transport system causes Na to enter and  Abnormal oxidative phosphorylation also leads to the formation of ROS which have
accumulate inside cells and K to diffuse out (SIPO) many deleterious effect
o Net gain of solute = isosmotic gain of water  cell swelling and  Mitochondria produce several proteins capable of activating apoptotic pathways
dilation of ER o Includes cytochrome c and proteins that indirectly activate apoptosis-
 Cellular energy ,metabolism is altered inducing enzymes called caspases
o If supply of oxygen is reduced = oxidative phosphorylation ceases o Increased permeability of outer membrane results in leakage of these
 ↓ ATP and ↑ AMP proteins in the cytosol and death by apoptosis
o Changes stimulate phosphofructokinase and phosphorylase
activities  lead to ↑ anaerobic glycolysis (generate ATP from
glycogen)  rapid ↓ glycogen stores
o Anaerobic glycolysis causes accumulation of lactic acid and INFLUX OF CALCIUM AND LOSS OF CALCIUM HOMEOSTASIS
inorganic phosphates  ↓ intracellular pH  ↓ cellular enzymes
activity  Calcium ions are important mediators or cell injury
 Failure of Ca2+ pump leads to influx of Ca2+  has damaging effects  Depleting calcium protects cells from injury induced by harmful stimuli
 Prolonged depletion of ATP  structural disruption of the protein synthetic  Cytosolic calcium – maintained at 0.1umol
apparatus occurs  manifested as detachment of ribosomes and  Extracellular levels 0 1.3 mmol
dissociation of polysomes with reduction in protein synthesis  Most intracellular calcium is sequestered in mitochondria and ER
 In cells deprived of oxygen or glucose, proteins become misfolded  Ischemia and certain toxins cause an increase in cytosolic calcium concentration
o Accumulation of misfolded proteins in ER triggers a cellular o Initially due to release of Ca2+ from intracellular stores and later due to
reaction called “unfolded protein response” that may increased influx across the plasma membrane
accumulate in cell inury and even death  Increased intracellular Ca2+ causes cell injury by several mechanisms:
 Ultimately, there is irreversible damage to mitochondrial and lysosomal o Accumulation of Ca2+ in mitochondria  opens MPTP and fails production
membranes, and the cell undergoes necrosis of ATP
o ↑ cytosolic calcium activates enzymes with harmful effects on cells
 Phospholipase – cause membrane damage
 Proteases – breakdown both membrane and cytoskeletal proteins
 Endonuclease – responsible for DNA and chromatin fragmentation
 ATPase – hastening ATP depletion
o ↑ intracellular Ca2+ levels result in the induction of apoptosis by direct
activation of caspases and by increasing mitochondrial permeability
abmrmtmd
ACCUMULATION OF OXYGEN-DERIVED FREE RADICALS (OXIDATIVE STRESS)  Series of enzymes acts as free radical-scavenging systems (breaks down H2O2 and
O2-); these enzymes are located near the sites of generation of the oxidants and
 Cell injury induced by free radicals, particularly ROS, is an important includes:
mechanism of cell damage in many pathologic conditions, such as: o Catalase – present in peroxisomes; decomposes H2O2
o chemical and radiation injury, o Superoxide dismutase (SOD) – found in many cell types; convert O2- to
o ischemia-reperfusion injury (induced by restoration of blood flow in H2O2
ischemic tissue)  Includes manganese SODs (mitochondria)
o cellular aging and microbial killing phagocytes  Copper-zinc SODs (cytosol)
 Free radicals – chemical species that have single unpaired electron in outer orbit  Glutathione peroxidise – protects against injury by catalyzing free radical
o Unpaired electrons are highly reactive; “attack” and modify adjacent breakdown
molecules (inorganic or organic chemicals – CHON, CHO, Lipids, nucleic o Intracellular ration of oxidized glutathione (GSSG) to reduced glutathione
acids) (GSH) is a reflection of the oxidative state of the cell and is an important
o Some reactions are catalytic  molecules that react with free radicals are indicator of the cell’s ability to detoxify ROS
themselves converted to free radicals
 Reactive oxygen species (ROS) – type of oxygen-derived free radicals
o Produced normally inc ells during mitochondrial respiration and energy
generation  degraded and removed by cellular defense system
o Do not cause damage at low concentrations
o ↑ production or ↓ scavenging of ROS  cause oxidative stress
o Oxidative stress – cell injury, cancer, aging and degenerative diseases
o ROS are also produced by leukocytes (neutrophils and macrophage)

Generation of Free Radicals

Free radicals may be generated within cells in several ways

 The reduction-oxidation reactions that occur during normal metabolic processes Pathologic Effects of Free Radicals
 Absorption of radiant energy
 Rapid bursts of ROS are produced in activated leukocytes during inflammation Effects of ROS and other free radicals are wide-ranging. Three actions relevant to cell injury:
 Transition metals (iron and copper) donate or accept free electrons during intracellular
and catalyze free electrons during intracellular reactions and catalyze free radical  Lipid peroxidation in membranes
formation (Fenton Reaction) o Presence of O2  peroxidation of lipids
 Nitric Oxide – important chemical mediator generated by endothelial cells, o Oxidative damage is initiated when the double bonds in unsaturated fatty
macropahges, neurons and other cell types acids of membrane lipids are attacked by O2 derived free radicals (.OH)
o Act as free radical and can also be converted to highly reactive peroxynitrite o Lipid free radical interactions yield peroxides  unstable and reactive 
anion as well as NO2 and NO3 autocatalytic chain reactions ensues (called propagation) extensive
membrane damage
Removal of Free Radicals  Oxidative modification of proteins
o Free radicals promote oxidation of amino acid side chains, formation of
Free radicals are unstable and decay spontaneously. Mechanisms to remove free radicals and covalent protein-protein cross –lins and oxidation of the protein backbones
minimize injury: o Oxidative modification of proteins damages the active sites of enzymes,
disrupt the conformation of structural proteins, and enhance the
 Antioxidants – block or inactivate free radicals proteasomal degradation of unfolded or misfolded proteins
o Ex: vitamin E and A; ascorbic acid; glutathione in cytosol  Lesions in DNA
 Free iron and copper can catalyze formation of ROS o Free radicals are capable of causing single and double-strand breaks in
o Reactivity of these metals is minimized by their binding to storage and DNA, cross-linking of DNA strands. And formation of adducts
transport proteins (e.g., transferrin, lactoferrin, ferritin, ceruloplasmin)  o Oxidative DNA damage has been implicated in cell aging and in malignant
prevents these metals from participating in reactions that generate ROS transformation of cells

abmrmtmd
DEFECTS IN MEMBRANE PERMEABILITY Reversible vs. Irreversible Injury

 Early loss of selective membrane permeability, leading ultimately to overt  “point of no return” – damage becomes irreversible
membrane damage, is a consisitent feature of most forms of cell injury  Two phenomena consistently characterized irreversibility
(except apoptosis) o the inability to reverse mitochondrial dysfunction – lack of oxidative
 Mechanisms and pathologic consequences of membrane damage: phosphorylation and ATP generation
o profound disturbances in membrane function
Mechanisms of Membrane Damage  Injury to lysosomal membranes results in the enzymatic dissolution of the injured cell
that is characteristic of necrosis
In ischemic cells, membrane defects may be result of : ↓ ATP and calcium-mediated activation of  Leakage of intracellular proteins through the damage cell membrane and ultimately
phospholipases. Plasma membrane can also be damages by: bacterial and viral toxins, lytic into the circulation provides a means of detecting tissue-specific cellular injury and
complement components, physical and chemical agents. necrosis using serum samples

ROS - Cause injury to cell membranes by lipid peroxidation Mechanisms of Cell Injury
↓ Phospholipid - Consequence of defective mitochondrial function or hypoxia   ATP depletion: failure of energy-dependent functions  reversible injury  necrosis
Synthesis both decrease the production of ATP  Mitochondrial damage: ATP depletion  failure of energy-dependent cellular
- Affect cellular membranes and mitochondria functions  ultimately, necrosis; under some conditions, leakge of mitochondrial
↑ Phospholipid - Due to activation of Ca-dependent phospholipases by ↑ levels proteins that cause apoptosis
Breakdown of cytosolic and mitochondrial Ca2+  Influx of calcium: activation of enzymes that damage cellular components and may
- Phospholipid breakdown  lead to accumulation of lipid also trigger apoptosis
breakdown products (FFA, acyl carnitine, lysosphospholipids   Accumulation of ROS: covalent modification of cellular proteins, lipids, nucleic acids
detergent effect on membranes)  Increased permeability of cellular membranes: may affect plasma membrane,
- May also insert in lipid bilayer  changes in permeability and lysosomal membranes, mitochondrial membranes; typically culminates in necrosis
electrophysiologic alterations  Accumulation of damaged DNA and misfolded proteins: triggers apoptosis
Cytoskeletal - Activation of protease  damage in cytoskeleton
abnormalities
CLINICOPATHOLOGIC CORRELATIONS: SELECTED EXAMPLES OF CELL INJURY AND
Consequences of Membrane Damage NECROSIS

The most important part of membrane damage during cell injury are: mitochondrial membrane, Common and clinically significant forms of cell injury that culminate in necrosis:
plasma membrane, membranes of lysosomes.
ISCHEMIC AND HYPOXIC INJURY
Mitochondrial - Results in opening of MPTP  lead to ↓ ATP generation and
membrane damage release of proteins that trigger apoptotic death  Ischemia is the most common type of cell injury in clinical medicine and it
Plasma membrane - Results in loss of osmotic balance and influx of fluids and results from hypoxia induced by reduced blood flow, most commonly due to
damage ions, as well as loss of cellular components
a mechanical arterial obstruction (also venous drainage)
- Cells may also leak metabolites vital for reconstitution of ATP
furthering the depletion of energy stores  Anaerobic glycolysis continue and exhaust glycolytic substrates  stops anaerobic
Injury to lysosomal - Results in leakage of enzymes into cytoplasma energy generation
membranes - Activates the acid hydrolase in the acidic intracellular pH of  Glycolysis is inhibited by accumulation of metabolites that would be washed out by
injured cell flowing blood
- Lysosomes contain RNAses, DNAses, proteases,  Ischemia tends to cause more rapid and severe cell and tissue injury than does
phosphatises, glucosidases  activation leads to digestion of hypoxia in the absence of ischemia
proteins, RNA, DNA, glycogen and cells die by necrosis
 Ischemia – deficient blood supply
 Hypoxia – deficient oxygen supply
DAMAGE TO DNA AND PROTEINS
Mechanisms of Ischemic Cell Injury
 Cells have mechanisms that repair damage to DNA, but if DNA damage is
 ↓ oxygen tension in cells  ↓ oxidative phosphorylation; ↓ATP generation
too seveer to be corrected (e.g., after exposure to DNA damaging drugs,
 ↓ ATP  failure of Na-pump  K out; Na and water in  cell swelling
radiation, or oxidative stress), the cell initiates a suicide program that
 Influx of calcium  harmful effects  loss of glycogen and ↓ protein synthesis
results in death by apoptosis
 If oxygen is restored, all of these disturbances are reversible
 If ischemia persists, irreversible injury and necrosis ensue
abmrmtmd
 Irreversible injury is associated with: Activation of - Contribute to ischemia-reperfusion injury
o Severe swelling of mitochondria Complement - IgM  deposit in ischemic tissues and when blood flow is
o Extensive damage to plasma membrane (give rise to myelin figures) System resumed, complement proteins bind to the deposited antibodies
 activated and cause cell injury and inflammation
o Swelling of lysosomes
 Large, flocculent, amorphous densities develop in the mitochondrial matrix 
indications of irreversible injury in the myocardium; seen 30-40mins after ischemia
CHEMICAL (TOXIC) INJURY
 Massive influx of calcium then occurs if ischemic zone is reperfused
 Death is mainly by necrosis, but apoptosis also contributes  Chemical injury remains a frequent problem in clinical medicine and is a
o Apoptotic pathway is activated by the release of pro-apoptotic molecules major limitation to drug therapy
from leaky mitochondria
 Liver – frequent target of drug toxicity
 Cell’s components are degraded  widespread leakage of cellular enzymes in the o Toxic liver injury – most frequent reason for terminating drug use
extracellular space and there is entry of extracellular molecules from the interstitial  Chemicals induce cell injury by one of two general mechanisms
cells into the dying cells
 Finally, dead cells become replaced by large masses composed of phospholipids in the Direct Toxicity
form of myelin figures  Some chemicals can injure cells directly by combining with critical molecular
o Phagocytosed by WBC or degraded further into fatty acids components
o Calcification of fatty acids  calcium soaps  Ex: mercuric chloride poisoning  mercury binds with sulfhydryl groups  cause ↑
 Leakage on intracellular enzymes  important clinical indicators of cell death membrane permeability and inhibition of ion transport
 Hypoxia-inducible factor-1  promotes new blood vessels, stimulates cell survival o Greatest damage is usually to the cells that use, absorb, excrete, or
concentrate the chemicals (GIT and kidney)
pathways, and enhance anaerobic glycolysis
 Cyanide – poisons mitochondrial cytochrome oxidase  inhibits oxidative
phosphorylation
ISCHEMIA-REPERFUSION INJURY  Antineoplastic chemo agents and antibiotics  induce cell damge by direct cytotoxic
effects
 Restoration of blood flow to ischemic tissues can promote recovery of cells Conversion to Toxic Metabolites
if they are reversibly injured, but can also paradoxically exacerbate the  Most toxic chemicals are not biologically active in their native form
injury and cause cell death o Converted to reactive toxic metabolites and act on target molecules
 Ischemia-reperfusion injury – process where reperfused tissues sustain loss of o Modification is usually accomplished by cytochrome P-450 mixed-function
cells in addition to the cells that are irreversibly damaged at the end of ischemia oxidases in the sER of liver and other organs
 Toxic metabolites cause membrane damage and cell injury mainly by formation of free
o Clinically important: contributes to tissue damage during myocardial and
radicals and subsequent lipid peroxidation, also direct covalent binding
cerebral infarction and following therapies to restore blood flow  Ex: CCL4  converted to highly reactive free radical CCL3 by cyt450
o Reperfusion injury occurs when new damaging processes happen during o Causes lipid peroxidation and damages many cellular structure
reperfusion , causing death of cells that might have recovered otherwise.  Ex: Acetaminophen  analgesic; converted to a toxic product in the liver  cause cell
 Several mechanisms: injury

Oxidative stress - New damage may be initiated during reoxygenation by ↑ ROS


- Free radicals may be produced in reperfused tissue as a result of KEY CONCEPTS: Ischemic and Toxic Injury
incomplete reduction of oxygen by damaged mitochondria, or  Mild Ischemia: ↓ oxidative phoaphorylation  ↓ ATP generation  failure of Na pump
because of action of oxidases from leukocytes, endothelial or  influx of Na and H2O  organelle and cellular swelling (reversible)
parenchymal cells  Severe/prolonged ischemia: severe swelling of mitochondria, Ca influx into
- Cellular antioxidant defense mechanism may be compromised by mitochondria and into cell w/ rupture of lysosomes and PM. Death by necrosis and
ischemia  favors accumulation of free radicals apoptosis due to release of cytC from mitochondria
Intracellular - Intracellular and mitochondrial calcium overload begins during  Reperfusions injury follows blood flow into ischemic area is due to oxidative sress by
Ca2+ overload acute ischemia  exacerbated during perfusion due to influx of the release of free radicals from WBC and endothelial cells. Blood brings Ca that
calcium resulting from cell membrane damage and ROS overloads reversibly injured cells with mitochondrial injury. Influx of WBC generated
mediated injury to SR free radicals and cytokines. Local activation of complement by IgM deposited in
- Ca2+ overload favors opening of the MPTP with resultant ischemic tissues
depletion of ATP  furthers cell injury  Chemicals may cause injury directly or by conversion to toxic metabolites. Direct
Inflammation - Result of: “dangers signals” released from dead cells, cytokines injury to critical organelles such as mitochondria or indirect injury from free radicals
released by macrophages; increased expression of adhesion generated from chemicals/toxins involved
molecules by hypoxic parenchymal and endothelial cells 
recruit circulating to reperfused tissue
- Inflammation causes additional tissue injury
abmrmtmd
APOPTOSIS  Accumulation of misfolded proteins
o May arise because of mutations in genes or because of extrinsic factors
 Apoptosis is a pathway of cell death that is induced by a tightly regulated o Excessive accumulation in ER leads to ER stress  ends in apoptotic cell
suicide program in which cells destined to die activate intrinsic enzymes death
that degrade the cell’s own nuclear DNA and nuclear and cytoplasmic o Apoptosis by accumulation of misfolded proteins  basis of several
proteins degenerative diseases of the CNS and other organs
 Apoptotic bodies – fragmented apoptotic cells  Cell death in certain infections (particularly viral infections)
o Contain portions of the cytoplasma and nucleus o Adenovirus, HIV or viral hepatitis
o Plasma membrane is intact but they become “tasty” targets for phagocytes o Important host response  cytotoxic T cells  induce apoptosis of infected
o Rapidly devoured before contents have leaked out  cell death does not cells  eliminate reservoir of infection
elicit inflammation o Cytotoxic T cells also responsible for cell death in tumors and cellular
 Apoptosis – programmed cell death; characterized by: rejection of transplants
o Loss of membrane integrity  Pathologic atrophy in parenchymal organs after duct obstruction
o Enzymatic digestion of cells o Pancreas, parotid glands, kidney
o Leakage of cellular components
o Host reaction MORPHOLOGIC AND BIOCHEMICAL CHANGES IN APOPTOSIS

CAUSE OF APOPTOSIS  Best seen in electron microscope

Apoptosis – serves to remove unwanted, aged, or potentially harmful cells; also a Cell shrinkage - Cell is smaller in size; cytoplasm is dense; organelles
pathologic event when diseased cells become damaged beyond repair are tightly packed
Chromatin condensation - Most characteristic feature of apoptosis
Apoptosis in Physiologic Situations - Chromatin aggregates peripherally,under the nuclear
membrane, into dense masses of shapes and sizes
- Nucleus fragmentation
 Death by apoptosis is a normal phenomenon that serves to eliminate cells that are no
Blebbing and apoptotic - Apoptotic cell first shows extensive surface blebbing,
longer needed, and to maintain a steady number of various cell populations in tissues.
bodies then undergo fragmentation into membrane-bound
 It is important in following physiologic situations: apoptotic bodies (w/ or w/o nuclear fragments
Phagocytosis of apoptotic - Apoptotic bodies are rapidly ingested by phagocytes
 The destruction of cells during embryogenesis cells or cell bodies, usually and degraded by the phagocytes lysosomal ezymes
o implantation, organogenesis, developmental involution, metamorphosis by macrophages
 Involution of hormone-dependent tissues upon hormone withdrawal  Plasma membranes are thought to remain intact during apoptosis until the last stages
o Endometrial cell breakdown during menstrual cycle, ovarian follicular atresia when they become permeable
in menopause, prostatic atrophy after castration  H & E: apoptotic cell appears as round or oval mass of intensely eosinophilic
 Cell loss in proliferating cell populations cytoplasm with fragments of dense nuclear chromatin
o Immature lymphocytes in bone marrow, thymus, B lymphocytes in germinal  Apoptosis does not elicit inflammation  difficult to detect histologically
centers, epithelial cells in intestinal crypts  to maintain homeostasis
 Elimination of potentially harmful self-reactive lymphocytes Mechanisms of Apoptosis
 Death of host cells that have served their purpose
o Neutrophils in acute response, lymphocytes at the end of immune response  Apoptosis results from the activation of enzymes called caspases (cysteine
 deprived of growth factors proteases that cleave proteins after aspartic residues)
 Presence of cleaved, active caspases is a marker for cells undergoing apoptosis
Apoptosis in Pathologic Conditions  Process of apoptosis may be divided into:
o Initiation phase – where caspases become active
 Apoptosis eliminates cells that are injured beyond repair without eliciting a host o Execution phase – other caspases trigger the degradation of critical
reaction  limit tissue damage cellular components
 Death by apoptosis is responsible for loss of cells in a variety of pathologic states:  Two distinct pathways converge on caspase activation:
o Mitochondrial pathway
 DNA damage o Death receptor pathway
o Radiation, cytotoxic anticancer drugs, hypoxia  damage DNA directly or
via free radicals

abmrmtmd
Intrinsic or Mitochondrial Pathway of Apoptosis Extrinsic or Death Receptor-Initiated Pathway of Apoptosis
 The mitochondrial pathway is the major mechanism of apoptosis in all  This pathway is initiated by engagement of plasma membrane death
mammalian cells receptors on a variety of cells
 Results from increased permeability of mitochondrial outer membrane with release of  Death receptors (members of TNF receptor family)
pro-apoptotic molecules from the mitochondrial intermembrane space into cytoplasm o Contain death domain protein-protein interaction; essential for
 Mitochondria are remarkable organelles  they contain cytochrome c which is delivering apoptotic signals
essential for life o TNFR1 & CD95
o Release of cytochrome c  indication that the cell is not healthy  initiate o Mechanism of apoptosis is induced by Fas (death receptor in many cel
the suicide program of apoptosis types)
 BCL2 – controls the release of mitochondrial pro-apoptotic proteins  FasL – ligand for Fas; expressed on T-cells that recognize self-antigens and cytotoxic
T cells
Anti- - BCL2, BCL-XL, MCL1 o FasL + FAS = FADD
apoptotic - Possess four BH domains (BH1-4) o FADD attached to death receptors binds an inactive form of caspase-8 or 10
- Reside in the outer mitochondrial membranes as well as the via death domain  cleave to form active caspase 8
cytosol and ER membranes  This pathway of apoptosis can be inhibited by FLIP  binds to pro-caspase 8but
- Prevent leakage of cytochrome c and other death-inducing cannot cleave and activate the caspase because it lacks a protease domain
proteins into the cytosol by keeping the mitochondrial outer o Inhibits FAS-mediated apoptosis
membrane impermeable
Pro- - BAX and BAK (they also have four BH domains)
apoptotic - Upon activation, they oligomerize w/in the outer mitochondrial The Execution Phase of Apoptosis
protein and promote permeability  The two initiating pathways converge to a cascade of caspase activation,
- They form a channel in the outer mitochondrial membrane  which mediates the final phase of apoptosis
allow leakage of cytochrome c from the intermembranous space  Mitochondrial pathway leads to activation of the initiator caspase 9
Sensors - BAD, BIM, BID, Puma, Noxa  Death receptor pathway leads to activation of caspases-8 and -10
- Contain only one BH (3rd of the four BH domains; hence they are  After initiator caspase is cleaved to active form  sequential activation of executioner
called BH3-only proteins) caspase (caspase-3 and -6)
- BH3-only proteins – acts as sensors of cellular stress and o Ex: caspase 3 and 6 cleave DNAse inhibitor  makes DNAse active 
damage; regulate balance between the other two groups  thus induce DNA cleavage
acting as arbiters of apoptosis  Caspase also degrade structural components and promote fragmentation of nuclei

 Growth factors and other survival signals stimulate the production of anti-apoptotic
proteins such as BCL2  prevent leakage of deatg-inducing proteins fron the outer
Removal of Dead Cells
mitochondrial membrane
 Formation of apoptotic bodies breaks cells up into “bite-sized” fragments that are
 When cells are deprived of survival or their DNA is damaged, or misfolded proteins
edible for phagocytes
induce ER stress  BH3-only proteins “sense” such damage and are activated
 Phosphatidylserine – present on the inner leaflet of plasma membrane of a healthy
o Sensors activate pro-apoptotic effectors (BAX and BAK)  allow leakage of
cell
proteins
o Flips out and expressed on the outer layer of membrane in apoptotic cells
 BH3-only proteins may also bind to and block the function of BCL2 and BCL-XL 
 recognize by macrophage receptors
BCL2 and BCL-XL may decline
 Dying cells by apoptosis secrete soluble factors that recruit phagocytes  target for
 BAX-BAK activation  activate caspase cascade and release cytochrome c  cyt c
engulfment; “eat me” signal
binds to APAF-1 (forms wheel-like hexamer called apoptosome)  complex is able to
o E.g., thrombospondin; C1q
bind to caspase-9
 This process is efficient that dead cells disappear within minutes without a trace 
 Caspase-9  critical initiator caspase of the mitochondrial pathway  set up an
inflammation is absent even in the face of extensive apoptosis
autoamplification process
 Cleavage activates caspase-9  triggers other pro-caspases  mediate the execution
phase of apoptosis
 Smac/Diablo – enter the cytoplasm and bind to and neutralize cytoplasmic proteins
that function as physiologic inhibitors of apoptosis  called IAPs
 IAPs – block the activation of caspases, including executioners like caspase-3 and
keep cells alive
o Neutrlaization of IAPs permits the initiation of a caspase cascade

abmrmtmd
CLINICOPATHOLOGIC CORRELATIONS: APOPTOSIS IN HEALTH AND DISEASE SELECTED DISEASES CAUSED BY MISFOLDED PROTEINS
Disease Affected Protein Disease
Examples of Apoptosis Cystic fibrosis CFTR Loss of CTFRdefect in chloride transport
Growth Factor - Hormone-sensitive cells, lymphocyte and neurons die by Familial LDL receptor Loss of LDL
Deprivation apoptosis if deprived by their growth factor hypercholesterolemia receptorhypercholesterolemia
- Apoptosis is triggered by the intrinsic (mitochondrial) Tay-Sachs disease Hexosaminidase β Loss of lysosomal enzyme storage of GM,
pathway  attributed by the ↓ synthesis of BCL2 and subunit gangliosides in neurons
BCL-XL and activation of BIM and other pro-apoptotic Alpha-1 antitrypsin A1-antitrypsin Storage of non-functional protein in
members of the BCL2 family deficiency hepatocytes cause apoptosis; absence of
DNA Damage - Exposure to radiation or chemotherapeutic agents  enzymatic activity in lungs causes
initiated by DNA damage or genotoxic stress  involves destruction in elastic tissue giving rise to
TP53 (tumor-suppressor gene) emphysema
- P53 accumulates in cells when DNA is damaged  Creutzfeldt-Jacob Prions Abnormal folding of PrPsc causes neuronal
arrest G1 phase of cell cycle to allow time for repair disease cell death
- If damage is too great  P53 triggers apoptosis Alzheimer disease Aβ peptide Abnormal folding of Aβ peptide causes
- If TP53 is mutated or absent (cancers)  cells with aggregation w/in neurons and apoptosis
damaged DNA fail to undergo p53-mediated apoptosis
and survive  may cause mutations and neoplastic
transformations Disorders Associated with Dysregulated Apoptosis (“too little or too much”)
- P53 serves as a critical “life or death” switch ff Disorders associated - Incorrect low rate of apoptosis  permit survival or
genotoxic stress with defective abnormal cells
o Triggers the distal death effector machinery; apoptosis and - E.g., TP53 mutations cause accumulation of defective
o Involve its function as a DNA-binding increased cell DNA repair  give rise to cancer
transcription factor survival - Defective apoptosis results in failure to eliminate harmful
o BAX, BAK, BH3-only proteins cells (e.g., self Ags)  basis of autoimmune disease
Protein Misfolding - Ubiquitinated and targeted for proteolysis in Disorders associated - Diseases characterized by loss of cells and include:
proteasomes with increased - (1) Neurodegenerative diseases – manifested by loss
- Accumulation of un/misfolded proteins  trigger apoptosis and of specific neurons (apoptosis caused by mutations and
“unfolded protein response”  activates signalling excessive cell death misfolded proteins)
pathways to ↑ production of chaperones  enhance - (2) Ischemic injury – e.g., myocardial infarction and
proteasomal degradation and slow protein translation  stroke
reduce load of misfolded proteins in cell - (3) death of virus-infected cells – e.g., viral infections
- If unable to cope with accumulation  induces
apoptosis called “ER stress”
- Alzheimer, Huntington, Parkinson Disease, Type 2 DM –
neurodegenerative diseases caused by intracellular KEY CONCEPTS: Apoptosis
accumulation of abnormally folded proteins  Regulated mechanism of cell death that serves to eliminate unwanted and irreparably
- Deprivation of glucose and oxygen, and stress (e.g., damaged cells, w/ the least possible host reaction
heat) – also result in protein misfolding  Cxd by enzymatic degradation of proteins and DNA, initiated by caspases; and by
Apoptosis Induced by - FasL on T cells binds to Fas on the same or neighboring recognition and removal of dead cells by phagocytosis
TNF Receptor Family lymphocytes  Initiated by two major pathways:
- Plays a role in the elimination of lymphocytes that Mitochondrial  Triggered by loss of survival signals, DNA damage, and
recognize self Ags, and mutations affecting Fas or FasL (intrinsic) accumulation of misfolded proteins (ER stress)
result in autoimmune diseases in humans and mice pathway  Asso. w/ leakage of pro-apoptotic proteins from mitochondrial
Cytotoxic T - CTL (cytotoxic T-lymphocytes) recognize foreign Ags on membrane into cytoplasm, where they activated caspases
Lymphocyte-Mediated the surface of host cells  Inhibited by anti-apoptotic members of BCL2 family (induced
Apoptosis - Activated CTLs secrete perforin – transmembrane pore- by survival signals; GF)
forming molecule  promotes entry of granzymes Death  Responsible for elimination of self-reactive lymphocytes and
- Granzymes – cleave proteins at aspartate residues receptor damaged cytotoxic T-lymphocytes
- CTL kills target cells by directly inducing the effector (extrinsic)  Initiated by engagement of death receptors (TNF receptor
phase of apoptosis pathway family) by ligand on adjacent cells

abmrmtmd
NECROPTOSIS KEY CONCEPTS: Necroptosis and Pyroptosis
 Necroptosis resembles necrosis morphologically and apoptosis mechanistically as a
 Form of cell death with aspects of both necrosis and apoptosis form of programmed cell death
 Characterization of necroptosis:  Necroptosis is triggered by ligation of TNFR1, and viral proteins of RNA and DNA virus
 Necroptosis is caspase-independent but dependent on signalling by RIP1 and RIP3
 RIP1-RIP3 signaling reduces mitochondrial ATP generation, causes production of ROS,
 Morphologicallyand biochemically, resemples necrosis
and permeabilizes lysosomal membranes, causing cellular swelling and membrane
o Cxd by loss of ATP damage as well as necrosis
o Swelling of cell and organelles  Release of cellular contents evokes an inflammatory reaction as in necrosis
o Generation of ROS  Pyroptosis occurs inc ells infected by microbes. It involves activation of caspase-1
o Release of lysosomal enxymes which cleaves the precursor form IL-1 to activate it. Caspase-1 along with closely
o Rupture of the plasma membrane related caspase-11 also cause of death of the infected cell
 Mechanistically, it is triggered by genetically programmed signal transduction that
culminate in cell death  resembles programmed cell death (hallmark of apoptosis)
AUTOPHAGY
 Necroptosis aka “programmed necrosis” or “ caspase-independent programmed cell
 Autophagy is a process in which a cell eats its own contents
death”
 Involves delivery of cytoplasmic materials to the lysosome for degradation
 In sharp contrast to apoptosis, the genetic program that drives necroptosis does not
 Can be categorized into three types:
result in caspase activation  “ caspase-independent programmed cell death”
 Initiation of necroptosis is similar with extrinsic form of apoptosis  by ligation of a
Chaperone-mediated - Direct translocation across the lysosomal membrane by
receptor by its ligand (TNFR1) autophagy chaperone proteins
o TNF can cause both apoptosis and necroptosis Microautophagy - Inward invagination of lysosomal membrane for delivery
 Necoptosis involves two unique kinases: RIP1 and RIP3 Macroautophagy - Referred to as autophagy
o Ligation of TNFR1 recuits RIP1 and RIP3 into a multiprotein complex that - Major form of autophagy involving the sequenstration and
contains caspase-8 transportation of portions of cytosol in a double-membrane
o Unlike apoptosis, caspases are not activated bound autophagic vacuole
 Autophagy is seen in single-celled organisms as well as mammalian cells
o Like in necrosis, terminal events include permeabilization of lysosomal
o Survival mechanism in states of nutrient deprivation  starved cell eats
membranes, generation of ROS, damage to mitochondria, and ↓ ATP levels
itself and recycles digestive contents
o This explains the morphologic similarity of necroptosis with necrosis initiated
o It is seen in physiologic states (e.g., aging and exercise) and pathologic
by other injuries
 Necroptosis – important death pathway in physiologic and pathologic conditions processes
o Proceeds through several steps:
o E.g., necroptosis occurs in mammalian growth plate, cell death in
steatohepatitis, acute pancreatitis, reperfusion injury and neurodegenerative
diseases (ex, Parkinson)
 Formation of an isolation membrane (aka phagosphore) and its nucleation; the
o Acts as a back up mechanism in host defense against viruses that encode isolation membrane is believed to be derived from the ER
caspase inhibitor (ex:CMV)
 Elongation of the vesicle
 Pyroptosis – form of programmed cell death accompanied by the release of fever  Maturation of the autophagosome, its fusion with lysosomes, and eventual
inducing cytokine IL-1 and bears some biochemical properties with apoptosis
degradation of the contents
 Microbial products that infected the cells are recognized by innate immune receptors
 Starvation or depletion of growth factors activate an initiation complex of four proteins
 activate multiprotein complex called “inflammasome”
that stimulates the assembly of a nucleation complex  promotes nucleation of
o Inflammasome – activate caspase-1 (aka IL-1b converting enzyme) 
autophagosomal membrane  surrounds and closes to form autophagosome
cleaves and activates IL-1
o Require LC3
o IL-1 – mediator of leukocyte recruitment and fever
o LC3 – augmented during autophagy; marker for identifying cells in which
o Caspase-1 and -11  also induce cell death
autophagy is occurring; “target” protein aggregates and effete organelles
o This pathway of death is czd by:
o Newly formed autophagosome fuses with endosome and lysosome to form
 Swelling of cells
autophagolysosome
 Loss of plasma membrane integrity
o Loading of cargo into autophagosome is “selective”
 Release of inflammatory mediators
 Autophagy is a survival mechanism that maintains the integrity of the cells by
 Pyroptosis results in the death of microbes the enters the cytosol and promotes
recycling essential metabolites and clearing cellular debris
release of inflammasome-generated IL-1
o Prominent in atrophic cells exposed to severe nutrient deprivation

abmrmtmd
 Autophagy is involved in the turnover of organelles like ER, mitochondria, and LIPIDS
lysosomes and the clearance of intracellular aggregates that accumulate during aging,
stress, and various disease states  All major lipids can accumulate in cells: triglycerides, cholesterol/cholesterol
 Autophagy can trigger cell death if it is inadequate to cope with the stress on the cell esters, phospholipids
 Autophagic vacuolization often precedes or accompanies call death  Phospholipids – components of the myelin figures found in necrotic cells
 Autophagy plays a role in human disease:  Abnormal complexes of lipids and carbohydrates accumulate in the lysosomal storage
diseases
Cancer - Autophagy can both promote cancer growth and act as a
defense against cancers Steatosis - Abnormal accumulations of triglycerides within
Neurodegerative - Associated with dysregulation of autophagy (Fatty parenchymal cells
Diseases - E.g., Alzheimer and Huntington Disease Change) - Fatty change is often seen in the liver because it is the major
Infectious diseases - E.g., mycobacteria, shigella, HSV-1 organ involved in fat metabolism (also occurs in heart, muscle,
Inflammatory bowel - E.g., Crohn disease and ulcerative colitis kidney)
diseases - Causes include: toxins, protein malnutrition, DM, obesity and
anoxia
- In developed nations, the most common cause of fatty change in
Key Concepts: Autophagy liver are alcohol abuse and non-alcoholic fatty liver disease,
associated with diabetes and obesity
 Autophagy involves sequestration of cellular organelles into cytoplasmic autophagic
Cholesterol - Accumulations are seen in several pathologic processes:
vacuoles (aurophagosomes) that fuse with lysosomes and digest the enclosed material
 Autophagy is an adaptive response that is enhanced during nutrient deprivation, and
allowing the cell to cannibalize itself to survive Cholesterol Atherosclerosis - Smooth muscle cells and
Esters macrophages w/in the intimal layer
 Autophagosome formation is regulated by more than an dozen proteins that act in a
coordinated manner of aorta and large arteries are filled
 Dysregulation of autophagy occurs in many diseases states including cancer, with lipid vacuoles, most of which
inflammatory bowel disease, and neurodegenerative disorders. Autophagy play a role are made up of cholesterol and
in host defense against certain microbes cholesterol esters
- Cells have foamy appearance  foam
cells
- Aggregation in intima produces yellow
INTRACELLULAR ACCUMULATIONS cholesterol-laden atheromas
characteristic of the disorder
 One of the manifestations of metabolic derangements - Extracellular cholesterol esters may
 May be located in the cytoplasm, within organelles (usually lysosomes), or in the crystallize in the shape of long needles,
nucleus; may be synthesized by the affected cells or may be produced elsewhere producing quite distinctive clefts in tissue
 Four main pathways of abnormal intracellular accumulations: sections
Xanthomas - Intracellular accumulation of
cholesterol within macrophages is
 Inadequate removal of a normal substance secondary to defects in mechanisms of
also characteristic of acquired and
packaging and transport (e.g., stetaosis) hereditary hyperlipidemic states
 Accumulation of an abnormal endogenous substance as a result of genetic or acquired - Clusters of foamy cells found in
defects: folding, packaging, transport or secretion (e.g., mutated a1-anitrypsin) subepithelial connective tissue of skin
 Failure to degrade a metabolite due to inherited enzyme deficiencies  storage and tendons  produce timorous
diseases masses aka xanthomas
 Deposition and accumulation of an abnormal exogenous substance when the cell has Cholesterolosis - Refers to the focal accumulations of
cholesterol-laden macrophages in lamina
neither the enzymatic machinery to degrade it nor the ability to transport. (e.g.,
propria of gall bladder
accumulation of carbon particles)
Niemann-Pick - Lysosomal storage disease cause by
diseases, type C mutations affecting enzyme involved in
 Accumulation is reversible if overload can be controlled or stopped cholesterol trafficking, resulting in
 Inherited storage disease  accumulation is progressive  overload causes cell injury cholesterol accumulation in multiple
that leads to death of tissue and patient organs

abmrmtmd
PROTEINS GLYCOGEN

 Intracellular accumulations of proteins usually appear as rounded,  Glycogen – energy source stored in the cytoplasm of healthy cells.
eosinophilic droplets, vacuoles, or aggregates in the cytoplasm  Excessive intracellular deposits of glycogen are seen in patients with an
 EM: amorphous, fibrillar, or crystalline appearance abnormality in either glucose or glycogen metabolism
 Amyloidosis – abnormal proteins deposit primarily in extracellular spaces  Glycogen masses appear as clear vacuoles within the cytoplasm  they dissolve in
 Excesses of proteins within the cells sufficient to cause morphologically visible aqueous fixatives
accumulation have diverse causes: o Readily identified when fixed in absolute alcohol
o Best carmine or PAS  rose to violet color
 Reabsorption droplets in proximal renal tubules are seen in renal diseases associated o Diastase digestion before staining serves as a control by hydrolyzing the
with proteinuria glycogen
o Small amounts of protein in kidney – absorbed by pinocytosis in PCT  Ex: Diabetes mellitus  prime example; glycogen is found in renal tubular epithelial
o Heavy protein leakage - ↑ reabsorption into vesicles  proteins appears as cells, liver cells, b cells of islets of Langerhans, heart muscle cells
pink hyaline droplets within the cytoplasm of tubular cell  Glycogen storage disease or glycogenoses – group of genetic disorders where
o Process is reversible; if proteinuria diminishes  protein droplets are glycogen accumulates within cells
metabolized and disappear
 Proteins that accumulate may be normal secreted proteins that are produed in PIGMENTS
excessive amounts.
o E.g., active synthesis of Ig  Pigments – colored substances (normal or abnormal) that accumulate in cells
o ER becomes hugely distended  produce large, homogenous eosinophilic  Can be exogenous (coming from outside the body) or endogenous (synthesized
inclusions called Russell bodies within the body itself
 Defective intracellular transport and secretion of critical proteins
o E.g., a1-antitrypsin deficiency  build-up of partly folded intermediates  Exogenous Pigments
aggregate in liver and not secreted  causes emphysema  Most common is carbon (coal dust), a ubiquitous air pollutant in urban
areas
 Accumulation of cytoskeletal proteins: microtubules, actin, myosin, intermediate
o accumulation of this pigment blacken the tissues of the lungs (anthracosis)
filaments and lymph nodes
o Intermediate filaments: keratin, neurofilaments, vimentin, glial filaments o coal worker’s pneumoconiosis
o Accumulations of keratin and neurofilaments  asso. w/ cell injury  Tattooing is a form of localized, exogenous pigmentation of the skin
o Alcoholic hyaline – eosinophilic cytoplasmic inclusion in liver cells that is  The pigments do not usually evoke any inflammation
characteristic of alcoholic liver disease; composed mainly of keratin
intermediate filaments
o Neurofibrillary tangle found in the brain in Alzheimer disease contains Endogenous Pigments
neurofilaments and other proteins  Lipofuscin is an insoluble pigment, aka lipochrome or wear-and-tear
pigment
 Aggregation of abnormal proteins
o Composed of polymers of lipids in complex with protein; derived rough lipid
o Abnormal/misfolded proteins may deposit in tissues and interfere with peroxidation of polyunsaturated lipids of subcellular membranes
normal functions o Not injurious to cell or function
o Deposits can e intracellular, extracellular or both o Telltale sign of free radical injury and lipid peroxidation
o Aggregates may either directly or indirectly cause the pathologic changes o Appears a yellow-brown, finely granular cytoplasmic, perinuclear pigment
o E.g., proteinopathies or protein-aggregation diseases such as amyloidosis o Seen in cells undergoing slow, regressive changes; prominent in liver and
heart of aging patients or with severe malnutrition and cancer cachexia
 Melanin – endogenous, brown-black, pigment formed when the enzyme tyrosinase
HYALINE CARTILAGE
catalyzes the oxidation of tyrosine to DHP in melanocytes; the only endogenous
brown-black pigment
 Hyaline – refers to an alteration w/in cells or in extracellular space that  Homogentisic acid – black pigment that occurs in patients with alkaptonuria;
gives a homogenous, glassy, pink appearance in routine H&E deposited in the skin, CT, cartilage and the pigmentation is known as ochronosis
 Intracellular accumulation of protein are examples of intracellular hyaline deposits  Hemosiderin – Hb-derived, golden yellow-to-brown, granular or crystalline
 Extracellular hyaline (e.g., collagenous fibrous tissue in old scars) pigment is one of the major storage forms of iron
 Hypertension and DM – walls of arterioles (esp kidney) are hyalinized  resulting o Iron is transported by transferring and stored with apoferritin to form
ferritin micelles
from extravasated plasma protein and deposition of basement membrane material
o When there is a local or systemic excess of iron, ferritin forms hemosiderin
granules (seen in LM)
o Hemosiderin represents aggregates of ferritin micelles
abmrmtmd
o BM, spleen, liver  engaged in RBC breakdown  Occur throughout the body but mostly affects interstitial tissues of gastric mucosa,
o Local hemosiderosis  e.g., common bruise kidneys, lungs, systemic arteries, pulmonary veins
o Green bile (biliverdin)  red bile (bilirubin) o Excrete acid  have internal alkaline compartment  predisposes to
 Hemosiderosis – deposition of hemosiderin in organs and tissues when there is metastatic calcification
systemic overload: main causes:  May occurs as noncrystalline amorphous deposits or hydrocyapatite crystals
o ↑ absorption of dietary iron due to an inborn error of metabolism called
hemochromatosis
o Haemolytic anemias  prematurelysis of RBC lead to abnormal quantities of KEY CONCEPTS: Abnormal Intracellular Depositions and Calcifications
iron  Deposition of lipids
o Repeated blood transfusion (transfused RBC – exogenous iron load) o Fatty change – accumulation of free TAGs in cells from excess intake or
defective transport; manifestation of reversible cell injury
o Cholesterol deposition – result of defective catabolism and excessive
PATHOLOGIC CALCIFICATION intake; in macrophage and smooth muscle cells of vessel walls in
atherosclerosis
 Pathologic calcification is the abnormal tissue deposition of calcium salts,  Deposition of proteins – reabsorbed proteins in kidney tubules; Ig in plasma cells
together with smaller amounts of iron, magnesium and other mineral salts  Deposition of glycogens – in macrophage of px w/ defects in lysosomal enzymes
 There are two forms: that breakdown glycogen (glycogenoses)
 Deposition of pigments – indigestible pigments such as carbon, lipofiscin (product
o Dystrophic calcification – deposition occurs locally in dying tissues;
of lipid peroxidation), or iron (due to overload; hemosiderosis)
 it occurs despite normal serum levels of calcium and in the  Pathologic calcification
absence of derangements in calcium metabolism o Dystrophic calcification – deposition of calcium at sites if cell injury and
o Metastatic calcification – deposition of calcium salts in abnormal tissues; necrosis
 almost always results from hypercalcemia secondary to some o Metastatic calcification – deposition of calcium in normal tissues, caused
disturbance in calcium metabolism by hypercalcemia (consequence of PTH excess)

Dystrophic Calcification CELLULAR AGING


 Dystrophic calcification is encountered in areas of necrosis, whether they
are of coagulative, caseous, or liquefactive type and in foci on enzymatic  Cellular aging is the result of a progressive decline in cellular function and
necrosis of fat viability caused by genetic abnormalities and the accumulation of cellular
 Calcification is almost always present in the atheromas of advanced atherosclerosis and molecular damage due to the effects of exposure to exogenous
 Commonly develops in aging, damaged heart valves influences
 Calcium salts appear macroscopically as fine, white granules or clumps, often felt as
gritty deposits (sometimes converted to stone in tuberculous lymph node)
DNA Damage
 Dystrophic calcification may simply be a telltale sign of previous cell injury and often
 Exogenous and endogenous factors threaten the integrity of nuclear and
cause of organ dysfunction
mitochondrial DNA
 Serum calcium is normal in dystrophic calcification
 Werner Syndrome – premature aging; DNA helicase is defective (protein involved in
DNA replication and repair
o Defect in DNA helicase causes rapid accumulation of chromosomal damage
Metastatic Calcification that mimic the injury that normally accumulates during cellular aging
 May occur in normal tissues whenever there is hypercalcemia  Genetic instability – characteristic of other disorders where px display some of the
 Hypercalcemia also accentuates dystrophic calcification manifestations of aging at increased rate
 Four principal causes of hypercalcemia: o Bloom syndrome and ataxia telangiectasia  mutated genes encode
o ↑ PTH (hyperparathyroidism, ectopic PTH secretion by malignant tumors) proteins involved in repairing double-strand breaks in DNA
o Resorption of bone tissue (secondary to primary tumors of BM, or
diffuse skeletal metastasis, accelerated bone turnover, or immobilization)
o Vitamin D-related disorders (vit. D intoxication, sarcoidosis, idiopathic
Cellular Senescence
hypercalcemia of infancy like Williams syndrome, cxd by abnormal
 All normal cells have a limited capacity for replication, and after a fixed
sensitivity to vit D)
number of divisions cells become arrested in a terminally nondividing state,
o Renal failure (causes retention of phosphate  secondary
known as replicative senescence
hyperparathyroidism)
 Aging is associated with progressive replicative senescence of cells
 Less common causes includes:
 Cells from children have the capacity to undergo more rounds of replication than do
o Aluminium intoxication (px w/ chronic renal dialysis)
cells from older people
o Milk-alkali syndrome (excessive ingestion of calcium and absorbable
 Two mechanisms underlie cellular senescence:
antacids such as milk or calcium carbonate

abmrmtmd
Telomere attrition - Progressive shortening of telomeres  results in
cell cycle arrest
- Telomeres – important for ensuring the complete
KEY CONCEPTS: Cellular Aging
replication of chromosome ends and for protecting the
 Cellular aging results from a combination of accumulating cellular damage, reduced
ends from fusion and degradation
capacity to divide (replicative senescence), reduced ability to repair damaged DNA,
- When somatic cells replicate  telomeres are shortened 
and defective protein homeostasis.
ends of chromosomes are not protected and seen as
o Accumulation of DNA damage: defective DNA repair mechanisms;
broken DNA  signals cell cycle arrest
conversely, caloric restriction activates DNA repair and is known to prolong
- Telomere length is maintained by nucleotide addition by
aging in model organisms
telomerase
o Replicative senescence : reduced capacity of cells to divide secondary to
- Immortalized cancer cells – telomerase is reactivated and
progressive shortening of chromosomal ends (telomeres)
telomere length is stabilized  allow proliferation of cells
o Defective protein homeostasis: resulting from impaired chaperone and
Activation of - CDKN2A locus encodes two tumor suppressor proteins,
proteasome functions
tumor suppressor known as p16 or INK4a  correlated with chronologic age
o Nutrient sensing system: caloric restriction increases longevity.
genes - By controlling G1 to S phase progression during the cell
Mediators may be reduced IGF-1 signaling and increase in sirtuins
cycle, p16 protects the cells from uncontrolled mitogenic
signals and pushes cells along the senescence pathway

Defective Protein Homeostasis


 Protein homeostasis involves two mechanisms:
o Those that maintain proteins in correct folded forms (mediated by
chaperones)
o Those that degrade misfolded proteins by the autophagy0lysosome system
and ubiquitin-proteasome system
 Normal folding and degradation of misfolded proteins are impaired with aging
 Rapamycin – promotes autophagy

Deregulated Nutrient Sensing


 Eating less increases longevity
 Caloric restriction increases life span in all eukaryotic species
 Two major neurohormonal circuits that regulate metabolism:
o Insulin and IGF-1 signaling pathway – IGF-1 mimics intracellular
signalling of insulin, informs cells of the availability of glucose and promote
anabolic state as well as cell growth and replication. IGF-1 has multiple
downstream targets; AKT and its downstream target mTOR (w/c is inhibited
by rapamycin
o Sirtuins – family of NAD-dependent protein deacetylases. There are seven
types of sirtuins distributed in different cellular compartments and have
nonredundant functions deisgned to adapt body functions to stress (food
deprivation and DNA damage). Sirtuins promote expression of genes whose
products increase longevity. Includes proteins that: inhibit metabolic
activity, reduce apoptosis, stimulate protein folding, and inhibit harmful
effects of oxygen free radicals.
 Sirtuins increase insulin sensitivity and glucose metabolism, (may be target for
treatment of diabetes
 Caloric restriction increases longevity both by reducing the signalling intensity of the
IGF-1 pathway and by increasing sirtuins
 Attenuation of IGF-1 leads to lower rates of cell growth and metabolism reduced
cellular damage  this effect can be mimicked by rapamycin
 Sirtuin-6 serves dual functions:
o Contribute to metabolic adaptations of caloric restriction
o Promote genomic integrity by activating DNA repair enzyme through
deacylation
abmrmtmd

You might also like