Fact-checked by Grok 2 weeks ago

Cell damage

Cell damage, also known as cellular injury, refers to the structural and functional alterations in cells resulting from exposure to injurious stimuli that disrupt normal , potentially leading to adaptive responses, reversible changes, or irreversible damage culminating in . The causes of cell damage are diverse and encompass both extrinsic and intrinsic factors, including oxygen deprivation through or ischemia, physical agents such as mechanical , extreme temperatures, or , chemical and toxic substances like drugs or environmental toxins, infectious agents including and viruses, immunological reactions such as autoimmune responses or , genetic derangements or mutations, nutritional imbalances like deficiencies, and the progressive effects of aging. These stressors initiate a cascade of biochemical events that can overwhelm cellular repair mechanisms, with the severity and duration of exposure determining the outcome. At the molecular level, cell damage primarily arises from four fundamental biochemical mechanisms: depletion of adenosine triphosphate (ATP), which impairs energy-dependent processes; permeabilization and dysfunction of cell membranes, leading to loss of integrity; disruption of key biochemical pathways, including protein synthesis and ion transport; and direct damage to DNA, often from reactive oxygen species (ROS) or other genotoxic agents. Additional critical processes include mitochondrial dysfunction, which amplifies ROS production and initiates permeability transitions; dysregulation of intracellular calcium (Ca²⁺) homeostasis, activating destructive enzymes like calpains and phospholipases; and oxidative stress, where excess ROS damages lipids, proteins, and nucleic acids. These mechanisms often interconnect, with early events like ATP loss exacerbating membrane permeability and calcium influx, marking a pivotal shift from reversible to irreversible injury. Morphologically, reversible cell damage manifests as cellular swelling (hydropic degeneration), fatty change (steatosis), and vacuolation of the cytoplasm, changes that resolve if the stressor is removed and homeostasis restored. In contrast, irreversible damage features severe nuclear alterations—pyknosis (shrinkage and basophilia), karyorrhexis (fragmentation), and karyolysis (dissolution)—along with plasma membrane rupture and mitochondrial swelling, progressing to cell death. Cell death assumes two primary forms: necrosis, an uncontrolled process involving cell swelling, organelle breakdown, and inflammatory response, with subtypes including coagulative (preservation of tissue architecture, as in ischemic infarcts), liquefactive (enzymatic digestion, common in bacterial infections), caseous (cheesy debris, seen in tuberculosis), and fat necrosis (saponification from lipases); or apoptosis, a programmed, energy-dependent mechanism characterized by cell shrinkage, chromatin condensation, and formation of apoptotic bodies without inflammation, often triggered by DNA damage or developmental signals. In response to milder or chronic stressors, cells may undergo adaptive changes to maintain function, such as (increased cell size, e.g., in under pressure overload), (increased cell number, e.g., endometrial proliferation), (reduced cell size, e.g., from disuse), or (replacement of one cell type by another, e.g., squamous metaplasia in smokers' airways). These adaptations represent a spectrum of cellular , but failure to adapt or excessive injury contributes to tissue dysfunction and diseases ranging from ischemia-related infarcts to chronic degenerative conditions.

Causes

Physical and Chemical Causes

Cell damage can arise from physical agents that exert mechanical or energetic forces on cellular structures, as well as chemical agents that interact molecularly with cellular components to disrupt normal function. These abiotic factors initiate injury through direct interactions, such as structural disruption or biochemical interference, independent of oxygen availability or biological invaders. Common mechanisms include membrane permeabilization, protein denaturation, DNA strand breaks, and oxidative stress via reactive species generation, leading to impaired homeostasis and potential progression to cell death if unresolved. Physical trauma represents a primary cause, involving blunt or penetrating forces that mechanically rupture membranes and organelles, often resulting in immediate or secondary ischemic effects from vascular disruption. For instance, crushing injuries to tissues can cause epithelial and erythrocyte breakdown, converting to in bruised areas. Mechanical stress from shearing or pressure similarly damages the , altering cell shape and motility; traumatic exemplifies this, where blunt force against over bony prominences induces rupture and release. Extreme temperatures further contribute to physical damage by altering biomolecular stability. , as in burn injuries, denatures proteins and enzymes while disrupting lipid bilayers, leading to membrane instability and leakage; severe burns can thus cause widespread in affected tissues. Conversely, induces and ischemia alongside ice crystal formation during freezing, which physically pierces membranes and organelles, as seen in resulting in dry . Radiation exposure constitutes another key physical agent, with ultraviolet (UV) and ionizing types eliciting distinct yet overlapping damages. UV radiation primarily generates cyclobutane pyrimidine dimers and 6-4 photoproducts in DNA, impeding replication and transcription while provoking oxidative stress that affects proteins and lipids; prolonged skin exposure, for example, leads to mutations and immunosuppression. Ionizing radiation, such as X-rays, directly ionizes atoms to produce DNA double-strand breaks and indirectly generates reactive oxygen species (ROS) that oxidize cellular macromolecules, compromising membrane integrity and mitochondrial function. Chemical agents damage cells by binding to or reacting with key biomolecules, often through covalent modification or enzyme inhibition. Toxins like cyanide exemplify this by tightly binding the heme iron in cytochrome c oxidase (Complex IV) of the mitochondrial electron transport chain, halting aerobic respiration and causing rapid ATP depletion; acute poisoning thus leads to histotoxic anoxia in high-oxygen tissues like the brain and heart. Therapeutic drugs can also induce damage at supratherapeutic doses via reactive metabolite formation. Acetaminophen overdose, a leading cause of acute liver failure, produces the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI), which depletes glutathione and forms protein adducts, triggering mitochondrial dysfunction and oxidative stress in hepatocytes. Environmental pollutants, including heavy metals such as lead, bind sulfhydryl groups on enzymes and generate ROS, disrupting antioxidant defenses and causing membrane peroxidation; chronic lead exposure notably forms intranuclear inclusions in renal tubular cells, impairing filtration and inducing oxidative injury. Certain chemicals promote , oxidizing polyunsaturated fatty acids in membranes to form toxic aldehydes that propagate chain reactions. Ethanol consumption, for example, enhances this in liver cells by boosting ROS from metabolism, leading to and ; studies in humans confirm elevated lipid peroxides and conjugated dienes in , correlating with disease severity. These mechanisms collectively underscore how physical and chemical insults initiate cellular disequilibrium through targeted molecular disruptions.

Biological and Hypoxic Causes

Biological agents, such as and viruses, induce cell damage through direct invasion or release. Gram-negative bacteria produce endotoxins, lipopolysaccharides in their cell walls that trigger intense inflammatory responses, leading to fever, vascular changes, and cellular disruption. Viral pathogens exacerbate this by hijacking host cellular machinery; for instance, HIV-1 commandeers ATPases and involved in protein trafficking to facilitate replication, thereby dysregulating and promoting immune evasion and cell death. Pathogen invasion further disrupts host by impairing mechanisms and altering metabolic pathways, resulting in genomic instability and tissue injury. Immune-mediated processes contribute to cell damage via aberrant responses to biological threats. Autoantibodies can directly attack cell surfaces or form immune complexes that activate complement and receptors, inciting and tissue destruction in autoimmune conditions. Exaggerated immune activation, such as cytokine storms, amplifies this harm by recruiting effector cells that release pro-inflammatory mediators, causing widespread endothelial and parenchymal damage. Nutritional deficiencies from biological stressors, like chronic infections or malabsorption, impair cellular integrity by hindering synthesis and repair processes. Micronutrient shortages, including vitamins A, C, and E, lead to DNA damage, mitochondrial dysfunction, and increased telomere shortening, compromising cell cycle regulation and energy production. For example, vitamin C deficiency disrupts collagen synthesis and antioxidant defenses, heightening susceptibility to oxidative stress and cellular breakdown in connective tissues. Hypoxic conditions arise from oxygen shortages that cripple aerobic metabolism, encompassing ischemia (reduced blood flow), (complete oxygen absence), and from high-altitude exposure or . In ischemia, such as during , neuronal cells experience rapid oxygen deprivation, triggering excitotoxic glutamate release and calcium overload that exacerbate energy failure. High-altitude induces neuronal injury via overactivation of NMDA receptors and loss of calcium homeostasis control. similarly limits oxygen delivery to tissues, mimicking ischemic effects and promoting cellular stress. The core mechanism in involves mitochondrial dysfunction, where low oxygen halts activity, causing ATP depletion and failure of ion pumps like Na+/K+-ATPase. This leads to ionic imbalances, permeabilization, and swelling, progressing to irreversible if oxygen is delayed. In biological contexts, pathogens can compound hypoxic injury by inflaming vasculature and further restricting .

Cellular Targets

Structural Components

Cell damage often begins with disruptions to the plasma membrane, the primary barrier separating the intracellular environment from the . Injury to this structure can lead to increased permeability, allowing uncontrolled influx of ions such as sodium (Na⁺) and calcium (Ca²⁺), while permitting efflux of (K⁺), resulting in an ionic imbalance that drives cellular swelling and dysfunction. This Na⁺/K⁺ disequilibrium disrupts the essential for cellular , as the Na⁺/K⁺-ATPase pump struggles to maintain ion concentrations amid the leak. In early stages of , plasma membrane blebbing occurs, where protrusions form due to cytoskeletal detachment and hydrostatic pressure imbalances, signaling initial compromise without immediate rupture. Organelles, as specialized intracellular compartments, are also vulnerable to structural alterations during cell damage. Mitochondria frequently undergo swelling, characterized by matrix expansion and cristae disorganization, which impairs their role in energy production and initiates release of pro-death signals. The endoplasmic reticulum (ER) experiences dilation of its cisternae, leading to unfolded protein accumulation and responses that exacerbate injury propagation. Lysosomes, containing hydrolytic enzymes, may rupture, spilling their contents into the and triggering autodigestion of cellular components. The provides mechanical support and maintains cell shape, but damage disrupts its filamentous networks. , composed of polymers, depolymerize under , compromising intracellular transport and structural integrity. filaments, forming dynamic networks, fragment or reorganize abnormally, leading to loss of and , which contributes to overall morphological instability. Specific examples illustrate these structural impacts. induces in plasma membranes, where oxidize polyunsaturated fatty acids, creating chain reactions that compromise and integrity. In bacterial infections, pore-forming toxins such as those from or perforate host cell membranes, forming conductive channels that facilitate ion dysregulation and rapid structural failure.

Functional Molecules

Cell damage often targets functional molecules such as proteins, lipids, and carbohydrates, leading to disruptions in enzymatic activities, , and signaling pathways essential for cellular . These alterations impair the cell's ability to perform metabolic processes, respond to stimuli, and maintain balances, ultimately contributing to injury or death. Proteins are highly susceptible to denaturation, a process where stressors like disrupt their native three-dimensional structure, resulting in loss of enzymatic and structural functions. In intact hepatocytes exposed to at 45°C, approximately 4-7% of proteins denature, inactivating critical components and triggering the synthesis of heat shock proteins as a protective response. Denaturation extends across cellular compartments, including mitochondria and microsomes, where it compromises energy production and machinery. Similarly, protein misfolding under cellular stress, such as during , leads to aggregation of unfolded polypeptides, overwhelming the unfolded protein response and pathways. In conditions like protein misfolding diseases, impaired heat shock factor 1 (Hsf1) activity exacerbates misfolding of proteins such as polyglutamine-expanded , disrupting cytosolic signaling and promoting through endoplasmic reticulum stress. Enzymatic inactivation further compounds these effects; for instance, toxins bind to sulfhydryl groups on enzymes, halting catalytic activity— is particularly vulnerable, as its inhibition blocks Krebs cycle progression and ATP synthesis. Lipids in cellular membranes undergo peroxidation, primarily affecting polyunsaturated fatty acids, which generates hydroperoxides and reactive aldehydes like (4-HNE) and (MDA). These products reduce by altering lipid packing and slowing lateral diffusion, while increasing permeability to ions and macromolecules, thereby collapsing electrochemical gradients critical for signaling. Lipid peroxidation also propagates chain reactions that modify adjacent and proteins, amplifying oxidative damage and contributing to ferroptosis-like pathways. oxidation yields oxysterols, such as 7-ketocholesterol, which integrate into membranes and induce domain formation, enhancing and endothelial barrier dysfunction. In arterial cells, these oxidation products promote in and endothelial cells by perturbing trafficking and intracellular calcium homeostasis, fostering inflammatory signaling via activation. Carbohydrates and related glycoconjugates face damage through non-enzymatic glycation, especially in hyperglycemia, where elevated glucose reacts with amino groups on proteins to form advanced glycation end products (AGEs). These AGEs create irreversible cross-links in extracellular matrix components like collagen and elastin, increasing tissue rigidity and impairing remodeling processes that support cellular adhesion and migration. Glycation activates receptors for AGEs (RAGE) on nearby cells, triggering oxidative stress and inflammatory cascades that further disrupt matrix integrity and intercellular signaling. In diabetic conditions, this leads to vascular stiffening and reduced elasticity, indirectly affecting cellular nutrient exchange and wound healing. Representative examples illustrate these molecular disruptions in pathological contexts. Arsenic poisoning exemplifies enzymatic inactivation, as binds to groups in and succinic , inhibiting mitochondrial respiration and elevating , which culminates in widespread cellular energy failure and . In , lipid modifications such as LDL oxidation produce oxLDL that macrophages internalize via scavenger receptors, forming foam cells laden with esters; this triggers chronic , endothelial , and plaque instability through release and defective cholesterol efflux. While membranes serve as primary carriers for these functional lipids, their peroxidation-induced alterations underscore the interconnectedness of molecular damage and cellular dysfunction.

Morphological Types of Damage

Reversible Damage

Reversible cell damage refers to early-stage cellular alterations induced by injurious stimuli that can be fully restored if the stressor is removed before progression to irreversible injury, preserving cellular function and structure without nuclear pyknosis, , or . These changes primarily involve disruptions in ion homeostasis and , allowing cells to recover through restoration of ATP levels and normalization of integrity. Common in mild hypoxic, toxic, or metabolic insults, reversible damage underscores the adaptive capacity of cells to sublethal stress, with key examples including cellular swelling and fatty change. Cellular swelling, also termed hydropic change or cloudy swelling, manifests as an increase in cell volume due to intracellular accumulation, imparting a pale, vacuolated, and cloudy appearance to affected tissues under light microscopy. The primary mechanism involves ATP depletion from mild ischemia or , which impairs the Na+/K+-ATPase pump, leading to sodium influx, loss of , and subsequent osmotic entry into the and organelles like mitochondria and . This results in early plasma membrane blebbing and increased permeability without nuclear involvement, creating clear vacuoles that displace organelles but remain reversible upon reoxygenation and ATP replenishment. A representative example is transient renal tubular swelling during brief renal ischemia, where cells regain normal morphology after blood flow restoration, preventing progression to . Fatty change, or steatosis, is characterized by the reversible accumulation of triglycerides within cytoplasmic lipid droplets, most prominently in hepatocytes but also in cardiac and renal cells, due to imbalances in lipid synthesis, uptake, or oxidation. Mechanisms include direct toxic effects on mitochondrial beta-oxidation or enhanced fatty acid mobilization from adipose tissue, often without significant osmotic shifts but involving early endoplasmic reticulum stress. In the liver, chronic alcohol exposure exemplifies this, as ethanol metabolism generates NADH excess that inhibits fatty acid oxidation while promoting lipogenesis, leading to macrovesicular steatosis that resolves within weeks of abstinence through normalized lipid export via lipoproteins. Unlike swelling, fatty change does not typically alter cell volume but imparts a foamy cytoplasmic appearance, reversible as long as hepatocellular integrity is maintained. Overall, both forms of reversible damage stem from osmotic imbalances in swelling and metabolic derangements in , highlighting the threshold beyond which early alterations escalate to permanent harm if the persists.

Irreversible Damage

Irreversible cell damage represents the point of no return in cellular , where structural and functional failures preclude , unlike earlier reversible changes such as cellular swelling that serve as warnings. These alterations culminate in collapse and loss of viability, often observed in severe insults like prolonged ischemia or . Mitochondrial dysfunction is a hallmark of irreversible damage, initiated by the opening of the permeability transition pore, which increases inner membrane permeability and leads to mitochondrial swelling, production, and depletion of NAD+. This event dissipates the proton gradient essential for ATP synthesis, committing the to energy failure. Nuclear changes further signify irreversible commitment to , beginning with , characterized by condensation and nuclear shrinkage due to severe protein denaturation. This progresses to , where the fragmented breaks into irregular clumps, reflecting advanced DNA degradation. Ultimately, occurs as enzymatic dissolution fully erodes the nuclear structure, marking complete loss of genetic integrity. Plasma membrane rupture accompanies these events, starting with extensive blebbing—irregular protrusions formed by cytoskeletal and intracellular imbalances. As fails, the membrane ruptures, allowing uncontrolled ion influx and cellular contents to spill, resulting in explosive and . In severe , such as during , irreversible damage manifests as eosinophilic cytoplasm from denatured proteins, staining brightly pink under light microscopy due to ribosomal loss. exposure, like (), induces nuclear clumping with basophilic debris, evident in hepatic where aggregates form dark, irregular masses.

Forms of Cell Death

Necrosis

Necrosis represents an uncontrolled form of cell death triggered by severe exogenous injuries, such as ischemia, toxins, or infections, resulting in the loss of cellular and plasma membrane integrity. This process differs from , which is a regulated, non-inflammatory mechanism of cell elimination. In necrosis, cellular contents spill into the , provoking a robust inflammatory response that distinguishes it as a pathological event rather than a programmed one. The primary mechanisms of necrosis involve progressive damage to the plasma membrane, often initiated by ATP depletion and ion dysregulation, leading to cellular swelling (oncosis) and eventual rupture. This membrane breakdown allows uncontrolled influx of water and ions, followed by the release of lysosomal enzymes such as proteases and hydrolases, which autodigest cellular components and exacerbate tissue destruction. The ensuing inflammatory cascade is driven by damage-associated molecular patterns (DAMPs), including and ATP, which activate immune pathways like the and promote release, such as interleukin-1β (IL-1β). Necrosis manifests in distinct morphological types based on the underlying insult and tissue involved. Coagulative necrosis, the most common form, features protein denaturation that preserves the basic tissue architecture, typically occurring in ischemic conditions outside the central nervous system; for instance, myocardial infarction leads to coagulative necrosis of cardiomyocytes, where nuclei fade and cytoplasm becomes eosinophilic within hours of occlusion. Liquefactive necrosis, in contrast, results from enzymatic liquefaction of tissue, often in bacterial infections or brain ischemia, transforming solid parenchyma into a viscous pus-like material; a classic example is brain abscess formation secondary to bacterial invasion, where neutrophils and microbes digest neural tissue. Fat necrosis occurs when lipases released from damaged pancreatic tissue or abdominal trauma hydrolyze neutral fats into fatty acids, which combine with calcium to form soaps (saponification), leading to chalky white areas in adipose tissue, as seen in acute pancreatitis. Gangrenous necrosis extends these patterns to extremities, combining ischemic coagulative changes with superimposed infection in "wet" gangrene, as seen in diabetic limb ischemia where vascular compromise and bacterial overgrowth cause rapid tissue putrefaction. Caseous necrosis presents as amorphous, cheese-like debris within granulomas, characteristically in tuberculosis, where Mycobacterium tuberculosis infection induces central acellular necrosis surrounded by epithelioid cells. The consequences of include widespread breakdown and a potent inflammatory that recruits to the site, amplifying through further release and . In infectious contexts, this neutrophil influx can culminate in formation, encapsulating the necrotic focus to contain spread, though it may also propagate if unchecked. Overall, necrotic serves as a nidus for secondary complications, underscoring the need for prompt intervention to mitigate .

Programmed Cell Death

Programmed cell death (PCD) encompasses genetically regulated processes that eliminate superfluous, damaged, or harmful cells in a controlled manner, maintaining tissue homeostasis and preventing inflammation. Unlike accidental cell death, PCD pathways such as apoptosis ensure orderly dismantling of cellular components, followed by rapid phagocytic clearance, which suppresses immune activation. These mechanisms are crucial during embryonic development, immune system maturation, and tumor suppression, where dysregulated PCD contributes to diseases like cancer and autoimmunity. Apoptosis, the prototypical form of PCD, proceeds via two primary pathways: the intrinsic (mitochondrial) pathway and the extrinsic (death receptor) pathway. In the intrinsic pathway, cellular stresses like DNA damage or growth factor deprivation trigger mitochondrial outer membrane permeabilization (MOMP), mediated by pro-apoptotic Bcl-2 family proteins such as Bax and Bak, while anti-apoptotic members like Bcl-2 inhibit this process. This releases cytochrome c, forming the apoptosome with Apaf-1 and procaspase-9, which activates executioner caspases (e.g., caspase-3 and -7) to cleave substrates, leading to morphological changes including chromatin condensation and DNA fragmentation into 180-200 bp multiples, visible as DNA laddering on electrophoresis. The extrinsic pathway, activated by extracellular signals, involves death receptors like Fas (CD95) or TNF receptors binding ligands, recruiting adaptor proteins (e.g., FADD) to activate initiator caspase-8, which either directly activates executioner caspases or amplifies via the mitochondrial route through Bid cleavage. Caspase activation is a central, irreversible step in both pathways, orchestrating nuclear fragmentation, cytoskeletal breakdown, and apoptotic body formation without leakage of intracellular contents. Beyond , other PCD modalities include , necroptosis, and , each with distinct molecular regulators and contexts. involves lysosomal degradation of cellular components via formation, primarily serving as a under (e.g., deprivation) but can culminate in when excessive, suppressing through contained self-digestion. Necroptosis, a regulated necrosis variant, is triggered by /RIPK3/MLKL signaling when is inhibited, forming a necrosome that compromises in a programmed fashion, yet it differs from by promoting if not cleared efficiently. is an iron-dependent process characterized by and 4 () inhibition, leading to accumulation and plasma rupture; it plays roles in suppressing tumorigenesis but can drive in states. These pathways often intersect, with modulating sensitivity and necroptosis serving as a backup to . PCD's non-inflammatory nature stems from exposure of "eat-me" signals like on apoptotic bodies, facilitating swift engulfment by , which further releases cytokines. In embryogenesis, apoptosis sculpts structures, such as digit formation via interdigital cell elimination, and in the , it ensures thymocyte selection: double-positive thymocytes with self-reactive T-cell receptors undergo negative selection via intrinsic to prevent . For cancer suppression, p53-induced eliminates genomically unstable cells, while evasion of PCD (e.g., overexpression) promotes tumor survival. In pathology, exemplifies PCD's dual role; in hepatocytes, as in hemochromatosis, triggers and ferroptotic death, contributing to liver without initial but potentially escalating to broader damage if unresolved.

Repair and Recovery

Regeneration

Regeneration refers to the process by which damaged tissues restore their original architecture through the and of surviving , particularly in response to or cell damage. This capacity varies among cell types and is most pronounced in tissues with high proliferative potential, enabling full functional recovery without scarring. In labile cells, which continuously divide throughout life, regeneration occurs rapidly via ongoing activity, allowing seamless replacement of lost cells. Labile cells, such as those in the skin and gut , maintain a constant turnover through stem cell-driven proliferation, facilitating complete regeneration after damage. For instance, epidermal stem cells in the basal layer and hair follicles activate upon injury, migrating and dividing to reepithelialize wounds and restore the skin barrier. Similarly, intestinal epithelial stem cells in crypts continuously renew the lining, enabling quick repair of superficial erosions. Stable cells, like liver hepatocytes, normally exhibit limited division but possess the ability to undergo compensatory following significant , thereby regenerating tissue mass and function. In the liver, this process restores lobule architecture after partial resection, with hepatocytes re-entering the to proliferate synchronously. Key mechanisms of regeneration involve the of resident or , stimulation by such as (EGF) and hepatocyte growth factor (HGF), and the re-entry of quiescent cells into the . EGF promotes keratinocyte proliferation in skin repair by binding to , enhancing migration and , while HGF initiates hepatocyte priming via c-Met signaling, activating pathways like MAPK for mitotic progression. These factors are released or upregulated post-, coordinating resolution with tissue rebuilding. niches, such as those in the liver's canals of Hering or skin's bulge region, provide progenitors that amplify regeneration when mature cells are depleted. A prominent example is liver lobule regeneration after partial , where up to two-thirds of the organ is removed; HGF surges within hours, followed by EGF-driven waves of division, restoring mass within weeks without under normal conditions. In epidermal , stem cells from wound margins and appendages, stimulated by EGF, proliferate to cover defects, forming a new stratified that integrates with surrounding tissue. In permanent tissues like neurons or , regeneration is limited, often leading to by non-functional rather than original restoration.

Replacement and Scarring

When regeneration of damaged tissue is not possible, particularly in permanent or post-mitotic cells that lack the ability to proliferate, the body initiates a repair process involving with non-functional , resulting in scarring or . , such as neurons in the and cardiomyocytes in , are unable to divide due to their terminally differentiated state, limiting regenerative potential and leading to formation () in the or fibrotic in the heart. This contrasts with labile tissues like , where regeneration can restore original architecture. The mechanisms of scarring begin with the proliferation of s, which migrate to the injury site and differentiate into myofibroblasts, depositing excessive components, primarily types I and III. This process is preceded by the formation of , characterized by —new blood vessel formation driven by factors like (VEGF)—to support nutrient delivery and fibroblast activity. Over time, the provisional granulation tissue matures into a dense, avascular through collagen cross-linking and remodeling by matrix metalloproteinases, stabilizing the wound but distorting tissue architecture. Scarring leads to several adverse consequences, including tissue contracture due to myofibroblast-mediated , which can reduce organ flexibility and function. Impaired organ performance arises from the replacement of functional with rigid fibrous tissue; for instance, in the liver, progressive culminates in , where scar tissue disrupts architecture and vascular flow, causing and synthetic dysfunction. Representative examples include the myocardial following , where necrotic cardiomyocytes are replaced by a -rich fibrotic area that prevents rupture but increases stiffness, contributing to . In skin, formation exemplifies excessive scarring, where fibroblasts produce hypertrophic nodules that extend beyond the original wound boundaries, often in response to and leading to cosmetic and functional deformities.

Biochemical Mechanisms

Metabolic Disruptions

Metabolic disruptions represent a fundamental aspect of cell damage, where alterations in energy production and maintenance of cellular lead to progressive functional impairment and potential . These changes primarily stem from interference with ATP synthesis and subsequent failure of energy-dependent processes, distinguishing them from other biochemical insults by their direct impact on core metabolic pathways. ATP depletion is a hallmark of metabolic disruption, occurring rapidly when oxygen supply is compromised or mitochondrial function is inhibited. In , cells shift to , which is inefficient and cannot sustain ATP levels, leading to a decline of up to 30% within minutes. Toxins such as exacerbate this by binding to in the , blocking and halting ATP production almost immediately. For instance, during ischemia, ATP levels can drop by 62% within 15 minutes, impairing cellular viability before complete exhaustion occurs after 60-90 minutes. This energy shortfall disrupts ion homeostasis, as ATP-dependent pumps like the Na+/K+-ATPase fail, causing sodium accumulation inside the cell and cellular swelling. Concurrently, calcium influx occurs through voltage-gated channels and the reversal of the Na+/Ca2+ exchanger, elevating cytosolic Ca2+ levels and activating damaging enzymes. Hypoxia-induced anaerobic metabolism further contributes to pH shifts via , where accumulation lowers intracellular pH, exacerbating enzyme dysfunction and membrane instability. Protein synthesis is also profoundly affected, with ATP depletion leading to ribosomal detachment from the and polysome disaggregation, thereby reducing rates. This inhibition occurs early in ischemic conditions, conserving energy but halting the production of essential proteins needed for repair and maintenance. can briefly compound these effects by further impairing ribosomal function, though metabolic shifts predominate.

Oxidative and Free Radical Damage

Oxidative damage to cells arises primarily from (ROS), which are chemically reactive molecules containing oxygen, such as anion (O₂⁻), (H₂O₂), and (•OH). These species can oxidize cellular components, leading to structural and functional impairments. Free radicals, a subset of ROS with unpaired electrons, exacerbate this damage by propagating chain reactions that amplify . Endogenous sources of ROS include mitochondrial leaks during aerobic respiration, where approximately 1-2% of oxygen is converted to ; activity during ; and in immune cells, which generates for killing but contributes to in non-infectious contexts. Lipid peroxidation represents a major type of ROS-induced damage, initiating chain reactions in polyunsaturated fatty acids of cell membranes. This process begins with hydrogen abstraction by a free radical, forming a peroxyl radical that propagates further oxidation, resulting in membrane rigidity, increased permeability, and loss of integrity. , another key damage mechanism, involves the irreversible addition of carbonyl groups to side chains, often via reactions with byproducts like or . This modification inactivates enzymes and disrupts signaling pathways, contributing to cellular dysfunction. For instance, carbonylation of key metabolic enzymes can impair overall cellular . Cells counter ROS through enzymatic antioxidants, including (SOD), which catalyzes the dismutation of to and oxygen, preventing •OH formation via Fenton reactions; , which decomposes into water and oxygen in peroxisomes; and (GPx), which reduces and organic hydroperoxides using as a cofactor, thereby protecting and proteins. These enzymes form a coordinated defense system, with deficiencies linked to heightened oxidative vulnerability. In following ischemia, the restoration of oxygen supply triggers a burst of ROS from activated and damaged mitochondria, amplifying tissue damage beyond the initial hypoxic insult and leading to in affected organs like the heart. Similarly, ultraviolet (UV) radiation generates hydroxyl radicals through photosensitization of cellular chromophores and subsequent Fenton-like reactions with iron, causing acute in cells and contributing to and .

DNA-Specific Damage

Types of DNA Lesions

DNA lesions encompass a variety of structural alterations to the DNA molecule, arising from both endogenous factors like metabolic byproducts and exogenous agents such as environmental toxins and . These lesions disrupt the integrity of the genetic material, potentially leading to if not addressed. The primary categories include base modifications, strand breaks, and crosslinks, each with distinct chemical natures and causative mechanisms. Base modifications involve chemical changes to the bases without breaking the phosphodiester backbone. , a common modification, occurs when alkyl groups are covalently attached to bases, often at or oxygen atoms; for instance, nitrosamines from or can methylate to form O6-methylguanine or N7-methylguanine, while endogenous S-adenosylmethionine can cause similar low-level . Oxidation, driven by (ROS) generated during or from exogenous , produces lesions like , which alters base pairing fidelity. Another example is spontaneous , an endogenous process where loses its amino group to become uracil, accelerated by heat or hydrolytic conditions. Strand breaks represent disruptions in the DNA sugar- backbone. Single-strand breaks (SSBs) occur when one strand is cleaved, often leaving a 3'- or other damaged ; these can result from endogenous ROS or exogenous like X-rays, which generate free radicals that abstract hydrogen atoms from the sugar. Double-strand breaks (DSBs), more severe as they sever both strands, are typically induced by exogenous agents such as drugs (e.g., or ) that create radical intermediates or directly cleave the backbone, though high-dose can also produce them through clustered SSBs. Crosslinks form covalent bonds that tether DNA components, impeding strand separation. Interstrand crosslinks (ICLs) link bases on opposite strands, commonly caused by exogenous bifunctional alkylating agents like cisplatin (which forms about 5-10% ICLs alongside more intrastrand lesions) or psoralens activated by UVA light in phototherapy. Intrastrand crosslinks, such as UV-induced cyclobutane pyrimidine dimers (CPDs) between adjacent thymines or cytosines on the same strand, arise from direct absorption of UVB radiation, distorting the helix. Protein-DNA adducts, another form of crosslink, involve covalent attachments between DNA and proteins; for example, benzopyrene from cigarette smoke forms bulky adducts with guanine, while endogenous formaldehyde can crosslink histones to DNA.

DNA Repair Processes

DNA repair processes are essential cellular mechanisms that detect and correct DNA lesions resulting from endogenous sources like or exogenous agents such as UV radiation and , thereby maintaining genomic integrity and preventing cell damage progression to , , or . These pathways respond to approximately 70,000 DNA damage events per day in a typical , primarily single-strand breaks, with double-strand breaks being the most cytotoxic if unrepaired. Deficiencies in these processes are linked to accelerated aging, neurodegeneration, and cancer predisposition, as unrepaired damage leads to genomic instability. The primary DNA repair pathways include direct reversal, (BER), (NER), mismatch repair (MMR), and double-strand break repair (DSBR) via (HR) or (NHEJ). Direct reversal involves enzymes that restore damaged bases without excision, such as photolyases that split UV-induced using light energy or O6-methylguanine-DNA methyltransferase () that transfers alkyl groups from O6-alkylguanine to itself, repairing damage in a single step. This pathway is highly efficient for specific lesions but limited in scope, failing to address more complex damage and leaving cells vulnerable to if overwhelmed. BER targets non-bulky base modifications, such as oxidative lesions like or deaminated bases, initiated by (e.g., OGG1 for ) that remove the damaged base, creating an abasic site processed by AP endonuclease 1 (APE1) to generate a single-strand break. DNA polymerase β then fills the gap with 1-10 nucleotides, and DNA ligase III seals the nick, often scaffolded by XRCC1 and activated by poly(ADP-ribose) polymerase 1 (). This short-patch repair predominates for endogenous , while long-patch BER incorporates more nucleotides using polymerase δ/ε; unrepaired BER substrates contribute to mutations in cancer and neurodegenerative diseases like Alzheimer's. NER addresses bulky helix-distorting lesions, such as UV-induced cyclobutane pyrimidine dimers or chemical adducts from carcinogens, through two subpathways: global genome NER (GG-NER), which scans the entire genome via XPC-RAD23B recognition, and transcription-coupled NER (TC-NER), which prioritizes actively transcribed genes using CSA/CSB proteins. The TFIIH complex (including XPB and XPD) unwinds DNA, followed by incision 24-32 nucleotides away from the lesion by XPG and ERCC1-XPF endonucleases, excision of the oligonucleotide, and resynthesis using the undamaged strand as template by polymerases δ/ε and ligation by ligase I. Defects in NER, as seen in xeroderma pigmentosum, increase skin cancer risk over 1,000-fold due to persistent UV damage. MMR corrects replication errors and small insertion/deletion loops, recognizing mismatches like G-T or A-C via MutSα (MSH2-MSH6) or MutSβ (MSH2-MSH3) complexes, which recruit MutLα (MLH1-PMS2) for strand discrimination and to the erroneous segment. PCNA directs the process on the newly synthesized strand, followed by resynthesis and ; this pathway reduces mutation rates by 100- to 1,000-fold, and its impairment, as in Lynch syndrome, leads to and . DSBR handles the most severe lesions—double-strand breaks from ionizing radiation or replication fork collapse—primarily through HR in S/G2 phases for error-free repair or NHEJ throughout the cell cycle for rapid but potentially mutagenic ligation. HR begins with MRN complex (MRE11-RAD50-NBS1) resection, loading of RPA and RAD51 nucleoprotein filament mediated by BRCA2, followed by strand invasion of a homologous template (e.g., sister chromatid) and resolution, ensuring high fidelity. Key players include BRCA1 for end resection and PALB2 for RAD51 stabilization; HR defects, such as BRCA1/2 mutations, cause Fanconi anemia and breast/ovarian cancer predisposition. NHEJ, conversely, uses Ku70/Ku80 to bind breaks, recruiting DNA-PKcs for autophosphorylation and Artemis for end processing, culminating in ligation by XRCC4-LIG4-XLF, often introducing small insertions/deletions. NHEJ deficiencies result in severe combined immunodeficiency and radiosensitivity, highlighting its role in immune development via V(D)J recombination. An alternative microhomology-mediated end joining (alt-MHEJ or alt-NHEJ) pathway, involving PARP1 and LIG3, serves as a backup but promotes chromosomal translocations. These interconnected pathways are regulated by modifications and , with sensors like /ATR kinases coordinating repair to minimize cell damage; their collective efficiency underscores the 2015 Nobel Prize in Chemistry awarded to , Paul Modrich, and for elucidating BER, MMR, and NER mechanisms.

References

  1. [1]
    Mechanisms and Morphology of Cellular Injury, Adaptation, and Death
    These fundamental underlying biochemical mechanisms of cell injury are (1) ATP depletion, (2) permeabilization of cell membranes, (3) disruption of biochemical ...
  2. [2]
    [PDF] Cell Injury, Adaptation and Death
    If the injury is too severe (“irreversible injury”), the affected cells die. Page 3. Causes of Cell Injury. • Hypoxia and ischemia. • “Chemical” agents.Missing: definition | Show results with:definition
  3. [3]
  4. [4]
    UV Radiation in DNA Damage and Repair Involving DNA ...
    Prolonged exposure to ultraviolet radiation on human skin can lead to mutations in DNA, photoaging, suppression of the immune system, and other damage up to ...
  5. [5]
    Ionizing radiation-induced DNA injury and damage detection in ...
    Ionizing radiation directly affects DNA structure by inducing DNA breaks, particularly, DSBs. Secondary effects are the generation of reactive oxygen species ( ...Ionizing Radiation Effects... · Dsb Repair Pathways · Radiosensitivity In Breast...<|control11|><|separator|>
  6. [6]
    Cyanide Toxicity - StatPearls - NCBI Bookshelf - NIH
    Feb 22, 2025 · Cyanide toxicity is a rare but often fatal poisoning that occurs through ingestion, inhalation, dermal absorption, or injection.
  7. [7]
    Mechanisms of acetaminophen hepatotoxicity and their translation ...
    This review focuses on the mechanisms behind APAP-induced hepatotoxicity and the relevance of these to the human pathophysiology.
  8. [8]
    Heavy Metals Toxicity: Mechanism, Health Effects, and Therapeutic ...
    Aug 19, 2025 · Heavy metals interfere with intracellular processes, disrupt cell membranes, cause DNA damage, and affect the nervous, kidney, cardiovascular, ...
  9. [9]
    Alcohol-induced generation of lipid peroxidation products in humans
    The development of hepatic disease in response to alcohol feeding in the rat is associated with an increase in lipid peroxides and in conjugated dienes in ...
  10. [10]
    Bacterial Pathogenesis - Medical Microbiology - NCBI Bookshelf - NIH
    Endotoxins: The lipopolysaccharide endotoxins on Gram-negative bacteria cause fever, changes in blood pressure, inflammation, lethal shock, and many other ...
  11. [11]
    HIV-1 Hijacking of Host ATPases and GTPases That Control Protein ...
    HIV-1 hijacks many host proteins to ensure an efficient replication cycle and to evade the immune response, leading to pathogenesis.
  12. [12]
    What's the Damage? The Impact of Pathogens on Pathways ... - NIH
    Pathogens impact host cell genomes directly through genotoxins and oncoproteins that induce changes to cellular DNA and by impairing DNA repair mechanisms (see ...
  13. [13]
    Mechanisms of Autoantibody-Induced Pathology - PMC
    According to our algorithm, autoantibodies can be classified into the following categories: (1) mimic receptor stimulation, (2) blocking of neural transmission, ...
  14. [14]
    The 'Cytokine Storm': molecular mechanisms and therapeutic ...
    Cytokine storm syndrome has generally been described as a collection of clinical manifestations resulting from an overactivated immune system.
  15. [15]
    Low micronutrient intake may accelerate the degenerative diseases ...
    Deficiencies in many micronutrients cause DNA damage, such as chromosome breaks, in cultured human cells or in vivo.
  16. [16]
    Vitamin C Deficiency - StatPearls - NCBI Bookshelf
    Nov 12, 2023 · Vitamin C deficiency, commonly called scurvy, is a well-documented nutritional disorder with historical significance that continues to impact global health ...<|separator|>
  17. [17]
    Hypoxia-Ischemia and Brain infarction - Basic Neurochemistry - NCBI
    Hypoglycemia produces brain injury that has several features in common with ischemic injury. Neurons are more susceptible than glial cells to ischemia, hypoxia ...
  18. [18]
    Insight into the Effects of High-Altitude Hypoxic Exposure on ...
    Under hypoxic stress, glutamate release increases, leading to overactivity of NMDARs and loss of control of Ca2+ influx, ultimately causing neuronal injury and ...
  19. [19]
    Hypoxia. 2. Hypoxia regulates cellular metabolism - PubMed Central
    Hypoxia diminishes ATP utilization by downregulating protein translation and the activity of the Na-K-ATPase. Hypoxia diminishes ATP production in part by ...
  20. [20]
    Cell Biology of Ischemia/Reperfusion Injury - PMC - PubMed Central
    Ischemia also depletes cellular ATP which inactivates ATPases (e.g., Na+/K+ ATPase), reduces active Ca2+ efflux, and limits the reuptake of calcium by the ...
  21. [21]
    Mechanisms of Cell and Tissue Damage - PMC - PubMed Central
    The damage inflicted by pathogens on their hosts is the result of direct and indirect collateral effects resulting from the activity of virulence factors ...
  22. [22]
    Membrane Repair: Mechanisms and Pathophysiology - PMC
    Cell swelling, acidosis, and oxidation compromises the plasma membrane. Membranes become leaky, with breaches sufficiently large to allow the release of ...
  23. [23]
    Blebbing, free Ca2+ and mitochondrial membrane ... - PubMed - NIH
    Cell surface 'blebbing' is an early consequence of hypoxic and toxic injury to cells. A rise in cytosolic free Ca2+ has been suggested as the stimulus for ...
  24. [24]
    Unravelling oncosis: morphological and molecular insights into a ...
    Oncosis, characterized by significant energy consumption, cell swelling, dilation of the endoplasmic reticulum, mitochondrial swelling, and nuclear chromatin ...5.4. Phospholipase A2 · 9.2. Cell Membrane Damage... · 9.3. Mitochondrial...<|control11|><|separator|>
  25. [25]
    Paraptosis—A Distinct Pathway to Cell Death - PMC - PubMed Central
    Paraptosis is a form of regulated cell death characterized by ER dilatation, mitochondrial swelling, and cytoplasmic vacuolation. Physiological paraptosis ...
  26. [26]
    Diversity and complexity of cell death: a historical review - PMC
    Lysosomal cell death results from lysosomal membrane permeabilization, which causes the release of lysosomal enzymes into the cytoplasm and activation of cell ...
  27. [27]
    Links of Cytoskeletal Integrity with Disease and Aging - PMC - NIH
    Sep 16, 2022 · When certain amino acid residues in microtubules and actin microfilaments are oxidized, the ability of microtubules to polymerize is reduced and ...
  28. [28]
    Cytoskeletal Disruption after Electroporation and Its Significance to ...
    (a) Actin disruption is the cytoskeletal component most frequently investigated by studies. Many studies also consider microtubules (MT) disruption. Few studies ...
  29. [29]
    Radiation-Induced Lipid Peroxidation Triggers Ferroptosis and ... - NIH
    We report here that ferroptosis, a form of non-apoptotic cell death driven by lipid peroxidation, is partly responsible for radiation-induced cancer cell death.
  30. [30]
    Role of Pore-Forming Toxins in Bacterial Infectious Diseases - PMC
    PFTs function to perforate membranes of host cells, predominantly the plasma membrane but also intracellular organelle membranes (6).
  31. [31]
    Histology, Cell Death - StatPearls - NCBI Bookshelf
    Jan 30, 2023 · Necrosis typically occurs when there is extensive damage to the cell membrane and internal structures that push the cell past reversible injury.Missing: Na+ imbalance
  32. [32]
    Alcohol-Associated Liver Disease - StatPearls - NCBI Bookshelf
    Jul 13, 2023 · Fatty liver is generally considered a reversible condition. On further progression, there is marked steatosis, hepatocellular necrosis, and ...
  33. [33]
    Fatty Liver: Overview, Etiology, Pathophysiology
    Aug 28, 2025 · Abstinence from alcohol may reverse steatosis in patients with alcohol-related fatty liver. The steatosis usually resolves within 2 weeks of ...
  34. [34]
    Necrosis Pathology - StatPearls - NCBI Bookshelf - NIH
    Morphology · 1) Coagulative necrosis: Ischemia in most organs except the brain can lead to coagulative necrosis. · 2) Liquefactive necrosis: This morphology is ...Anatomical Pathology · Clinical Pathology · Morphology · Mechanisms
  35. [35]
    Apoptosis, Pyroptosis, and Necrosis: Mechanistic Description of ...
    Necrosis is a term used by pathologists to designate the presence of dead tissues or cells and is the sum of changes that have occurred in cells after they ...
  36. [36]
    Acute Myocardial Infarction - StatPearls - NCBI Bookshelf - NIH
    Sep 3, 2023 · At 4 to 12 hours, the myocardium undergoes coagulation necrosis and edema. At 12 to 24 hours, the gross specimen appears dark and mottled ...<|separator|>
  37. [37]
    Cell Liquefactive Necrosis - StatPearls - NCBI Bookshelf
    Jan 20, 2025 · Fat Necrosis. Gangrenous Necrosis. Fibrinoid Necrosis. The other types of necrosis listed above do not represent distinct pathological entities.
  38. [38]
    Molecular mechanisms of cell death: recommendations of ... - Nature
    Jan 23, 2018 · Limited mitochondrial permeabilization causes DNA damage and genomic instability in the absence of cell death. Mol Cell. 2015;57:860–72. Article ...
  39. [39]
    Molecular mechanisms of cell death in neurological diseases - Nature
    Jun 7, 2021 · Apoptosis can be triggered by two distinct pathways: the intrinsic (also called mitochondrial or BCL-2-regulated) pathway and the death receptor ...
  40. [40]
    Emerging roles of caspase-3 in apoptosis | Cell Death & Differentiation
    Jan 28, 1999 · Caspase-3 is a death protease essential for apoptosis, brain development, and chromatin condensation, and is required for cell dismantling.<|control11|><|separator|>
  41. [41]
    Autophagy: Regulator of cell death | Cell Death & Disease - Nature
    Oct 4, 2023 · The switch between necroptosis and apoptosis is achieved by increasing cellular ATP utilization through “self-eating” (when ATP reserves are ...
  42. [42]
    Ferroptosis: past, present and future | Cell Death & Disease - Nature
    Feb 3, 2020 · Ferroptosis is a new type of programmed cell death, which occurs with iron dependence. Ferroptosis plays an important regulatory role in the ...<|control11|><|separator|>
  43. [43]
    50 years on and still very much alive: 'Apoptosis: a basic biological ...
    Nov 11, 2022 · It is now well established in the cell death field, that apoptosis can be triggered via two different pathways, depending on the origin of the ...
  44. [44]
    The role of apoptosis in the development and function of T ... - Nature
    Oct 1, 2005 · Apoptosis plays an essential role in T cell biology. Thymocytes expressing nonfunctional or autoreactive TCRs are eliminated by apoptosis during development.
  45. [45]
    The multifaceted role of ferroptosis in liver disease - Nature
    Jan 24, 2022 · Under iron-overload conditions, excess iron results in redox-active non-transferrin-bound iron (NTBI), the uptake of which is mediated by metal ...
  46. [46]
    Tissue Homeostasis, Inflammation, and Repair - NCBI
    Labile tissues have stem cells like epithelial cells and hematopoietic cells, and differentiated cells in the tissue are continuously renewed by stem cells.
  47. [47]
    Epidermal Stem Cells in Skin Wound Healing - PMC - PubMed Central
    Sep 1, 2017 · In this review, we will dissect the biological process of adult skin wound healing and emphasize the importance of epidermal stem cells during the wound ...
  48. [48]
    Liver Regeneration after Partial Hepatectomy - NIH
    Hepatocyte proliferation after partial hepatectomy (PHx) depends on a concerted action between MET (the HGF receptor), EGFR (EGF receptor), and several ...
  49. [49]
    Tissue repair: The hidden drama - PMC - PubMed Central
    Continuously dividing tissues (also known as labile tissues) are comprised of cells that are constantly proliferating in order to replace dead or sloughed-off ...
  50. [50]
    Reactive gliosis and the multicellular response to CNS damage and ...
    This article examines and reviews the multiple cell types involved in, and contributing to, different types of CNS insults.
  51. [51]
    Mechanisms of Cardiac Regeneration - PMC - PubMed Central
    In response to cardiac injury, adult mammals—including humans—fail to regenerate the majority of the lost cardiomyocytes and instead replace necrotic muscle ...
  52. [52]
    Wound repair and regeneration: Mechanisms, signaling, and ...
    Furthermore, keratinocyte growth factor-1 (KGF-1; also known as FGF-7), which targets epidermal cells, has been reported to promote healing of skin wounds in ...
  53. [53]
    Scar Formation: Cellular Mechanisms - NCBI - NIH
    Dec 8, 2020 · Fibroblasts are key players in the maintenance of skin homeostasis and in orchestrating physiological tissue repair.
  54. [54]
    Wound Healing Phases - StatPearls - NCBI Bookshelf
    The proliferative phase is characterized by the formation of granulation tissue, reepithelialization, and neovascularization. This phase can last several ...
  55. [55]
    Wound healing: cellular mechanisms and pathological outcomes - NIH
    Sep 30, 2020 · Fibroblasts degrade the provisional matrix by producing MMPs and replace it with a granulation tissue rich in fibronectin, immature collagens ...Wound Healing: Cellular... · 2.1. Haemostasis · 2.2. Inflammation<|control11|><|separator|>
  56. [56]
    Extracellular Targets to Reduce Excessive Scarring in Response to ...
    Apr 27, 2023 · Excessive scarring of the skin, tendon, muscle, and ligament alters the mechanical functions of these tissues. Similarly, ocular scars may ...<|control11|><|separator|>
  57. [57]
    Hepatic Cirrhosis - StatPearls - NCBI Bookshelf - NIH
    After a chronic injury, most of the liver tissue becomes fibrotic, leading to loss of function and the development of cirrhosis.
  58. [58]
    Cardiac fibrosis in myocardial infarction—from repair and ...
    Ischemic cell death during a myocardial infarction leads to a multiphase reparative response in which the damaged tissue is replaced with a fibrotic scar ...
  59. [59]
    Hypertrophic Scarring and Keloids: Epidemiology, Molecular ...
    Oct 4, 2025 · Although traditionally considered a low‐tension site, the earlobe is particularly prone to keloid formation, primarily due to piercing‐induced ...
  60. [60]
    Intracellular energy production and distribution in hypoxia
    Jul 26, 2023 · However, as cells undergo hypoxia, ATP production is reduced due to reduced mitochondrial metabolism. Hypoxia then presents a “non-ideal state” ...
  61. [61]
    Glyoxylate protects against cyanide toxicity through metabolic ...
    Mar 23, 2022 · By inhibiting Complex IV, cyanide blocks electron flux from cytochrome c to oxygen coupled with pumping of protons across the mitochondrial ...
  62. [62]
    Prolonged depletion of ATP and of the adenine nucleotide pool due ...
    The mean ATP after 15 min of ischemia was reduced 62% from 5.42 ± 0.33 to 2.08 ± 0.21 μmol/g; and the total nucleotide content was reduced by 50%.<|control11|><|separator|>
  63. [63]
    THE CELL BIOLOGY OF ACUTE MYOCARDIAL ISCHEMIA!
    Noninhibited ischemic tissue does not suffer complete depletion of A TP nor develop contracture-rigor until 60 to 90 minutes of ischemia have passed. Tissue ADP ...
  64. [64]
    Mechanisms of cell injury and death
    Cell injury occurs as a result of physical, chemical or biological insults or as a result of vital substrate deficiency (table 1). The cellular response to ...
  65. [65]
    Lactic Acidosis: It's Not Just Hypoxia Anymore - CHEST Journal
    Oct 23, 2012 · Lactic acidosis is the most common cause of metabolic acidosis. Although generation of lactate is generally associated with hypoxia causing the body to shift ...
  66. [66]
    Current Mechanistic Concepts in Ischemia and Reperfusion Injury
    Apr 20, 2018 · The detachment of ribosomes decreases protein synthesis. After the reperfusion stage, restoring blood flow to ischemic tissue provides ...
  67. [67]
    Mitochondrial ROS in myocardial ischemia reperfusion and ...
    ... detachment of ribosomes with subsequently impaired protein synthesis [4,5]. Inhibition of the electron transport chain (ETC) during IR contributes to ATP ...
  68. [68]
    ROS: Basic Concepts, Sources, Cellular Signaling, and its ...
    Reactive oxygen species (ROS) are bioproducts of cellular metabolism. There is a range of molecules with oxidizing properties known as ROS.
  69. [69]
    Reactive Oxygen Species: the Dual Role in Physiological and ...
    Other cellular sources of ROS include neutrophils, monocytes, cardiomyocytes, endothelial cells, xanthine oxidases, cytochrome P450, lipoxygenases, and ...
  70. [70]
    Defining roles of specific reactive oxygen species (ROS) in cell ...
    Feb 21, 2022 · Comprehensive review of methodology to detect reactive oxygen species (ROS) in mammalian species and establish its relationship with ...
  71. [71]
    Exploring the Biology of Lipid Peroxidation Derived Protein ...
    Protein modification through carbonylation reactions is a documented consequence of oxidative stress, first defined as “a disturbance in the prooxidant ...
  72. [72]
    Protein carbonylation as a major hallmark of oxidative damage
    Protein carbonylation is a harmful, irreversible oxidative protein modification and a major hallmark of oxidative stress-related disorders.
  73. [73]
    Protein Carbonylation and Metabolic Control Systems - PMC
    Protein carbonylation is the non-enzymatic modification of proteins by reactive lipid aldehydes, linked to oxidative stress and metabolic states.
  74. [74]
    First line defence antioxidants-superoxide dismutase (SOD ...
    First line defense antioxidants are superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX), which suppress free radical formation.
  75. [75]
    The importance of antioxidants which play the role in cellular ...
    The most efficient enzymatic antioxidants contain glutathione peroxidase, catalase and superoxide dismutase. Non-enzymatic antioxidants include Vitamin E ...
  76. [76]
    Reperfusion injury and reactive oxygen species - PubMed Central
    This review provides a synopsis of the evidence implicating ROS in reperfusion injury, the clinical implications of this phenomenon, and summarizes current ...
  77. [77]
    Role of reactive oxygen species in ultraviolet-induced photodamage ...
    Jan 12, 2024 · Long-term exposure to UV can cause DNA damage, oxidative stress, inflammation, and cell apoptosis [22]. UV can interact with chromogenic groups ...
  78. [78]
    DNA damage repair: historical perspectives, mechanistic pathways ...
    Jul 9, 2021 · Several types of DNA damage have been reported previously, as follows: (i) single-strand breaks; (ii) double-strand breaks (DSBs); (iii) base ...
  79. [79]
    Mechanisms of DNA damage, repair and mutagenesis - PMC
    DNA damage can be categorized into two main classes based on its origin: endogenous and exogenous. The majority of the endogenous DNA damage arises from the ...
  80. [80]
    DNA damage and mutation. Types of DNA damage - BioDiscovery
    Feb 23, 2014 · This review outlines the basic types of DNA damage caused by exogenous and endogenous factors, analyses the possible consequences of each type of damage
  81. [81]
    Mechanisms of interstrand DNA crosslink repair and human disorders
    May 1, 2016 · Interstrand DNA crosslinks (ICLs) are lesions that are a covalent linkage between opposite strands of double-stranded DNA. They are formed in ...
  82. [82]
    Exploring DNA Damage and Repair Mechanisms - NIH
    Jan 16, 2024 · This comprehensive review paper consolidates research efforts, focusing on DNA repair mechanisms, computational research methods, and associated databases.
  83. [83]
    DNA-damage repair; the good, the bad, and the ugly - PMC
    The NER pathway. The NER pathway is a multistep process that serves to repair a variety of DNA damage, including DNA lesions caused by UV radiation, mutagenic ...