You are on page 1of 13

Far Eastern University – Nicanor Reyes Medical Foundation CELLULAR RESPONSES TO STRESS AND NOXIOUS STIMULI

Pathology A – Cellular Responses to Stress and Toxic Insults:


Adaptation, Injury, and Death.
Pedrito Tagayuna M.D.

PATHOLOGY
 Study of structural, biochemical, and functional changes in
tissues, and organs that underlie disease.
 Pathos (Gk.) – Suffering.
 Divisions of Pathology:
 General Pathology
- Concerned with common reactions of cells and tissues to
injurious stimuli.
- Such reactions are not tissue specific.
 Systemic Pathology
- Examine alterations and underlying mechanisms in organ
specific diseases.

Four aspects of the disease process  Cells able to handle physiologic demands and maintain
homeostasis.
ETIOLOGY PATHOGENESIS  Adaptation is a reversible functional and structural response to
change in physiologic states (e.g pregnancy) or in some
pathologic stimuli, during which altered steady states are already
MORPHOLOGIC CLINICAL
achieved.
CHANGES MANIFESTATION  Adaptive changes can be in the form of:
 Hypertrophy
1.) ETIOLOGY  Hyperplasia
 The cause of the disease, which can be grouped into 2 classes:  Atrophy
 Genetic – inherited mutations and disease-associated gene  Metaplasia
variants or polymorphisms.  If the injury is mild and the stimuli are removed, reversible cell
 Acquired – Infectious, Nutritional, Chemical or Physical. injury will happen and it can return to normal state so long that
 Most of the common afflictions can be considered multifactorial the noxious stimuli are removed.
wherein various external triggers can affect genetically  If limits of adaptive responses are exceeded, irreversible cell
susceptible individuals. injury will happen and will progress to cell death.
 Cell death is the end result of progressive cell injury which may
2.) PATHOGENESIS be due to ischemia, infection, toxin, mechanical stressors or
 Sequence of cellular, biochemical, and molecular events that even some physiologic needs (e.g. embryogenesis).
follow the exposure of cells or tissues to an injurious agent.  There are two principal ways of cell death:
 One of the main domains of pathology.  Apoptosis
 Necrosis
3.) MORPHOLOGIC CHANGES
 Structural alterations in cells or tissues that are either ADAPTATIONS OF CELLULAR GROWTH AND DIFFERENTIATION
characteristic of the disease itself or diagnostic of an etiologic  Cellular adaptations are reversible changes in the cell.
process.
 Morphologically identical lesions can be differentiated by 1.) HYPERTROPHY
molecular analysis such as in cases of tumors.  Increase in the size of cells that result in an overall increase in
the size of the organ.
4.) CLINICAL MANIFESTATIONS (Functional Derangements)  There are no new cells produced, the cells just get larger.
 End results of genetic, biochemical, and morphologic changes in  This is due to increased synthesis and assembly of intracellular
the tissues. structural components.
 Symptoms and signs of the disease.  More prominent in non-dividing cells because they cannot
 Clinical course and outcome. undergo hyperplasia.
 Rudolf Virchow: “Virtually all forms of disease start with  Hypertrophy can be physiologic or pathologic.
molecular or structural alterations in cells.”

Page 1 of 13
2.) HYPERPLASIA
 Increase in the number of cells in an organ or tissue.
 Frequently occurs together with hypertrophy in cells capable of
mitotic division.

Physiologic Hyperplasia
 Occurs due to actions of hormones or growth factors.
 Occurs when there is increased functional capacity or
compensatory increase after damage.
 Hormonal Hyperplasia: proliferation of glandular epithelium of
 Cells hypertrophy due to increased functional demands. the female breast during pregnancy.
 Stimulation of hormones and growth factors.  Compensatory Hyperplasia: Liver regeneration after
 Most common stimulus for muscle hypertrophy is increased hepatectomy.
workload (e.g. cardiac and skeletal muscles)
 Uterine Hypertrophy is a good example of hormone-induced Pathologic Hyperplasia
hypertrophy during pregnancy. (Physiologic Hypertrophy)  Occurs when there is an excessive or abnormal action of
 Faulty valves or increased hemodynamic load (e.g. Hypertension) hormones or growth factors.
is a stimulus for Cardiac hypertrophy. (Pathologic Hypertrophy)  Ex:
 Endometrial Hyperplasia – due to a stop in rising levels of
Mechanism of Hypertrophy progesterone.
 Increased production of cellular protein.  Benign Prostatic Hyperplasia – abnormal hormonal
 Myocardial Hypertrophy biochemical mechanisms: stimulation by androgens.

 Hyperplasia is different from cancer, but it constitutes a fertile


soil where cancer proliferation and malignancies may arise.
 Hyperplasia is also a characteristic response to papillomaviruses.
 Viral proteins can interfere with host proteins that regulate cell
proliferation.

Mechanism of Hyperplasia
 Growth factor driven cell proliferation.

3.) ATROPHY
 Reduction in the size of the organ.

Physiologic Atrophy:
 Integrated actions of mechanical sensors (which are  Such as during embryogenesis and developmental pathways.
triggered by increased workload), growth factors, and
vasoactive agents. Pathologic Atrophy:
 These three signals from the plasma membrane trigger two Decreased Workload
signal transduction pathways:  Initially the decrease in cell size is reversible as long as the disuse
a.) Phosphoinositide 3-kinase – important in physiologic. is not prolonged.
b.) G-Protein coupled messenger – important in pathologic.  Prolonged disuse can lead to permanent atrophy such as in
 The signaling pathways activate set of transcription factors: muscle atrophy.
a.) GATA4 Loss of Innervation (Denervation atrophy)
b.) NFAT (Nuclear Factor of Activated T-Lymphocyte)  Damage to nerve leads to disuse of muscles resulting to atrophy.
c.) MEF2 (Myocyte Enhancer Factor 2) Diminished Blood Supply
 Gradual decrease in blood supply could lead to atrophy.
 Hypertrophy is also associated with the shift of adult proteins to  Senile atrophy – due to atherosclerotic plaques.
neonatal proteins. Inadequate Nutrition
 Example in muscle hypertrophy, α-isoform of myosin heavy  Protein wasting (Marasmus)
chain is transformed to β-isoform in hypertrophic conditions.  Muscle Wasting (Cachexia)
 Genes expressed only during early development is reexpressed  TNF suppresses appetite and causes lipid depletion.
in hypertrophy.

Page 2 of 13
Loss of Endocrine Stimulation  However, the squamous epithelium removes the muco-ciliary
 Prominent on hormone dependent organs such as the breast clearance system of the respiratory epithelium, rendering it
and uterus. available to infectious agents.
Pressure  Persistent squamous metaplasia can lead to squamous cell
 Tissue compression for any length of time can cause atrophy. carcinoma of the respiratory tract.
 Benign tumors can compress vital surrounding tissues.

 Atrophic tissues have decrease cell size and organelles which


results to less metabolic requirements.
 In atrophic muscle, there may be fewer mitochondria and less Squamous to Columnar
ribosome in the ER.  Present in Barrett Esophagus.
 Early in the process, atrophic tissues have diminished function  The squamous lining of the esophageal epithelium is replaced by
but cell death is not prominent. columnar in response to gastric acid reflux.
 Cancer cells can also arise in these areas (adenocarcinomas)
Mechanism of Atrophy
 Decreased Protein Synthesis due to decreased metabolic activity. Connective Tissue Metaplasia
 Increased Protein Degradation.  Formation of cartilage, bone, or adipose tissue in areas where
 Occurs mainly by Ubiquitin Proteosome Pathway. they are normally not present.
 Ubiquitin (76-AA chain) tag cellular proteins which then become  Myositis ossificans – formation of bone in muscle fibers which
target for proteasomes for degradation. normally occur after intramuscular hemorrhage.
 Accelerated proteolysis.
 Autophagy is also prominent in atrophy, leaving some cell debris Mechanism of Metaplasia
and increase in autophagic vacuoles (e.g. Lipofuscin granules).  It is the result of reprogramming of stem cells.
 The precursor cells differentiate along new pathways driven by
4.) METAPLASIA cytokines, growth factors.
 Reversible change one differentiated cell type is replaced by  Retinoic Acid deficiency can lead to transcription dysfunction
another cell type. since it is a lipophilic substance and a direct regulator of gene
 Adaptive response wherein one cell type is replaced by another transcription.
cell type because the latte can handle the stress compared to
the first one. OVERVIEW OF CELL INJURY AND DEATH
Reversible Cell Injury
Columnar to Squamous (Squamous Metaplasia)  Injury is reversible if the damaging stimuli are removed.
 Columnar ciliated epithelium of the respiratory tract is replaced  Reduced oxidative phosphorylation results to reduced ATP.
with stratified squamous epithelium in response to chronic  Reduced ATP makes the Na-K pump to stop working.
irritation.  There will be Na influx and K efflux, together with Na, water will
 Prominent in smokers. go inside the cell and cause cellular swelling.
 It is also present in secretory epithelium of glandular organs
undergoing squamous metaplasia in response to calculi Cell Death (Irreversible Cell Injury)
formations.  Continuous damaging stimuli will result in irreversible cell death.
 Vitamin A deficiency also causes squamous metaplasia in the  Cell Death can happen via two ways:
respiratory epithelium.  Apoptosis
 The stratified squamous is able to survive circumstances that the  Necrosis
columnar epithelium may have succumbed to.  The two processes vary in their morphology, mechanism, and
roles in physiology and diseases.

Page 3 of 13
Necrosis Nutritional Imbalances
 Accidental and unregulated form of cell death.  Protein-Calorie Deficiencies.
 Results from damage to cell membrane and loss of ion  Vitamin Deficiencies.
homeostasis.  Nutritional Excess.
 Lysozymes digest the cytoplasm and cytoplasmic content leak
which often elicit inflammatory reactions. MORPHOLOGIC CHANGES IN CELL INJURY
 Necrosis is commonly encountered in injuries.

Apoptosis
 Occurs in cells when DNA damage is beyond repair.
 Programmed cell death.
 Nuclear dissolution and cell fragmentation without leaking of the
contents.
 No inflammatory reaction.

 Necrosis is always a pathologic process.


 Apoptosis can be physiologic or pathologic.
 In some cases, a different set of genes can trigger a necrosis-like
mechanism of cell death, it is called NECROPTOSIS.
 All stresses and noxious influences exert their effects first at the
CAUSES OF CELL INJURY molecular or biochemical level.
Oxygen Deprivation  There is a time lag between the stress and the morphologic
 Hypoxia is a deficiency of oxygen. changes.
 It causes cell injury by reducing aerobic oxidative mechanisms.  Ultrastructural and Biochemical changes can be seen in minutes
 Causes of Hypoxia: or hours.
 Ischemia – reduced blood flow (Atherosclerotic plaques)  However, it takes days to weeks to see microscopic and gross
 Inadequate oxygenation of blood (Cardiorespiratory failure) morphological changes.
 Decreased oxygen carrying capacity (Carbon monoxide
Morphologic features of Reversible Injury
Poisoning – formation of irreversible monoxyhemoglobin.)
1.) Cellular Swelling – because of reduced oxidative
phosphorylation, there is no ATP to be used in the Na-K pump,
Physical Injuries
allowing sodium influx with water causing swelling.
 Mechanical Trauma.
2.) Fatty Change – Occurs in hypoxic injuries presents as lipid
 Extreme Temperatures.
vacuoles in the cytoplasm, prominent in cells which are dependent
 Radiation, Pressure, Electric Shock.
on lipid metabolism (e.g. hepatocytes and cardiac myocytes)

Chemical Agents and Drugs


 Can cause cell injury by deranging electrolyte balance.
MORPHOLOGY OF CELLULAR SWELLING
 Poisons, Air Pollutants, High conc. of oxygen.
 Causes some pallor (paleness)
 Increased Turgor
Infectious Agents
 Increase in the weight of the organ
 Virus, Bacteria, Fungi, Tapeworms.
 Microscopically, small clear vacuoles and eosinophilia.

Immunologic Reactions
1.) Plasma Membrane Alteration
 Endogenous self-antigens can be responsible for autoimmune
 Blebbing, Blunting, Loss of Microvilli.
diseases.
2.) Mitochondrial Changes
 Immune reaction to external stimuli can also cause cell injury.
 Swelling and Small Amorphous Densities.
3.) Dilation of the ER
Genetic Derangements
 Detachment of polysomes, presence of intracytoplasmic myelin
 Extra chromosomes (Down Syndrome)
figures.
 Singe Base Pair Substitution (Sickle Cell Anemia)
4.) Nuclear Alterations
 Enzyme defects in inborn errors of metabolisms (PKU)
 Diaggregation of granular and fibrillary elements.
 Accumulation of damaged DNA and misfolded proteins
 All of them can trigger cell death.

Page 4 of 13
 Fatty acids from the cell can be degraded by lipases and combine
with calcium (saponification) to form calcium soaps and
ultimately become calcified.
 Electron Microscopy:
 Discontinuation of the plasma and organelle membranes.
 Dilation of the mitochondria with large amorphouse
densities.
 Amorphous debris, myelin figures, aggregates.
 Nuclear Change:
 Karyolysis – Basophilia of the chromatin and enzymatic
A Normal kidney tubules with viable epithelial cells. degradation of the DNA.
B Early reversible ischemic injury, with surface blebs and  Pyknosis – Nuclear Shrinkage and Increased Basophilia.
increased eosinophilia.
 Karyorrhexis – Pyknotic nucleus undergoes fragmentation.
C Necrotic tubule. (Irreversible)
Loss of nuclei, fragmentation, and cytoplasmic leakage PATTERNS OF NECROSIS
Coagulative Necrosis
 Preserved architecture of the dead tissue.
NECROSIS VS APOPTOSIS  Denatures all proteins including enzymes.
 Since even the cellular enzymes are denatured, the cells are
removed by phagocytosis of leukocytes.
 Ischemia secondary to blood vessel injury is the most common
cause of coagulative in all organs EXCEPT the brain.
 Localized coagulative necrotic area is called an Infarct.

Liquefactive Necrosis
 Digestion of the dead cells.
 Transformation of the solid tissue to liquid viscous mass.
 Bacterial or Fungal infections is the most common cause of
liquefactive necrosis.
 There is accumulation of dead leukocytes or pus.
 Hypoxic death of cells in the CNS is considered liquefactive.

Gangrenous Necrosis
 Not a specific pattern of necrosis.
 Commonly used on the limbs, especially the lower limbs.
 The area usually loses blood supply and undergoes necrosis
Morphologic Changes in Necrosis
(coagulative).
 Result of denaturation of intracellular proteins and enzymatic
 When bacterial infection is superimpoased, there is more
digestion of the lethally injured cell.
liquefactive then coagulative necrosis (wet gangrene).
 Unable to maintain membrane integrity and contents leak out.
 Lysosomes of the cell itself leak their own enzymes combined
Caseous Necrosis
with lysozymes from the leukocytes causes degradation of the
 Tuberculous Infections is the most common cause.
cell.
 “Cheese-like” appearance.
 Increased Eosinophilia: since the proteins are denatured and
 Necrotic area appears as structure less collection of lysed cells
RNA in the cytoplasm is lost, which are basophilic, there is
and amorphous granular debris.
nothing to absorb the blue dye.
 What remains is the leaked cytoplasm which is eosinophilic and  It is usually enclosed within a distinctive inflammatory border,
common appearance of granulomas.
absorbs all the red dye.
 Myelin Figures: Whorled Phospholipid masses derived from
Fat Necrosis
damaged cell membranes.
 Not a specific pattern of necrosis.
 Cytoplasm becomes vacuolated having a moth-eaten
 Refers to focal areas of fat destruction, primarily due to release
appearance.
of pancreatic lipases (Acute Pancreatitis).
 Cells have more glassy homogenous appearance due to lost
glycogen particles.  The lipase splits TAG and Free FA’s combine with Calcium
(Saponification).

Page 5 of 13
 In histologic preparations, it appears as shadowy outlines of  Disruption of protein synthesis apparatus.
necrotic fat cells, with basophilic calcium deposits, surrounded  Accumulation of misfolded proteins in the ER resulting to
by inflammatory reaction. Unfolded Protein Response that can culminate cell death.
 Damage to lysosomal and mitochondrial membrane
Fibrinoid Necrosis resulting to cell death.
 Immune Reactions involving Blood vessels.
 Antigen-antibody complexes that get deposited in the arterial B.) MITOCHONDRIAL DAMAGE
wall. The immune complex leak out with fibrin.
 Bright pink amorphous appearance: Fibrinoid.
 Vasculitis Syndrome.

MECHANISM OF CELL INJURY


1.) Cellular response to injurious stimuli depends on the nature,
duration, and severity of the injury.
2.) The consequences of cell injury depend on the type, state, and
adaptability of the injured cell.
3.) Cell injuries results from different biochemical mechanisms
acting on several essential cellular components.

BIOCHEMICAL MECHANISMS IN RESPONSE TO INJURY:

A.) DEPLETION OF ATP

 Mitochondria can be damaged by:


 Increased Cytosolic Ca++
 ROS (Reactive Oxygen Species)
 Oxygen Deprivation
 There are 3 major consequences of mitochondrial damage:
1. Mitochondrial Permeability Transition Pore
- Formation of this high conductance channel leads to the
loss of the mitochondrial potential.
- Failure of oxidative phosphorylation.
- Progressive depletion of ATP.
- Cyclophilin D – a protein component of the mitochondrial
transition pore, targeted by immunosuppressive drugs (e.g.
Cyclosporin) which prevent opening of the transition pores.
2. Abnormal Oxidative Phosphorylation
- Formation of Reactive Oxygen Species (ROS)
3. Activation of Apoptotic Pathways
 Associated with both hypoxic and chemical injury. - Apoptotic proteins are present in the intermembrane
 Major pathway is oxidative phosphorylation of ADP. space that can trigger cell death when leaked in the cytosol
 Resulting to reduction of oxygen by the ETC. (e.g. Cytochrome C and Caspases).
 Second pathway is glycolytic pathway, ATP generation in the
absence of oxygen via hydrolysis of glycogen stores. C.) INFLUX OF CALCIUM AND LOSS OF CALCIUM HOMEOSTASIS
 Depletion of ATP to 10-15% causes the following:  Cytosolic free Ca++ is normally maintained at very low
 Energy-dependent pumps are reduced, specifically the Na- concentrations ~0.1µmol compared with extracellular levels of
K pump. Without this transport, Na will go in, and K will go 1.3mmol.
out of the cell. Together with Na, water will follow and  Increased intracellular Ca++ causes damage by:
cause cell swelling.  Accumulation of Ca++ in the mitochondria increases its
 Cellular Energy Metabolism is altered, cell shifts first to permeability, resulting to failed ATP generation.
glycolytic pathway, after the depletion of glycogen stores, it  Activates intracellular enzymes (nucleases, proteases, etc.)
shifts to anaerobic glycolysis but results to accumulation of  Activates the caspase cascade system results to apoptosis.
lactic acid.
 Failure of Ca++ pump, causes calcium influx.

Page 6 of 13
D.) ACCUMULATION OF OXYGEN-DERIVED FREE RADICALS Removal of Free Radicals
(OXIDATIVE STRESS)  Free radicals are generally unstable and decay spontaneously.
 Free Radicals are chemical species that have a single unpaired  Antioxidants, such as Vitamin A and E, ascorbic acid, and
electron in an outer orbit. glutathione block free radical formation.
 These unpaired electrons are highly reactive and can modify  Binding of metals to their transport protein, such as Ferritin and
adjacent molecules. Ceruloplasmin, can decrease the reaction of Iron and Copper
 Reactions most of the time are autocatalytic. with Free radicals.
 Reactive Oxygen Species (ROS) are produced normally during  Enzyme located near the generation sites of ROS can help
respiration but degraded and removed by the cellular defense degrade them:
system.  Catalase – degrades H2O2 to H2O and O2.
 Increased Production and Decreased scavenging of ROS can lead  Superoxide Desmutase – degrades Superoxide to
to Oxidative Stress. Hydrogen Peroxide (H2O2). Has 2 forms:
 Oxidative stress has been implicated in variety of pathologic - Manganese SOD: located in the mitochondria.
processes (e.g. Aging, Cancer, Cell Injury, and neurodegenerative - Copper-Zinc SOD: located in the cytosol.
diseases).  Glutathione Peroxidase – catalyzes free radical
 ROS is produced in large amount by neutrophils and breakdown by recycling glutathione. Glutathione serves as
macrophages to kill bacteria. a donor of electron.

Generation of Free Radicals Pathologic Effects of Free Radicals


 Redox Reactions during normal metabolic processes, O2 is 1.) Lipid Peroxidation of the Membrane
reduced by the transfer of four electrons to H2 to generate 2  Double bonds in unsaturated FAs of the membrane are attacked
water molecules. In this process small amounts of partially by O2 derived free radicals (autocatalytic).
reduced intermediates are produced in the forms of: 2.) Oxidative Modification of Proteins
 Hydrogen Peroxide  Formation of disulfide linkages.
 Superoxide Anion  Oxidation of protein backbones.
 Hydroxyl Ions  Disrupt conformational structure of proteins resulting to
 Absorption of radiant energy: Ionizing radiation can convert proteosomal degradation.
water to hydroxyl ions and H free radicals. 3.) Lesions in the DNA
 Inflammation: Leukocytes produce large amounts of ROS,  Single and Double Strand breaks.
carried out by the NADPH oxidase in the plasma membrane.  Formation of adducts
Xanthine Oxidase and other intracellular enzymes can also  Cross-linking
generate ROS.  Malignant transformation.
 Enzymatic Metabolism of Drugs: Can generate free radicals but
not ROS (e.g CCl4 can generate CCl3). E.) DEFECTS IN MEMBRANE PERMEABILITY
 Transition Metals: Iron and Copper can donate free electrons,  Loss of membrane’s selective permeability.
since they need to be reduced to be absorbed (Ferric to Ferrous).
It is explained by Fenton’s Reaction: Mechanism of Membrane Damage:
 Result in ATP depletion and Ca-mediated activation of
phospholipases.
 Can also be a direct damage from bacterial toxins, viral proteins,
 Nitric Oxide: Produced by endothelial cells and macrophages can lytic complement, physical, and chemical agents.
be converted to highly reactive Peroxynitrite Anion.  ROS can cause lipid peroxidation.
 Decreased Phospholipid Synthesis due to defective
mitochondrial function and decreased ATP affecting energy-
dependent biosynthetic pathways.
 Increased Phospholipid Breakdown due to activation of Ca-
dependent phospholipases. Unesterified free fatty acids in the
membrane have a detergent effect.
 Cytoskeletal Abnormalities: An increase in cytosolic calcium can
activate proteases that damage the cytoskeleton and detach
them from the membrane making it susceptible to stretch and
rupture.

Page 7 of 13
Consequences of Membrane Damage:  Mitochondria are usually swollen.
 Mitochondiral Membrane Damage: Opening of the permeability  Endoplasmic Reticulum remains dilated.
pores.  However, if oxygen is restored cell injury is reversible. But if
 Plasma Membrane Damage: Loss of osmotic balance, influx of ischemia persists, it can lead to irreversible cell injury:
fluids, and leak of metabolites.  Severe mitochondrial swelling.
 Injury to Lysosomal Membranes: Since the lysosomes are also  Extensive damage to the plasma membrane (myelin figures).
membrane bound, phospholipases can degrade their membrane  Swelling of the lysosomes.
and leak their enzymes in the cytoplasm.  Death is mainly by necrosis but apoptosis can partially contribute
due to activation of pro-apoptotic molecules from the leaky
F.) DNA AND PROTEIN DAMAGE mitochondria.
 DNA Damage beyond repair results to cell death.  Calcification via calcium soaps may occur.
 Same with proteins, too much misfolded proteins can trigger cell  Hypoxia Inducibe Factor 1: a protective mechanism in response
death. to hypoxic stress that:
 Irreversible due to:  Promote the new formation of blood vessels.
 Mitochondrial dysfunction  Stimulate cell survival pathways.
 Membrane Disturbances (leakages)  Enhance anaerobic glycolysis.

CLINICOPATHOLOGIC CORRELATIONS: Selected examples of Cell 2.) ISCHEMIA REPERFUSION INJURY


Injury and Necrosis  Restoration of blood flow to ischemic tissues can promote
recovery of cells but paradoxically can exacerbate the injury and
1.) ISCHEMIA AND HYPOXIC INJURY cause cell death.
 Ischemia is the most common type of cell injury.  Clinically important in myocardial and cerebral infarctions.
 Results from hypoxia induced by reduced blood flow (most  Mechanisms of new damaging process:
commonly by mechanical obstruction in the arteries)  Oxidative Stress – increased generation of ROS, it can also
 Oxidative phosphorylation and glycogen stores are be due to incomplete reduction of oxygen.
compromised.  Intracellular Calcium Overload – Ca influx can happen
 Ischemia tends to cause more rapid and severe cell and tissue during reperfusion and mediate mitochondrial damage.
injury than hypoxia without ischemia.  Inflammation:
- Danger signals from dead cells. (“Eat me” signals)
Mechanism of Ischemia and Hypoxia - Cytokines secreted by resident immune cells.
- Increased expression of adhesion molecules by hypoxic
parenchymal and endothelial cells can recruit circulating
neutrophils to reperfused tissue.
 Activation of the Complement System, IgM have a
propensity to deposit in ischemic tissues for an unknown
reason, upon reperfusion, complement system can bind to
this antibodies.

3.) CHEMICAL (TOXIC) INJURIES


 A frequent problem in clinical medicine.
 A major limitation to drug therapy.
 Toxic liver injury is the most frequent reason for stopping
therapeutic use or development of drugs.

Direct Toxicity
 Injures the cell directly.
 Mercury Chloride Poisoning, Hg binds to sulfhydryl groups of cell
membrane proteins that can inhibit ion transport. Greatest
 Aside from the previous mentioned mechanism of hypoxia,
damage on cells involved in absorption, excretion, and
cytoskeletons disperse and result in loss of ultrastructural
concentration of chemicals. In HgCl2 poisoning, it affects GIT and
features such as microvilli.
Kidney tissues.
 Formation of membrane blebs.
 Cyanide Poisoning, CN binds to Complex 4 of the ETC inhibiting
 Myelin figures also form from derived degenerating plasma
oxidative phosphorylation inducing direct cell damage.
membrane.

Page 8 of 13
Indirect Toxicity Morphologic Changes in Apoptosis
 Usually accomplished by the Cytochrome P450 in the smooth ER  Cell shrinkage, cell is smaller and cytoplasm becomes dense,
of the liver. organelles become tightly packed.
 Formation of free radicals.  Chromatin Condensation most characteristic feature of
 CCl4 used in cleaning industry is converted by CytP450 to a free apoptosis wherein the chromatin aggregates peripherally.
radical CCl3.  Formation of cytoplasmic blebs and apoptotic body’s extensive
 Acetaminophen, an analgesic drug, is also converted into a toxic surface blebbing results to fragmentation of the cytoplasm.
product during liver detoxification.  Phagocytosis of the apoptotic fragments, rapidly ingested by
macrophages.
APOPTOSIS  Apoptotic cells appear as round or oval masses of intensely
 Tightly regulated suicide program. eosinophilic cytoplasm with fragments of dense nuclear
 Apoptotic cells break up into fragments and form apoptotic chromatin.
bodies.
 Plasma membrane of apoptotic cells remains intact but it is Mechanism of Apoptosis
altered to attract macrophages and phagocytize them before  Apoptosis results from the activation of enzymes called caspases
their contents leak out.  They are cysteine proteases that cleave proteins after aspartic
 Genetically regulated process. residues.
 The process of apoptosis can be divided into two:
Causes of Apoptosis  Initiation Phase – some caspases become catalytically
Physiologic Apoptosis active.
 Destruction of cells during embryogenesis (implantation,  Execution Phase – other caspases trigger degradation of
organogenesis, developmental pathways, and metamorphosis). critical cellular components.
 Involution of hormone dependent tissue upon hormone  Two distinct pathways of caspase activation:
withdrawal:  Mitochondrial (Intrinsic) Pathway
 Endometrial cell breakdown during menstruation  Death Receptor (Extrinsic) Pathway
 Ovarian Follicular Atresia during menopause.
 Prostatic Atrophy after castration. INTRINSIC PATHWAY OF APOPTOSIS
 Cell loss in proliferating cell population  Major pathway.
 Immature lymphocytes in bone and thymus  Results from the increased permeability of the outer membrane.
 B Lymphocytes in germinal centers  Cytochrome C, when released into the cytoplasm from the
 Epithelial cells in intestinal crypts. mitochondria initiates apoptosis.
 Elimination of potentially harmful self-reactive T-lymphocytes
 Death of host cells after it has served its function. Anti-Apoptotic Proteins
 Neutrophil dies after acute inflammation.  BCL2, BCL-XL, MCL1
 Lymphocytes at the end of an immune response.  These reside in the outer membrane of the mitochondrial
membranes, as well as the cytosol and ER Membranes.
Pathologic Apoptosis  These proteins keep the mitochondrial outer membrane
 Apoptosis eliminates cells that are injured beyond repair, impermeable, preventing the leakage of cytochrome c.
without eliciting a host reaction.
 DNA damage from radiation or cytotoxic cancer drugs, the cell Pro-Apoptotic Proteins
triggers intrinsic mechanisms that induce apoptosis.  BAX and BAK
 Accumulation of misfolded proteins that may arise from  These proteins oligomerize within the outer membrane of the
mutations in genes from free radical damage. This can cause mitochondria to promote permeability.
accumulation of misfolded proteins in the ER causing ER Stress.
 Infections induced by viruses can trigger apoptosis. It can also be Sensors
triggered by the host immune response. Cytotoxic T lymphocytes  BAD, BIM, BID, Puma, Noxa (BH3-only proteins)
can induce apoptosis.  Sensors of cellular stress and damage.
 Atrophy after duct obstruction such as calculi or stone
formation in the ducts of the pancreas, parotid, and kidney.  Growth factors stimulate the production of anti-apoptotic
proteins preventing leakage of cytochrome c.
 However, when the DNA is damaged, BH3 proteins can detect it
and activate the pro-apoptotic proteins.
 Anti-apoptotic proteins’ synthesis may decline because of the
relative deficiency of survival signals.

Page 9 of 13
 Once in the cytosol, cytochrome c, binds to a protein called REMOVAL OF THE DEAD CELL
Apoptosis-Activating Factor 1 (APAF1) and form a wheel-like  Phosphatidylserine, a part of the plasma membrane, flips out
hexamer called apoptosome. and expressed outside the plasma membrane, recognized by
 The complex binds to CASPASE-9 and initiates the cascade via many macrophages as an “eat me” signal.
autoamplification, activating the other pro-caspases.  Thrombospondin coats most of the apoptotic bodies which is an
 Proteins such as Smac and Diablo bind and neutralize proteins adhesion glycoprotein recognized by phagocytes.
that function as physiologic inhibitors of apoptosis.  Cq1, a part of the complement system, can naturally coat the
apoptotic bodies for macrophages to recognize them.
EXTRINSIC PATHWAY OF APOPTOSIS
 Initiated by engagement of plasma membrane death receptors CLINICOPATHOLOGIC CORRELATIONS: Apoptosis in Health and Disease.
on a variety of cells. Examples of Apoptosis
 TNF-receptor family, contain a cytoplasmic domain involved in  Growth Factor Deprivation
protein-protein interaction called death domain. - Triggered by intrinsic pathway of apoptosis.
 TNFR1 and Fas (CD95) are best known death receptors. - Hormone-sensitive cells deprived from relevant hormone.
 Fas Ligand (FasL) for Fas receptor is expressed on T cells that - Lymphocyte not stimulated by antigens.
recognize self-antigens. Upon binding of 3 or more FasL to Fas - Neurons deprived of nerve growth factor.
receptors, they are brought together and their death domain  DNA Damage
form an adaptor protein that contains another death domain - Tumor suppressor gene (TP53) triggers apoptosis.
called Fas-associated Death Domain (FADD). - TP53 stimulates the production of pro-apoptotic proteins and
 FADD binds inactive caspase 8 (caspase 10 in humans), when BH3 proteins.
multiple inactive caspase 8 molecules are brought to proximity,  Protein Misfolding
they tend to cleave one another and activate the cascade. - Ubiquinated proteins are target for proteosomal degradation.
 FLIP protein inhibits this pathway of apoptosis by binding to pro- - Accumulation of misfolded proteins can trigger unfolded
caspase 8 rendering it inactive. FLIP is produced by some viruses protein response which activates protein signaling pathway
to protect them from cell death. increasing the production of chaperones that enhance
proteosomal degradation.
- Unable to cope pu with misfolded protein activates the
caspases and induce apoptosis, this is called ER Stress.
 TNF-induced Apoptosis
- FasL on T cells eliminates lymphocytes that recognize self-
antigens.
- Mutations on Fas gene result in autoimmune diseases.
 Cytotoxic T-Lymphocyte Mediated Apoptosis
- CTLs recognize foreign antigens in infected host cells.
- CTLs release perforin molecule that forms pores allowing the
entry of Granzymes (Granule Serine Protease) in the cytosol ti
activate caspases,

Dysregulated Apoptosis
 Defective apoptosis, increased survival.
- TP53 mutations
- Autoimmune disorders
 Increased apoptosis, excessive cell death.
- Neurodegenerative diseases
- Ischemia
 Combined activation of both pathways can be fatal to the cell. - Death of Virus-infected cell

EXECUTION PHASE OF APOPTOSIS


 Two initiating pathways converge to a cascade of caspase
activation (intrinsic – caspase 9; extrinsic – caspase 8 & 10)
 Executioner caspases such as caspase 3 and 6 act on many
cellular components (e.g. it can cleave an inhibitor of a
cytoplasmic DNase making it active).

Page 10 of 13
NECROPTOSIS Steps in Autophagy
 Morphologically and biochemically, resembles necrosis.
 Triggered by genetic programmed signal transduction, like
apoptosis.
 Can be called as programmed necrosis.
 Driven passively by toxic or anoxic injuries.
 It does not result to caspase activation; hence it is sometimes
called caspase-independent programmed cell death.

Mechanism of Necroptosis
 Starts in a manner similar to extrinsic apoptosis via ligand-
receptor. 1. Formation of an isolation membrane called phagophore,
 Necroptosis involves 2 major unique kinases: derived from the ER.
 Repector associated kinase 1 and 3 (RIP1 & RIP3) 2. Elongation of the vesicle.
 When TNF binds to TNFR1, it recruits RIP1 and RIP3 molecules 3. Maturation of the autophagosome.
into a multiprotein complex that also contains caspase 8. 4. Fusion with the lysosomes.
5. Degradation of the content.
 However, the caspase is not activated, but the progress goes like
that of necrosis (ROS, lysosomal membrane destruction)
Roles of Autophagy
 Pathologic Necroptosis: Steatohepatitis, acute pancreatitis,
 Cancer: Both promote and inhibit cancer growth.
reperfusion injury, and neurodegenerative diseases.
 Neurodegenerative disorders:
 Physiologic Necroptosis: Formation of the mammalian bone
growth plate.  Alzheimer’s – formation of autophagosomes.
 Necroptosis also acts as a backup mechanism in host defense  Huntington’s D – Mutant ‘huntingtin’ (impaired
against viral infections that bypass the caspase system such as autophagy)
the cytomegalovirus.  Infection
 Shigella sp. And HSV-1
PYROPTOSIS  Macrophage deletion of ATG5, increases susceptibility to
 Accompanied by release of fever inducing cytokine IL-1 forming tuberculosis.
a multiprotein complex called inflammasome.  Inflammatory Bowel Diseases
 Inflammasomes activate caspase 1 (also known as Interleukin-1β  Crohn’s Disease
converting enzyme) which cleaves IL-β1 making it biologically  Ulcerative Colitis
active.
 Caspase 1 and caspase 11 induce cell death. INTRACELLULAR ACCUMULATION
 Unlike apoptosis, this present with cell swelling, loss of plasma  Intracellular accumulation of abnormal amounts of various
membrane integrity, and release of inflammatory mediators. substances may be harmless or associated with varying degrees
of injury.
AUTOPHAGY  Four main pathways of abnormal intracellular accumulation:
 A process where in a cell eats its own components. 1.) Inadequate Removal of a substance
 Involves the delivery of cytoplasmic materials to the lysosome  Defects in mechanisms of packaging and transport
for degradation  Ex: Hepatic Steatosis
 Three types:
 Chaperone-mediated 2.) Accumulation of an abnormal endogenous substance
- Direct translocation across the lysosomal membrane.  A result of genetic or acquired defects in its folding, packaging,
 Microautophagy and transport.
- Inward invagination of lysosomal membrane.  Ex: α1-antitrypsin
 Macroautophagy
- Major form of autophagy. 3.) Failure to degrade a metabolite
- Transport of portions of cytosol in a double membrane  Due to enzyme deficiencies
bound autophagic vaguole (autophagosome).  Ex: Storage diseases
 Autophagy is an evolutionarily conserved survival mechanisms:
 Used in states of nutrient deprivation 4.) Deposition and accumulation of an abnormal exogenous subs.
 Cells live by cannibalizing or recycling themselves  Cell has neither the enzymatic machinery to degrade the
 Controlled by the AGTS gene. substance, nor the ability to transport it to the other site
 Ex: Accumulation of Carbon or Silica particles

Page 11 of 13
LIPIDS  Glycogen storage diseases or Glycogenoses have enzymatic
Steatosis (Fatty Changes) defects or storage defect.
 Describe abnormal accumulations of triglycerides within
parenchymal cells. PIGMENTS
 Seen mainly in liver due to its site of fat metabolism. Exogenous Pigments
 Caused by toxins, protein malnutrition, diabetes, obesity, and  Common exogenous pigment is CARBON.
anorexia.  Accumulation of this pigment blackens the tissue (Anthracosis).
 Coal Worker’s Pneumoconiosis.
Cholesterol and Cholesterol Esters  Tattooing is also a form of localized exogenous pigmentation
 Cholesterol is used in constructing the cell membrane but it is where in the ink is being phagocytosed by dermal macrophages.
sometimes seen in pathologic conditions:
 Atherosclerosis – plaque formation in the intimal layer and Endogenous Pigments
smooth muscle layer of arteries filled with lipid vacuoles Lipofuscin
producing yellow cholesterol-laden arethoma.  An insobluble pigmen, also known as lipo-chrome or wear-and-
 Xanthoma – intracellular accumulation of cholesterol within tear pigment.
macrophages, seen in hyperlipidemic states.  Derived through lipid peroxidation of polyunsaturated lipids.
 Cholesterolosis – accumulation of cholesterol-laden  Not injurious but an indication of cell injury and lipid
macrophages in the gall bladder. peroxidation.
 Niemann-Pick disease type C – lysosomal storage disorder,  Appears as brown lipid or brown in actual slides.
resulting to cholesterol trafficking and accumulation in different
organs. Melanin
 Brown-black pigment.
PROTEINS  Clinically significant in PKU, deficient homogentisic acid oxidase
 Usually appear as rounded, eosinophilic droplets, vacuoles, or leading to alkaptonuria.
aggregates in the cytoplasm.  Pigments deposit in the skin, connective tissue, and cartilage, a
 In electron microscopy: Amorphous, fibrillary, or crystalline. condition known as ochronosis.
 Caused by:
 Reabsorption droplets in the PCT Hemosiderin
- Seen in renal disease associated with proteinuria.  Hgb-derived golden yellow to brown granular or crystalline
- During heavy protein leakage, protein appears as pink, pigment.
hyaline substance seen in tubular cells.  One of the major storage forms of Iron.
 Russell Bodies  In systemic or local excess iron, ferritin forms hemosiderin.
- When ER becomes hugely distended, producing large,  Hemosiderin represents aggregates of ferritin micelles.
homogenous, eosinophilic inclusions.  Local excess example is bruise.
 Defective intracellular transport  Systemic overload of iron, hemosiderin can be deposited in
- In α anti trypsin deficiency, there is slow protein folding organs causing a condition called hemosiderosis, caused mainly
leading to partially folded ones. by:
 Accumulation of cytoskeletal proteins  Increased absorption of dietary ion.
- Neurofibillary tangles  Hemolytic anemia
- Alcoholic hyaline is eosinophilic on cirrhotic patients.  Repeated blood transfusion
 Aggregation of abnormal proteins.
- Amyloidosis. PATHOLOGIC CALCIFICATION
- Proteinopathies or Protain-aggregation disorders.  Abnormal tissue deposition of calcium salts together with small
amounts of other minerals.
HYALINE CHANGE  Can be divided into Dystrophic or Metastatic.
 Alterations within the cells or in the extracellular space.
 Gives homogenous, glassy, pink appearance. Dystrophic Calcification
 Descriptive rather than specific.  Occurs despite normal serum levels of calcium.
 Encountered in areas of necrosis, whether they are coagulative,
GLYCOGEN caseous, or liquefactive, also in fat necrosis.
 Seen in patients affected by enzymatic or storage disorders of  Calcium appears as fine, white granules or clumps often felt as
glucose metabolism. gritty deposits.
 Diabetes Mellitus, glycogen is found on renal tubular cells, liver  Ex: Valvular Stenosis
cells, islet of Langerhans, cardiac myocytes.

Page 12 of 13
Metastatic Calcification
 Occurs in tissue due to disturbances in calcium metabolism.
 Mineral salts ‘usually’ cause no clinical dysfunction.
 Can be cause by:

1.) Increased secretion of PTH


 Hyperparathyroidsim
 Increased bone resportion of Calcium

2.) Resorption of Bone Tissue


 Secondary to primary bone tumors.

3.) Vitamin-D related disorders


 Sarcoidosis, Idiopathic Hypercalcemia.

4.) Renal Failure


 Causes phosphate retention. DEFECTIVE PROTEIN HOMEOSTASIS
 Usually secondary to hyperparathyroidism.  Protein homeostasis is maintained by 2 mechanisms:
 Maintain in proper folding state
CELLULAR AGING  Degrade misfolded ones by autophagy-lysosome system.

DEREGULATED NUTRIENT SENSING


 Eating less, increases longevity.
 Caloric restriction increases life span.
 IGF-1 mimics insulin hormone, informing the cell the availability
of glucose promoting anabolic state.
 Sirtuins are family of NAD-dependent protein deacetylases that
are designed to adapt to bodily functions to various
environmental stresses, including food deprivation.

 Result of a progressive decrease in cell function and viability.


 Caused by:
 Genetic Abnormalities
 Accumulation of cellular and molecular damage.

DNA DAMAGE
 Defective DNA repair mechanisms.
 Accumulation of chromosomal damages.

CELLULAR SENSENCE
 Limited capacity for replication, after a fixed number of divisions,
the cell arrests in a terminally non-dividing state.
 Telemere Attrition – progressive shortening of telomeres result “Pyryzy napasirossa vokemilzi, va daorunta nelli
to cell cycle arrest. qringaomna nojo zalari”
 Activation of Tumor Suppresion Genes – CKN2A locus encode (They will purify nonbelievers by the thousands, burning
TSGs that help regulate cell senescence. their sins and flesh away)

Page 13 of 13

You might also like