Wound healing

Last updated
Hand abrasion
Hand Abrasion - 32 minutes after injury.JPG Hand Abrasion - 2 days 22 hours 12 minutes after injury.JPG Hand Abrasion - 17 days 11 hours 30 minutes after injury.JPG Hand Abrasion - 30 days 4 hours 43 minutes after injury.JPG
Initial injury3 days17 days30 days

Wound healing refers to a living organism's replacement of destroyed or damaged tissue by newly produced tissue. [1]

Contents

In undamaged skin, the epidermis (surface, epithelial layer) and dermis (deeper, connective layer) form a protective barrier against the external environment. When the barrier is broken, a regulated sequence of biochemical events is set into motion to repair the damage. [1] [2] This process is divided into predictable phases: blood clotting (hemostasis), inflammation, tissue growth (cell proliferation), and tissue remodeling (maturation and cell differentiation). Blood clotting may be considered to be part of the inflammation stage instead of a separate stage. [3]

Deep wound on shin with stitches healing over five weeks Wound Healing.jpg
Deep wound on shin with stitches healing over five weeks

The wound-healing process is not only complex but fragile, and it is susceptible to interruption or failure leading to the formation of non-healing chronic wounds. Factors that contribute to non-healing chronic wounds are diabetes, venous or arterial disease, infection, and metabolic deficiencies of old age. [4]

Wound care encourages and speeds wound healing via cleaning and protection from reinjury or infection. Depending on each patient's needs, it can range from the simplest first aid to entire nursing specialties such as wound, ostomy, and continence nursing and burn center care.

Stages

Approximate times of the different phases of wound healing on a logarithmic scale, with faded intervals marking substantial variation, depending mainly on wound size and healing conditions, but image does not include major impairments that cause chronic wounds. Wound healing phases.png
Approximate times of the different phases of wound healing on a logarithmic scale, with faded intervals marking substantial variation, depending mainly on wound size and healing conditions, but image does not include major impairments that cause chronic wounds.

Timing and re-epithelialization

Timing is important to wound healing. Critically, the timing of wound re-epithelialization can decide the outcome of the healing. [11] If the epithelization of tissue over a denuded area is slow, a scar will form over many weeks, or months; [12] [13] If the epithelization of a wounded area is fast, the healing will result in regeneration. [13]

Early vs cellular phase

A fluorescence micrograph of cells in Drosophila larvae healing after a puncture wound. The arrow points to cells that have fused to form syncytia, and the arrowheads point to cells that are oriented to face the wound. Fluorescent wound healing.jpg
A fluorescence micrograph of cells in Drosophila larvae healing after a puncture wound. The arrow points to cells that have fused to form syncytia, and the arrowheads point to cells that are oriented to face the wound.

Wound healing is classically divided into hemostasis, inflammation, proliferation, and remodeling. Although a useful construct, this model employs considerable overlapping among individual phases. A complementary model has recently been described [1] where the many elements of wound healing are more clearly delineated. The importance of this new model becomes more apparent through its utility in the fields of regenerative medicine and tissue engineering (see Research and development section below). In this construct, the process of wound healing is divided into two major phases: the early phase and the cellular phase: [1]

The early phase, which begins immediately following skin injury, involves cascading molecular and cellular events leading to hemostasis and formation of an early, makeshift extracellular matrix that provides structural staging for cellular attachment and subsequent cellular proliferation.

The cellular phase involves several types of cells working together to mount an inflammatory response, synthesize granulation tissue, and restore the epithelial layer. [1] Subdivisions of the cellular phase are:

  1. Macrophages and inflammatory components (within 1–2 days)
  2. Epithelial-mesenchymal interaction: re-epithelialization (phenotype change within hours, migration begins on day 1 or 2)
  3. Fibroblasts and myofibroblasts: progressive alignment, collagen production, and matrix contraction (between day 4 and day 14)
  4. Endothelial cells and angiogenesis (begins on day 4)
  5. Dermal matrix: elements of fabrication (begins on day 4, lasting 2 weeks) and alteration/remodeling (begins after week 2, lasting weeks to months—depending on wound size). [1]

Inflammatory phase

Just before the inflammatory phase is initiated, the clotting cascade occurs in order to achieve hemostasis, or the stopping of blood loss by way of a fibrin clot. Thereafter, various soluble factors (including chemokines and cytokines) are released to attract cells that phagocytise debris, bacteria, and damaged tissue, in addition to releasing signaling molecules that initiate the proliferative phase of wound healing.

Clotting cascade

When tissue is first wounded, blood comes in contact with collagen, triggering blood platelets to begin secreting inflammatory factors. [15] Platelets also express sticky glycoproteins on their cell membranes that allow them to aggregate, forming a mass. [7]

Fibrin and fibronectin cross-link together and form a plug that traps proteins and particles and prevents further blood loss. [16] This fibrin-fibronectin plug is also the main structural support for the wound until collagen is deposited. [7] Migratory cells use this plug as a matrix to crawl across, and platelets adhere to it and secrete factors. [7] The clot is eventually lysed and replaced with granulation tissue and then later with collagen.

Platelets, the cells present in the highest numbers shortly after a wound occurs, release mediators into the blood, including cytokines and growth factors. [15] Growth factors stimulate cells to speed their rate of division. Platelets release other proinflammatory factors like serotonin, bradykinin, prostaglandins, prostacyclins, thromboxane, and histamine, [3] which serve several purposes, including increasing cell proliferation and migration to the area and causing blood vessels to become dilated and porous. In many ways, extravasated platelets in trauma perform a similar function to tissue macrophages and mast cells exposed to microbial molecular signatures in infection: they become activated, and secrete molecular mediators – vasoactive amines, eicosanoids, and cytokines – that initiate the inflammatory process.

Vasoconstriction and vasodilation

Immediately after a blood vessel is breached, ruptured cell membranes release inflammatory factors like thromboxanes and prostaglandins that cause the vessel to spasm to prevent blood loss and to collect inflammatory cells and factors in the area. [3] This vasoconstriction lasts five to ten minutes and is followed by vasodilation, a widening of blood vessels, which peaks at about 20 minutes post-wounding. [3] Vasodilation is the result of factors released by platelets and other cells. The main factor involved in causing vasodilation is histamine. [3] [15] Histamine also causes blood vessels to become porous, allowing the tissue to become edematous because proteins from the bloodstream leak into the extravascular space, which increases its osmolar load and draws water into the area. [3] Increased porosity of blood vessels also facilitates the entry of inflammatory cells like leukocytes into the wound site from the bloodstream. [17] [18]

Polymorphonuclear neutrophils

Within an hour of wounding, polymorphonuclear neutrophils (PMNs) arrive at the wound site and become the predominant cells in the wound for the first two days after the injury occurs, with especially high numbers on the second day. [19] They are attracted to the site by fibronectin, growth factors, and substances such as kinins. Neutrophils phagocytise debris and kill bacteria by releasing free radicals in what is called a respiratory burst. [20] [21] They also cleanse the wound by secreting proteases that break down damaged tissue. Functional neutrophils at the wound site only have life-spans of around two days, so they usually undergo apoptosis once they have completed their tasks and are engulfed and degraded by macrophages. [22]

Other leukocytes to enter the area include helper T cells, which secrete cytokines to cause more T cells to divide and to increase inflammation and enhance vasodilation and vessel permeability. [17] [23] T cells also increase the activity of macrophages. [17]

Macrophages

One of the roles of macrophages is to phagocytize other expended phagocytes, [24] bacteria and damaged tissue, [19] and they also debride damaged tissue by releasing proteases. [25]

Macrophages function in regeneration [26] [27] and are essential for wound healing. [19] They are stimulated by the low oxygen content of their surroundings to produce factors that induce and speed angiogenesis [20] and they also stimulate cells that reepithelialize the wound, create granulation tissue, and lay down a new extracellular matrix. [28] By secreting these factors, macrophages contribute to pushing the wound healing process into the next phase. They replace PMNs as the predominant cells in the wound by two days after injury. [24]

The spleen contains half the body's monocytes in reserve ready to be deployed to injured tissue. [29] [30] Attracted to the wound site by growth factors released by platelets and other cells, monocytes from the bloodstream enter the area through blood vessel walls. [31] Numbers of monocytes in the wound peak one to one and a half days after the injury occurs. [23] Once they are in the wound site, monocytes mature into macrophages. Macrophages also secrete a number of factors such as growth factors and other cytokines, especially during the third and fourth post-wounding days. These factors attract cells involved in the proliferation stage of healing to the area. [15]

In wound healing that result in incomplete repair, scar contraction occurs, bringing varying gradations of structural imperfections, deformities and problems with flexibility. [32] Macrophages may restrain the contraction phase. [27] Scientists have reported that removing the macrophages from a salamander resulted in failure of a typical regeneration response (limb regeneration), instead bringing on a repair (scarring) response. [33] [34]

Decline of inflammatory phase

As inflammation dies down, fewer inflammatory factors are secreted, existing ones are broken down, and numbers of neutrophils and macrophages are reduced at the wound site. [19] These changes indicate that the inflammatory phase is ending and the proliferative phase is underway. [19] In vitro evidence, obtained using the dermal equivalent model, suggests that the presence of macrophages actually delays wound contraction and thus the disappearance of macrophages from the wound may be essential for subsequent phases to occur. [27]

Because inflammation plays roles in fighting infection, clearing debris and inducing the proliferation phase, it is a necessary part of healing. However, inflammation can lead to tissue damage if it lasts too long. [7] Thus the reduction of inflammation is frequently a goal in therapeutic settings. Inflammation lasts as long as there is debris in the wound. Thus, if the individual's immune system is compromised and is unable to clear the debris from the wound and/or if excessive detritus, devitalized tissue, or microbial biofilm is present in the wound, these factors may cause a prolonged inflammatory phase and prevent the wound from properly commencing the proliferation phase of healing. This can lead to a chronic wound.

Proliferative phase

About two or three days after the wound occurs, fibroblasts begin to enter the wound site, marking the onset of the proliferative phase even before the inflammatory phase has ended. [35] As in the other phases of wound healing, steps in the proliferative phase do not occur in a series but rather partially overlap in time.

Angiogenesis

Also called neovascularization, the process of angiogenesis occurs concurrently with fibroblast proliferation when endothelial cells migrate to the area of the wound. [36] Because the activity of fibroblasts and epithelial cells requires oxygen and nutrients, angiogenesis is imperative for other stages in wound healing, like epidermal and fibroblast migration. The tissue in which angiogenesis has occurred typically looks red (is erythematous) due to the presence of capillaries. [36]

Angiogenesis occurs in overlapping phases in response to inflammation:

  1. Latent period: During the haemostatic and inflammatory phase of the wound healing process, vasodilation and permeabilisation allow leukocyte extravasation and phagocytic debridement and decontamination of the wound area. Tissue swelling aids later angiogenesis by expanding and loosening the existing collagenous extracellular matrix.
  2. Endothelial activation: As the wound macrophages switches from inflammatory to healing mode, it begins to secrete endothelial chemotactic and growth factors to attract adjacent endothelial cells. Activated endothelial cells respond by retracting and reducing cell junctions, loosening themselves from their embedded endothelium. Characteristically the activated endothelial cells show enlarged nucleoli.
  3. Degradation of endothelial basement membrane: The wound macrophages, mast cells and the endothelial cells themselves secrete proteases to break down existing vascular basal lamina.
  4. Vascular sprouting: With the breakdown of endothelial basement membrane, detached endothelial cells from pre-existing capillaries and post-capillary venules can divide and migrate chemotactically towards the wound, laying down new vessels in the process. Vascular sprouting can be aided by ambient hypoxia and acidosis in the wound environment, as hypoxia stimulates the endothelial transcription factor, hypoxia inducible factor (HIF) to transactivate angiogenic genes such as VEGF and GLUT1. Sprouted vessels can self-organise into luminal morphologies, and fusion of blind channels give rise to new capillary networks.
  5. Vascular maturation: the endothelium of vessels mature by laying down new endothelial extracellular matrix, followed by basal lamina formation. Lastly the vessel establishes a pericyte layer.

Stem cells of endothelial cells, originating from parts of uninjured blood vessels, develop pseudopodia and push through the ECM into the wound site to establish new blood vessels. [20]

Endothelial cells are attracted to the wound area by fibronectin found on the fibrin scab and chemotactically by angiogenic factors released by other cells, [37] e.g. from macrophages and platelets when in a low-oxygen environment. Endothelial growth and proliferation is also directly stimulated by hypoxia, and presence of lactic acid in the wound. [35] For example, hypoxia stimulates the endothelial transcription factor, hypoxia-inducible factor (HIF) to transactivate a set of proliferative genes including vascular endothelial growth factor (VEGF) and glucose transporter 1 (GLUT1).

To migrate, endothelial cells need collagenases and plasminogen activator to degrade the clot and part of the ECM. [3] [19] Zinc-dependent metalloproteinases digest basement membrane and ECM to allow cell migration, proliferation and angiogenesis. [38]

When macrophages and other growth factor-producing cells are no longer in a hypoxic, lactic acid-filled environment, they stop producing angiogenic factors. [20] Thus, when tissue is adequately perfused, migration and proliferation of endothelial cells is reduced. Eventually blood vessels that are no longer needed die by apoptosis. [37]

Fibroplasia and granulation tissue formation

Simultaneously with angiogenesis, fibroblasts begin accumulating in the wound site. Fibroblasts begin entering the wound site two to five days after wounding as the inflammatory phase is ending, and their numbers peak at one to two weeks post-wounding. [19] By the end of the first week, fibroblasts are the main cells in the wound. [3] Fibroplasia ends two to four weeks after wounding.

As a model the mechanism of fibroplasia may be conceptualised as an analogous process to angiogenesis (see above) - only the cell type involved is fibroblasts rather than endothelial cells. Initially there is a latent phase where the wound undergoes plasma exudation, inflammatory decontamination and debridement. Oedema increases the wound histologic accessibility for later fibroplastic migration. Second, as inflammation nears completion, macrophage and mast cells release fibroblast growth and chemotactic factors to activate fibroblasts from adjacent tissue. Fibroblasts at this stage loosen themselves from surrounding cells and ECM. Phagocytes further release proteases that break down the ECM of neighbouring tissue, freeing the activated fibroblasts to proliferate and migrate towards the wound. The difference between vascular sprouting and fibroblast proliferation is that the former is enhanced by hypoxia, whilst the latter is inhibited by hypoxia. The deposited fibroblastic connective tissue matures by secreting ECM into the extracellular space, forming granulation tissue (see below). Lastly collagen is deposited into the ECM.

In the first two or three days after injury, fibroblasts mainly migrate and proliferate, while later, they are the main cells that lay down the collagen matrix in the wound site. [3] Origins of these fibroblasts are thought to be from the adjacent uninjured cutaneous tissue (although new evidence suggests that some are derived from blood-borne, circulating adult stem cells/precursors). [39] Initially fibroblasts utilize the fibrin cross-linking fibers (well-formed by the end of the inflammatory phase) to migrate across the wound, subsequently adhering to fibronectin. [37] Fibroblasts then deposit ground substance into the wound bed, and later collagen, which they can adhere to for migration. [15]

Granulation tissue functions as rudimentary tissue, and begins to appear in the wound already during the inflammatory phase, two to five days post wounding, and continues growing until the wound bed is covered. Granulation tissue consists of new blood vessels, fibroblasts, inflammatory cells, endothelial cells, myofibroblasts, and the components of a new, provisional extracellular matrix (ECM). The provisional ECM is different in composition from the ECM in normal tissue and its components originate from fibroblasts. [28] Such components include fibronectin, collagen, glycosaminoglycans, elastin, glycoproteins and proteoglycans. [37] Its main components are fibronectin and hyaluronan, which create a very hydrated matrix and facilitate cell migration. [31] Later this provisional matrix is replaced with an ECM that more closely resembles that found in non-injured tissue.

Growth factors (PDGF, TGF-β) and fibronectin encourage proliferation, migration to the wound bed, and production of ECM molecules by fibroblasts. Fibroblasts also secrete growth factors that attract epithelial cells to the wound site. Hypoxia also contributes to fibroblast proliferation and excretion of growth factors, though too little oxygen will inhibit their growth and deposition of ECM components, and can lead to excessive, fibrotic scarring.

Collagen deposition

One of fibroblasts' most important duties is the production of collagen. [36]

Collagen deposition is important because it increases the strength of the wound; before it is laid down, the only thing holding the wound closed is the fibrin-fibronectin clot, which does not provide much resistance to traumatic injury. [20] Also, cells involved in inflammation, angiogenesis, and connective tissue construction attach to, grow and differentiate on the collagen matrix laid down by fibroblasts. [40]

Type III collagen and fibronectin generally begin to be produced in appreciable amounts at somewhere between approximately 10 hours [41] and 3 days, [37] depending mainly on wound size. Their deposition peaks at one to three weeks. [28] They are the predominating tensile substances until the later phase of maturation, in which they are replaced by the stronger type I collagen.

Even as fibroblasts are producing new collagen, collagenases and other factors degrade it. Shortly after wounding, synthesis exceeds degradation so collagen levels in the wound rise, but later production and degradation become equal so there is no net collagen gain. [20] This homeostasis signals the onset of the later maturation phase. Granulation gradually ceases and fibroblasts decrease in number in the wound once their work is done. [42] At the end of the granulation phase, fibroblasts begin to commit apoptosis, converting granulation tissue from an environment rich in cells to one that consists mainly of collagen. [3]

Epithelialization

The formation of granulation tissue into an open wound allows the reepithelialization phase to take place, as epithelial cells migrate across the new tissue to form a barrier between the wound and the environment. [37] Basal keratinocytes from the wound edges and dermal appendages such as hair follicles, sweat glands and sebaceous (oil) glands are the main cells responsible for the epithelialization phase of wound healing. [42] They advance in a sheet across the wound site and proliferate at its edges, ceasing movement when they meet in the middle. In healing that results in a scar, sweat glands, hair follicles [43] [44] and nerves do not form. With the lack of hair follicles, nerves and sweat glands, the wound, and the resulting healing scar, provide a challenge to the body with regards to temperature control. [44]

Keratinocytes migrate without first proliferating. [45] Migration can begin as early as a few hours after wounding. However, epithelial cells require viable tissue to migrate across, so if the wound is deep it must first be filled with granulation tissue. [46] Thus the time of onset of migration is variable and may occur about one day after wounding. [47] Cells on the wound margins proliferate on the second and third day post-wounding in order to provide more cells for migration. [28]

If the basement membrane is not breached, epithelial cells are replaced within three days by division and upward migration of cells in the stratum basale in the same fashion that occurs in uninjured skin. [37] However, if the basement membrane is ruined at the wound site, reepithelization must occur from the wound margins and from skin appendages such as hair follicles and sweat and oil glands that enter the dermis that are lined with viable keratinocytes. [28] If the wound is very deep, skin appendages may also be ruined and migration can only occur from wound edges. [46]

Migration of keratinocytes over the wound site is stimulated by lack of contact inhibition and by chemicals such as nitric oxide. [48] Before they begin to migrate, cells must dissolve their desmosomes and hemidesmosomes, which normally anchor the cells by intermediate filaments in their cytoskeleton to other cells and to the ECM. [23] Transmembrane receptor proteins called integrins, which are made of glycoproteins and normally anchor the cell to the basement membrane by its cytoskeleton, are released from the cell's intermediate filaments and relocate to actin filaments to serve as attachments to the ECM for pseudopodia during migration. [23] Thus keratinocytes detach from the basement membrane and are able to enter the wound bed. [35]

Before they begin migrating, keratinocytes change shape, becoming longer and flatter and extending cellular processes like lamellipodia and wide processes that look like ruffles. [31] Actin filaments and pseudopodia form. [35] During migration, integrins on the pseudopod attach to the ECM, and the actin filaments in the projection pull the cell along. [23] The interaction with molecules in the ECM through integrins further promotes the formation of actin filaments, lamellipodia, and filopodia. [23]

Epithelial cells climb over one another in order to migrate. [42] This growing sheet of epithelial cells is often called the epithelial tongue. [45] The first cells to attach to the basement membrane form the stratum basale. These basal cells continue to migrate across the wound bed, and epithelial cells above them slide along as well. [45] The more quickly this migration occurs, the less of a scar there will be. [49]

Fibrin, collagen, and fibronectin in the ECM may further signal cells to divide and migrate. Like fibroblasts, migrating keratinocytes use the fibronectin cross-linked with fibrin that was deposited in inflammation as an attachment site to crawl across. [25] [31] [42]

A scab covering a healing wound Scab.jpg
A scab covering a healing wound

As keratinocytes migrate, they move over granulation tissue but stay underneath the scab, thereby separating the scab from the underlying tissue. [42] [47] Epithelial cells have the ability to phagocytize debris such as dead tissue and bacterial matter that would otherwise obstruct their path. Because they must dissolve any scab that forms, keratinocyte migration is best enhanced by a moist environment, since a dry one leads to formation of a bigger, tougher scab. [25] [37] [42] [50] To make their way along the tissue, keratinocytes must dissolve the clot, debris, and parts of the ECM in order to get through. [47] [51] They secrete plasminogen activator, which activates plasminogen, turning it into plasmin to dissolve the scab. Cells can only migrate over living tissue, [42] so they must excrete collagenases and proteases like matrix metalloproteinases (MMPs) to dissolve damaged parts of the ECM in their way, particularly at the front of the migrating sheet. [47] Keratinocytes also dissolve the basement membrane, using instead the new ECM laid down by fibroblasts to crawl across. [23]

As keratinocytes continue migrating, new epithelial cells must be formed at the wound edges to replace them and to provide more cells for the advancing sheet. [25] Proliferation behind migrating keratinocytes normally begins a few days after wounding [46] and occurs at a rate that is 17 times higher in this stage of epithelialization than in normal tissues. [25] Until the entire wound area is resurfaced, the only epithelial cells to proliferate are at the wound edges. [45]

Growth factors, stimulated by integrins and MMPs, cause cells to proliferate at the wound edges. Keratinocytes themselves also produce and secrete factors, including growth factors and basement membrane proteins, which aid both in epithelialization and in other phases of healing. [52] Growth factors are also important for the innate immune defense of skin wounds by stimulation of the production of antimicrobial peptides and neutrophil chemotactic cytokines in keratinocytes.

Keratinocytes continue migrating across the wound bed until cells from either side meet in the middle, at which point contact inhibition causes them to stop migrating. [31] When they have finished migrating, the keratinocytes secrete the proteins that form the new basement membrane. [31] Cells reverse the morphological changes they underwent in order to begin migrating; they reestablish desmosomes and hemidesmosomes and become anchored once again to the basement membrane. [23] Basal cells begin to divide and differentiate in the same manner as they do in normal skin to reestablish the strata found in reepithelialized skin. [31]

Contraction

Contraction is a key phase of wound healing with repair. If contraction continues for too long, it can lead to disfigurement and loss of function. [32] Thus there is a great interest in understanding the biology of wound contraction, which can be modelled in vitro using the collagen gel contraction assay or the dermal equivalent model. [27] [53]

Contraction commences approximately a week after wounding, when fibroblasts have differentiated into myofibroblasts. [54] In full thickness wounds, contraction peaks at 5 to 15 days post wounding. [37] Contraction can last for several weeks [46] and continues even after the wound is completely reepithelialized. [3] A large wound can become 40 to 80% smaller after contraction. [31] [42] Wounds can contract at a speed of up to 0.75 mm per day, depending on how loose the tissue in the wounded area is. [37] Contraction usually does not occur symmetrically; rather most wounds have an 'axis of contraction' which allows for greater organization and alignment of cells with collagen. [54]

At first, contraction occurs without myofibroblast involvement. [55] Later, fibroblasts, stimulated by growth factors, differentiate into myofibroblasts. Myofibroblasts, which are similar to smooth muscle cells, are responsible for contraction. [55] Myofibroblasts contain the same kind of actin as that found in smooth muscle cells. [32]

Myofibroblasts are attracted by fibronectin and growth factors and they move along fibronectin linked to fibrin in the provisional ECM in order to reach the wound edges. [25] They form connections to the ECM at the wound edges, and they attach to each other and to the wound edges by desmosomes. Also, at an adhesion called the fibronexus, actin in the myofibroblast is linked across the cell membrane to molecules in the extracellular matrix like fibronectin and collagen. [55] Myofibroblasts have many such adhesions, which allow them to pull the ECM when they contract, reducing the wound size. [32] In this part of contraction, closure occurs more quickly than in the first, myofibroblast-independent part. [55]

As the actin in myofibroblasts contracts, the wound edges are pulled together. Fibroblasts lay down collagen to reinforce the wound as myofibroblasts contract. [3] The contraction stage in proliferation ends as myofibroblasts stop contracting and commit apoptosis. [32] The breakdown of the provisional matrix leads to a decrease in hyaluronic acid and an increase in chondroitin sulfate, which gradually triggers fibroblasts to stop migrating and proliferating. [19] These events signal the onset of the maturation stage of wound healing.

Maturation and remodeling

When the levels of collagen production and degradation equalize, the maturation phase of tissue repair is said to have begun. [20] During maturation, type III collagen, which is prevalent during proliferation, is replaced by type I collagen. [17] Originally disorganized collagen fibers are rearranged, cross-linked, and aligned along tension lines. [31] The onset of the maturation phase may vary extensively, depending on the size of the wound and whether it was initially closed or left open, [28] ranging from approximately three days [41] to three weeks. [56] The maturation phase can last for a year or longer, similarly depending on wound type. [28]

As the phase progresses, the tensile strength of the wound increases. [28] Collagen will reach approximately 20% of its tensile strength after three weeks, increasing to 80% after 12 months. The maximum scar strength is 80% of that of unwounded skin. [57] Since activity at the wound site is reduced, the scar loses its red appearance as blood vessels that are no longer needed are removed by apoptosis. [20]

The phases of wound healing normally progress in a predictable, timely manner; if they do not, healing may progress inappropriately to either a chronic wound [7] such as a venous ulcer or pathological scarring such as a keloid scar. [58] [59]

Factors affecting wound healing

Many factors controlling the efficacy, speed, and manner of wound healing fall under two types: local and systemic factors. [2]

Local factors

Systemic factors

In the 2000s there arose the first Mathematical models of the healing process, based on simplified assumptions and on a system of differential equations solved through MATLAB. The models show that the "rate of the healing process" appears to be "highly influenced by the activity and size of the injury itself as well as the activity of the healing agent." [69]

Research and development

Up until about 2000, the classic paradigm of wound healing, involving stem cells restricted to organ-specific lineages, had never been seriously challenged. Since then, the notion of adult stem cells having cellular plasticity or the ability to differentiate into non-lineage cells has emerged as an alternative explanation. [1] To be more specific, hematopoietic progenitor cells (that give rise to mature cells in the blood) may have the ability de-differentiate back into hematopoietic stem cells and/or transdifferentiate into non-lineage cells, such as fibroblasts. [39]

Stem cells and cellular plasticity

Multipotent adult stem cells have the capacity to be self-renewing and give rise to different cell types. Stem cells give rise to progenitor cells, which are cells that are not self-renewing, but can generate several types of cells. The extent of stem cell involvement in cutaneous (skin) wound healing is complex and not fully understood.[ citation needed ] Stem cell injection leads to wound healing primarily through stimulation of angiogenesis. [70]

It is thought that the epidermis and dermis are reconstituted by mitotically active stem cells that reside at the apex of rete ridges (basal stem cells or BSC), the bulge of hair follicles (hair follicular stem cell or HFSC), and the papillary dermis (dermal stem cells). [1] Moreover, bone marrow may also contain stem cells that play a major role in cutaneous wound healing. [39]

In rare circumstances, such as extensive cutaneous injury, self-renewal subpopulations in the bone marrow are induced to participate in the healing process, whereby they give rise to collagen-secreting cells that seem to play a role during wound repair. [1] These two self-renewal subpopulations are (1) bone marrow-derived mesenchymal stem cells (MSC) and (2) hematopoietic stem cells (HSC). Bone marrow also harbors a progenitor subpopulation (endothelial progenitor cells or EPC) that, in the same type of setting, are mobilized to aid in the reconstruction of blood vessels. [39] Moreover, it is thought that extensive injury to skin also promotes the early trafficking of a unique subclass of leukocytes (circulating fibrocytes) to the injured region, where they perform various functions related to wound healing. [1]

Wound repair versus regeneration

An injury is an interruption of morphology and/or functionality of a given tissue. After injury, structural tissue heals with incomplete or complete regeneration. [71] [72] Tissue without an interruption to the morphology almost always completely regenerates. An example of complete regeneration without an interruption of the morphology is non-injured tissue, such as skin. [73] Non-injured skin has a continued replacement and regeneration of cells which always results in complete regeneration. [73]

There is a subtle distinction between 'repair' and 'regeneration'. [1] [71] [72] Repair means incomplete regeneration. [71] Repair or incomplete regeneration, refers to the physiologic adaptation of an organ after injury in an effort to re-establish continuity without regards to exact replacement of lost/damaged tissue. [71] True tissue regeneration or complete regeneration, [72] refers to the replacement of lost/damaged tissue with an 'exact' copy, such that both morphology and functionality are completely restored. [72] Though after injury mammals can completely regenerate spontaneously, they usually do not completely regenerate. An example of a tissue regenerating completely after an interruption of morphology is the endometrium; the endometrium after the process of breakdown via the menstruation cycle heals with complete regeneration. [73]

In some instances, after a tissue breakdown, such as in skin, a regeneration closer to complete regeneration may be induced by the use of biodegradable (collagen-glycoaminoglycan) scaffolds. These scaffolds are structurally analogous to extracellular matrix (ECM) found in normal/un-injured dermis. [74] Fundamental conditions required for tissue regeneration often oppose conditions that favor efficient wound repair, including inhibition of (1) platelet activation, (2) inflammatory response, and (3) wound contraction. [1] In addition to providing support for fibroblast and endothelial cell attachment, biodegradable scaffolds inhibit wound contraction, thereby allowing the healing process to proceed towards a more-regenerative/less-scarring pathway. Pharmaceutical agents have been investigated which may be able to turn off myofibroblast differentiation. [75]

A new way of thinking derived from the notion that heparan sulfates are key player in tissue homeostasis: the process that makes the tissue replace dead cells by identical cells. In wound areas, tissue homeostasis is lost as the heparan sulfates are degraded preventing the replacement of dead cells by identical cells. Heparan sulfate analogues cannot be degraded by all known heparanases and glycanases and bind to the free heparin sulfate binding spots on the ECM, therefore preserving the normal tissue homeostasis and preventing scarring. [76] [77] [78]

Repair or regeneration with regards to hypoxia-inducible factor 1-alpha (HIF-1a). In normal circumstances after injury HIF-1a is degraded by prolyl hydroxylases (PHDs). Scientists found that the simple up-regulation of HIF-1a via PHD inhibitors regenerates lost or damaged tissue in mammals that have a repair response; and the continued down-regulation of Hif-1a results in healing with a scarring response in mammals with a previous regenerative response to the loss of tissue. The act of regulating HIF-1a can either turn off, or turn on the key process of mammalian regeneration. [79] [80]

Scarless wound healing

Scarless wound healing is a concept based on the healing or repair of the skin (or other tissue/organs) after injury with the aim of healing with subjectively and relatively less scar tissue than normally expected. Scarless healing is sometimes mixed up with the concept of scar free healing, which is wound healing which results in absolutely no scar (free of scarring). However they are different concepts.

A reverse to scarless wound healing is scarification (wound healing to scar more). Historically, certain cultures consider scarification attractive; [81] however, this is generally not the case in the modern western society, in which many patients are turning to plastic surgery clinics with unrealistic expectations. Depending on scar type, treatment may be invasive (intralesional steroid injections, surgery) and/or conservative (compression therapy, topical silicone gel, brachytherapy, photodynamic therapy). [82] Clinical judgment is necessary to successfully balance the potential benefits of the various treatments available against the likelihood of a poor response and possible complications resulting from these treatments. Many of these treatments may only have a placebo effect, and the evidence base for the use of many current treatments is poor. [83]

Since the 1960s, comprehension of the basic biologic processes involved in wound repair and tissue regeneration have expanded due to advances in cellular and molecular biology. [84] Currently, the principal goals in wound management are to achieve rapid wound closure with a functional tissue that has minimal aesthetic scarring. [85] However, the ultimate goal of wound healing biology is to induce a more perfect reconstruction of the wound area. Scarless wound healing only occurs in mammalian foetal tissues [86] and complete regeneration is limited to lower vertebrates, such as salamanders, and invertebrates. [87] In adult humans, injured tissue are repaired by collagen deposition, collagen remodelling and eventual scar formation, where fetal wound healing is believed to be more of a regenerative process with minimal or no scar formation. [86] Therefore, foetal wound healing can be used to provide an accessible mammalian model of an optimal healing response in adult human tissues. Clues as to how this might be achieved come from studies of wound healing in embryos, where repair is fast and efficient and results in essentially perfect regeneration of any lost tissue.

The etymology of the term scarless wound healing has a long history. [88] [89] [90] In print the antiquated concept of scarless healing was brought up in the early 20th century and appeared in a paper published in the London Lancet. This process involved cutting at a surgical slant to the skin surface, rather than at a right angle it; the process was described in various Newspapers. [88] [89] [90]

Cancer

After inflammation, restoration of normal tissue integrity and function is preserved by feedback interactions between diverse cell types mediated by adhesion molecules and secreted cytokines. Disruption of normal feedback mechanisms in cancer threatens tissue integrity and enables a malignant tumor to escape the immune system. [91] [92] An example of the importance of the wound healing response within tumors is illustrated in work by Howard Chang and colleagues at Stanford University studying Breast cancers. [8]

Oral collagen supplements

Preliminary results are promising for the short and long-term use of oral collagen supplements for wound healing and skin aging. Oral collagen supplements also increase skin elasticity, hydration, and dermal collagen density. Collagen supplementation is generally safe with no reported adverse events. Further studies are needed to elucidate medical use in skin barrier diseases such as atopic dermatitis and to determine optimal dosing regimens. [93]

Wound dressings

Modern wound dressing to aid in wound repair has undergone considerable research and development in recent years. Scientists aim to develop wound dressings which have the following characteristics: [94]

Cotton gauze dressings have been the standard of care, despite their dry properties that can adhere to wound surfaces and cause discomfort upon removal. Recent research has set out to improve cotton gauze dressings to bring them closer in line to achieve modern wound dressing properties, by coating cotton gauze wound dressing with a chitosan/Ag/ZnO nanocomposite. These updated dressing provide increase water absorbency and improved antibacterial efficacy. [94]

Wound cleansing

Dirt or dust on the surface of the wound, bacteria, tissue that has died, and fluid from the wound may be cleaned. The evidence supporting the most effective technique is not clear and there is insufficient evidence to conclude whether cleaning wounds is beneficial for promoting healing or whether wound cleaning solutions (polyhexamethylene biguanide, aqueous oxygen peroxide, etc.) are better than sterile water or saline solutions to help venous leg ulcers heal. [95] It is uncertain whether the choice of cleaning solution or method of application makes any difference to venous leg ulcer healing. [95]  

Simulating wound healing from a growth perspective

Considerable effort has been devoted to understanding the physical relationships governing wound healing and subsequent scarring, with mathematical models and simulations developed to elucidate these relationships. [96] The growth of tissue around the wound site is a result of the migration of cells and collagen deposition by these cells. The alignment of collagen describes the degree of scarring; basket-weave orientation of collagen is characteristic of normal skin, whereas aligned collagen fibers lead to significant scarring. [97] It has been shown that the growth of tissue and extent of scar formation can be controlled by modulating the stress at a wound site. [98]

The growth of tissue can be simulated using the aforementioned relationships from a biochemical and biomechanical point of view. The biologically active chemicals that play an important role in wound healing are modeled with Fickian diffusion to generate concentration profiles. The balance equation for open systems when modeling wound healing incorporates mass growth due to cell migration and proliferation. Here the following equation is used:

Dtρ0 = Div (R) + R0,

where ρ represents mass density, R represents a mass flux (from cell migration), and R0 represents a mass source (from cell proliferation, division, or enlargement). [99] Relationships like these can be incorporated into an agent-based models, where the sensitivity to single parameters such as initial collagen alignment, cytokine properties, and cell proliferation rates can be tested. [100]

Wound closure intentions

Successful wound healing is dependent on various cell types, molecular mediators and structural elements. [101]

Primary intention

Primary intention is the healing of a clean wound without tissue loss. [101] In this process, wound edges are brought together, so that they are adjacent to each other (re-approximated). Wound closure is performed with sutures (stitches), staples, or adhesive tape or glue.

Primary intention can only be implemented when the wound is precise and there is minimal disruption to the local tissue and the epithelial basement membrane, e.g. surgical incisions. [102]

This process is faster than healing by secondary intention. [101] There is also less scarring associated with primary intention, as there are no large tissue losses to be filled with granulation tissue, though some granulation tissue will form. [101]

Secondary intention

Tertiary intention

(Delayed primary closure):

If the wound edges are not reapproximated immediately, delayed primary wound healing transpires. This type of healing may be desired in the case of contaminated wounds. By the fourth day, phagocytosis of contaminated tissues is well underway, and the processes of epithelization, collagen deposition, and maturation are occurring. Foreign materials are walled off by macrophages that may metamorphose into epithelioid cells, which are encircled by mononuclear leukocytes, forming granulomas. Usually the wound is closed surgically at this juncture, or the scab is eaten, and if the "cleansing" of the wound is incomplete, chronic inflammation can ensue, resulting in prominent scarring.

Overview of involved growth factors

Following are the main growth factors involved in wound healing:

Growth factorAbbreviationMain originsEffects
Epidermal growth factor EGF
Transforming growth factor-α TGF-α
  • Activated macrophages
  • T-lymphocytes
  • Keratinocytes
  • Hepatocyte and epithelial cell proliferation
  • Expression of antimicrobial peptides
  • Expression of chemotactic cytokines
Hepatocyte growth factor HGF
Vascular endothelial growth factor VEGF
  • Mesenchymal cells
  • Vascular permeability
  • Endothelial cell proliferation
Platelet derived growth factor PDGF
  • Platelets
  • Macrophages
  • Endothelial cells
  • Smooth muscle cells
  • Keratinocytes
Fibroblast growth factor 1 and 2FGF-1, −2
  • Macrophages
  • Mast cells
  • T-lymphocytes
  • Endothelial cells
  • Fibroblasts
  • Fibroblast chemotaxis
  • Fibroblast and keratinocyte proliferation
  • Keratinocyte migration
  • Angiogenesis
  • Wound contraction
  • Matrix (collagen fibers) deposition
Transforming growth factor-β TGF-β
  • Platelets
  • T-lymphocytes
  • Macrophages
  • Endothelial cells
  • Keratinocytes
  • Smooth muscle cells
  • Fibroblasts
  • Granulocyte, macrophage, lymphocyte, fibroblast and smooth muscle cell chemotaxis
  • TIMP synthesis
  • Angiogenesis
  • Fibroplasia
  • Matrix metalloproteinase production inhibition
  • Keratinocyte proliferation
Keratinocyte growth factor KGF
  • Keratinocytes
  • Keratinocyte migration, proliferation and differentiation
Unless else specified in boxes, then reference is: [107]

Complications of wound healing

The major complications are many:

  1. Deficient scar formation: Results in wound dehiscence or rupture of the wound due to inadequate formation of granulation tissue.
  2. Excessive scar formation: Hypertrophic scar, keloid, desmoid.
  3. Exuberant granulation (proud flesh).
  4. Deficient contraction (in skin grafts) or excessive contraction (in burns).
  5. Others: Dystrophic calcification, pigmentary changes, painful scars, incisional hernia

Other complications can include infection and Marjolin's ulcer.

Biologics, skin substitutes, biomembranes and scaffolds

Advancements in the clinical understanding of wounds and their pathophysiology have commanded significant biomedical innovations in the treatment of acute, chronic, and other types of wounds. Many biologics, skin substitutes, biomembranes and scaffolds have been developed to facilitate wound healing through various mechanisms. [108] This includes a number of products under the trade names such as Epicel, Laserskin, Transcyte, Dermagraft, AlloDerm/Strattice, Biobrane, Integra, Apligraf, OrCel, GraftJacket and PermaDerm. [109]

See also

Notes and references

  1. 1 2 3 4 5 6 7 8 9 10 11 12 Nguyen DT, Orgill DP, Murphy GT (2009). "4 The Pathophysiologic Basis for Wound Healing and Cutaneous Regeneration". In Orgill DP, Blanco C (eds.). Biomaterials for Treating Skin Loss. Elsevier. pp. 25–57. ISBN   978-1-84569-554-5.
  2. 1 2 Rieger S, Zhao H, Martin P, Abe K, Lisse TS (January 2015). "The role of nuclear hormone receptors in cutaneous wound repair". Cell Biochemistry and Function. 33 (1): 1–13. doi:10.1002/cbf.3086. PMC   4357276 . PMID   25529612.
  3. 1 2 3 4 5 6 7 8 9 10 11 12 Stadelmann WK, Digenis AG, Tobin GR (August 1998). "Physiology and healing dynamics of chronic cutaneous wounds". American Journal of Surgery. 176 (2A Suppl): 26S–38S. doi:10.1016/S0002-9610(98)00183-4. PMID   9777970.
  4. Enoch, S. Price, P. (2004). Cellular, molecular and biochemical differences in the pathophysiology of healing between acute wounds, chronic wounds and wounds in the elderly Archived 2017-07-06 at the Wayback Machine .
  5. Rasche H (2001). "Haemostasis and thrombosis: an overview". European Heart Journal Supplements. 3 (Supplement Q): Q3–Q7. doi: 10.1016/S1520-765X(01)90034-3 .
  6. Versteeg HH, Heemskerk JW, Levi M, Reitsma PH (January 2013). "New fundamentals in hemostasis". Physiological Reviews. 93 (1): 327–58. doi:10.1152/physrev.00016.2011. PMID   23303912. S2CID   10367343.
  7. 1 2 3 4 5 6 7 8 Midwood KS, Williams LV, Schwarzbauer JE (June 2004). "Tissue repair and the dynamics of the extracellular matrix". The International Journal of Biochemistry & Cell Biology. 36 (6): 1031–7. doi:10.1016/j.biocel.2003.12.003. PMID   15094118.
  8. 1 2 Chang HY, Sneddon JB, Alizadeh AA, Sood R, West RB, Montgomery K, et al. (February 2004). "Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds". PLOS Biology. 2 (2): E7. doi: 10.1371/journal.pbio.0020007 . PMC   314300 . PMID   14737219.
  9. Garg, H.G. (2000). Scarless Wound Healing. New York Marcel Dekker, Inc. Electronic book.
  10. Reference list is found on image main page.
  11. Cubison TC, Pape SA, Parkhouse N (December 2006). "Evidence for the link between healing time and the development of hypertrophic scars (HTS) in paediatric burns due to scald injury". Burns. 32 (8): 992–9. doi:10.1016/j.burns.2006.02.007. PMID   16901651.
  12. Kraft J, Lynde C. "Giving Burns the First, Second and Third Degree - Classification of burns". skincareguide.ca. Archived from the original on 26 December 2011. Retrieved 31 January 2012. Formation of a thick eschar, slow healing (>1month), Obvious scarring,
  13. 1 2 "POST BURN SCAR RELATIVE TO RE-EPITHELIALIZATION". Burnsurgery.org. 2011. Archived from the original on 25 April 2012. Retrieved 16 March 2011. Healing in 2 weeks – minimal to no scar; Healing in 3 weeks – minimal to no scar except in high risk scar formers;Healing in 4 weeks or more – hypertrophic in more than 50% of patients
  14. Galko MJ, Krasnow MA (August 2004). "Cellular and genetic analysis of wound healing in Drosophila larvae". PLOS Biology. 2 (8): E239. doi: 10.1371/journal.pbio.0020239 . PMC   479041 . PMID   15269788.
  15. 1 2 3 4 5 Rosenberg L., de la Torre J. (2006). Wound Healing, Growth Factors Archived 2008-11-21 at the Wayback Machine . Emedicine.com. Accessed January 20, 2008.
  16. Sandeman SR, Allen MC, Liu C, Faragher RG, Lloyd AW (November 2000). "Human keratocyte migration into collagen gels declines with in vitro ageing". Mechanisms of Ageing and Development. 119 (3): 149–57. doi:10.1016/S0047-6374(00)00177-9. PMID   11080534. S2CID   21115977.
  17. 1 2 3 4 Dealey C. (1999). The care of wounds: A guide for nurses. Oxford; Malden, Mass. Blackwell Science. Electronic book.
  18. Theoret CL (2004). "Update on wound repair". Clinical Techniques in Equine Practice. 3 (2): 110–122. doi:10.1053/j.ctep.2004.08.009.
  19. 1 2 3 4 5 6 7 8 de la Torre J., Sholar A. (2006). Wound healing: Chronic wounds Archived 2008-10-29 at the Wayback Machine . Emedicine.com. Accessed January 20, 2008.
  20. 1 2 3 4 5 6 7 8 Greenhalgh DG (September 1998). "The role of apoptosis in wound healing". The International Journal of Biochemistry & Cell Biology. 30 (9): 1019–30. doi:10.1016/S1357-2725(98)00058-2. PMID   9785465.
  21. Muller MJ, Hollyoak MA, Moaveni Z, Brown TL, Herndon DN, Heggers JP (December 2003). "Retardation of wound healing by silver sulfadiazine is reversed by Aloe vera and nystatin". Burns. 29 (8): 834–6. doi:10.1016/S0305-4179(03)00198-0. PMID   14636760.
  22. Martin P, Leibovich SJ (November 2005). "Inflammatory cells during wound repair: the good, the bad and the ugly". Trends in Cell Biology. 15 (11): 599–607. doi:10.1016/j.tcb.2005.09.002. PMID   16202600.
  23. 1 2 3 4 5 6 7 8 Santoro MM, Gaudino G (March 2005). "Cellular and molecular facets of keratinocyte reepithelization during wound healing". Experimental Cell Research. 304 (1): 274–86. doi:10.1016/j.yexcr.2004.10.033. PMID   15707592.
  24. 1 2 "The phases of cutaneous wound healing" (PDF). Expert Reviews in Molecular Medicine. Cambridge University Press. 5. 21 March 2003. Archived from the original (PDF) on 8 March 2008.
  25. 1 2 3 4 5 6 Deodhar AK, Rana RE (1997). "Surgical physiology of wound healing: a review". Journal of Postgraduate Medicine. 43 (2): 52–6. PMID   10740722. Archived from the original on 2011-02-26. Retrieved 2005-10-27.
  26. Ovchinnikov DA (September 2008). "Macrophages in the embryo and beyond: much more than just giant phagocytes". Genesis. 46 (9): 447–62. doi: 10.1002/dvg.20417 . PMID   18781633. S2CID   38894501. Macrophages are present essentially in all tissues, beginning with embryonic development and, in addition to their role in host defense and in the clearance of apoptotic cells, are being increasingly recognized for their trophic function and role in regeneration.
  27. 1 2 3 4 Newton PM, Watson JA, Wolowacz RG, Wood EJ (August 2004). "Macrophages restrain contraction of an in vitro wound healing model". Inflammation. 28 (4): 207–14. doi:10.1023/B:IFLA.0000049045.41784.59. PMID   15673162. S2CID   9612298.
  28. 1 2 3 4 5 6 7 8 Mercandetti M, Cohen AJ (2005). "Wound Healing: Healing and Repair". Emedicine.com. Archived from the original on 21 November 2008. Retrieved 20 January 2008.
  29. Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P, Figueiredo JL, Kohler RH, et al. (July 2009). "Identification of splenic reservoir monocytes and their deployment to inflammatory sites". Science. 325 (5940): 612–6. Bibcode:2009Sci...325..612S. doi:10.1126/science.1175202. PMC   2803111 . PMID   19644120.
  30. Jia T, Pamer EG (July 2009). "Immunology. Dispensable but not irrelevant". Science. 325 (5940): 549–50. Bibcode:2009Sci...325..549J. doi:10.1126/science.1178329. PMC   2917045 . PMID   19644100.
  31. 1 2 3 4 5 6 7 8 9 Lorenz HP, Longaker MT (2008). "Wounds: Biology, Pathology, and Management" (PDF). In Norton JA (ed.). Surgery. New York, NY: Springer. pp. 191–208. doi:10.1007/978-0-387-68113-9_10. ISBN   978-0-387-30800-5. S2CID   83849346. Archived from the original (PDF) on 24 August 2014.
  32. 1 2 3 4 5 Hinz B (April 2006). "Masters and servants of the force: the role of matrix adhesions in myofibroblast force perception and transmission". European Journal of Cell Biology. 85 (3–4): 175–81. doi:10.1016/j.ejcb.2005.09.004. PMID   16546559.
  33. Souppouris A (2013-05-23). "Scientists identify cell that could hold the secret to limb regeneration". the verge.com. Archived from the original on 2017-07-31. Retrieved 2017-09-18. Researchers have identified a cell that aids limb regrowth in Salamanders. Macrophages are a type of repairing cell that devour dead cells and pathogens, and trigger other immune cells to respond to pathogens.
  34. Godwin JW, Pinto AR, Rosenthal NA (June 2013). "Macrophages are required for adult salamander limb regeneration". Proceedings of the National Academy of Sciences of the United States of America. University of Texas. 110 (23): 9415–20. Bibcode:2013PNAS..110.9415G. doi: 10.1073/pnas.1300290110 . PMC   3677454 . PMID   23690624.
  35. 1 2 3 4 Falanga V. (2005). Wound Healing. American Academy of Dermatology (AAD).
  36. 1 2 3 Kuwahara R.T. and Rasberry R. 2007. Chemical Peels Archived 2008-10-25 at the Wayback Machine . Emedicine.com. Accessed September 15, 2007.
  37. 1 2 3 4 5 6 7 8 9 10 Romo T. and Pearson J.M. 2005. Wound Healing, Skin Archived 2008-12-07 at the Wayback Machine . Emedicine.com. Accessed December 27, 2006.
  38. Lansdown AB, Sampson B, Rowe A (February 2001). "Experimental observations in the rat on the influence of cadmium on skin wound repair". International Journal of Experimental Pathology. 82 (1): 35–41. doi:10.1046/j.1365-2613.2001.00180.x. PMC   2517695 . PMID   11422539.
  39. 1 2 3 4 Song G, Nguyen DT, Pietramaggiori G, Scherer S, Chen B, Zhan Q, Ogawa R, Yannas IV, Wagers AJ, Orgill DP, Murphy GF (2010). "Use of the parabiotic model in studies of cutaneous wound healing to define the participation of circulating cells". Wound Repair and Regeneration. 18 (4): 426–32. doi:10.1111/j.1524-475X.2010.00595.x. PMC   2935287 . PMID   20546556.
  40. Ruszczak Z (November 2003). "Effect of collagen matrices on dermal wound healing". Advanced Drug Delivery Reviews. 55 (12): 1595–611. doi:10.1016/j.addr.2003.08.003. PMID   14623403.
  41. 1 2 Fig. 9-1. The cellular, biochemical, and mechanical phases of wound healing. Pollock RE, Brunicardi FC, Andersen DK, Billiar TR, Dunn D, Hunter JG, Matthews JJ (2009). Schwartz's Principles of Surgery, Ninth Edition. McGraw-Hill Professional. ISBN   978-0-07-154769-7.
  42. 1 2 3 4 5 6 7 8 DiPietro LA, Burns AL, eds. (2003). Wound Healing: Methods and Protocols. Methods in Molecular Medicine. Totowa, N.J.: Humana Press.
  43. Fu XB, Sun TZ, Li XK, Sheng ZY (February 2005). "Morphological and distribution characteristics of sweat glands in hypertrophic scar and their possible effects on sweat gland regeneration". Chinese Medical Journal. 118 (3): 186–91. PMID   15740645. Archived from the original on 2018-06-20. Retrieved 2014-06-18.
  44. 1 2 "BURN INJURIES". nationaltraumainstitute.org. Archived from the original on 3 March 2016. Retrieved 13 July 2016. When the dermis is destroyed, the scars do not regrow hair, nerves or sweat glands, providing additional challenges to body temperature control.
  45. 1 2 3 4 Bartkova J, Grøn B, Dabelsteen E, Bartek J (February 2003). "Cell-cycle regulatory proteins in human wound healing". Archives of Oral Biology. 48 (2): 125–32. doi:10.1016/S0003-9969(02)00202-9. PMID   12642231.
  46. 1 2 3 4 Mulvaney M. and Harrington A. 1994. Chapter 7: Cutaneous trauma and its treatment. In, Textbook of Military Medicine: Military Dermatology. Office of the Surgeon General, Department of the Army. Virtual Naval Hospital Project. Accessed through web archive on September 15, 2007.
  47. 1 2 3 4 Larjava H., Koivisto L., and Hakkinen L. 2002. Chapter 3: Keratinocyte Interactions with Fibronectin During Wound Healing. In, Heino, J. and Kahari, V.M. Cell Invasion. Medical Intelligence Unit; 33. Georgetown, Tex., Austin, Tex Landes Bioscience, Inc. Electronic book.
  48. Witte MB, Barbul A (April 2002). "Role of nitric oxide in wound repair". American Journal of Surgery. 183 (4): 406–12. doi:10.1016/S0002-9610(02)00815-2. PMID   11975928.
  49. Son HJ, Bae HC, Kim HJ, Lee DH, Han D, Park J (2005). "Effects of β-glucan on proliferation and migration of fibroblasts". Current Applied Physics. 5 (5): 468–71. Bibcode:2005CAP.....5..468S. doi:10.1016/j.cap.2005.01.011.
  50. Falanga V (2004). "The chronic wound: impaired healing and solutions in the context of wound bed preparation". Blood Cells, Molecules & Diseases. 32 (1): 88–94. doi:10.1016/j.bcmd.2003.09.020. PMID   14757419.
  51. Etscheid M, Beer N, Dodt J (December 2005). "The hyaluronan-binding protease upregulates ERK1/2 and PI3K/Akt signalling pathways in fibroblasts and stimulates cell proliferation and migration". Cellular Signalling. 17 (12): 1486–94. doi:10.1016/j.cellsig.2005.03.007. PMID   16153533.
  52. Bayram Y, Deveci M, Imirzalioglu N, Soysal Y, Sengezer M (October 2005). "The cell based dressing with living allogenic keratinocytes in the treatment of foot ulcers: a case study". British Journal of Plastic Surgery. 58 (7): 988–96. doi: 10.1016/j.bjps.2005.04.031 . PMID   16040019.
  53. Grinnell F (February 1994). "Fibroblasts, myofibroblasts, and wound contraction". The Journal of Cell Biology. 124 (4): 401–4. doi:10.1083/jcb.124.4.401. PMC   2119916 . PMID   8106541.
  54. 1 2 Eichler MJ, Carlson MA (February 2006). "Modeling dermal granulation tissue with the linear fibroblast-populated collagen matrix: a comparison with the round matrix model". Journal of Dermatological Science. 41 (2): 97–108. doi:10.1016/j.jdermsci.2005.09.002. PMID   16226016.
  55. 1 2 3 4 Mirastschijski U, Haaksma CJ, Tomasek JJ, Agren MS (October 2004). "Matrix metalloproteinase inhibitor GM 6001 attenuates keratinocyte migration, contraction and myofibroblast formation in skin wounds". Experimental Cell Research. 299 (2): 465–75. doi:10.1016/j.yexcr.2004.06.007. PMID   15350544.
  56. worldwidewounds.com Archived 2011-07-05 at the Wayback Machine > Figure 3 – The time relationship between the different processes of wound healing. Archived 2011-07-18 at the Wayback Machine by Gregory S Schultz, Glenn Ladwig and Annette Wysocki – in turn adapted from Asmussen PD, Sollner B. Mechanism of wound healing. In: Wound Care. Tutorial Medical Series. Stuttgart: Hippokrates Verlag, 1993.
  57. Morton LM, Phillips TJ (April 2016). "Wound healing and treating wounds: Differential diagnosis and evaluation of chronic wounds". Journal of the American Academy of Dermatology. 74 (4): 589–605, quiz 605–6. doi:10.1016/j.jaad.2015.08.068. PMID   26979352.
  58. O'Leary R, Wood EJ, Guillou PJ (2002). "Pathological scarring: strategic interventions". The European Journal of Surgery = Acta Chirurgica. 168 (10): 523–34. PMID   12666691.
  59. Desmoulière A, Chaponnier C, Gabbiani G (2005). "Tissue repair, contraction, and the myofibroblast". Wound Repair and Regeneration. 13 (1): 7–12. doi:10.1111/j.1067-1927.2005.130102.x. PMID   15659031. S2CID   2590702.
  60. Metzger S (September 2004). "Clinical and financial advantages of moist wound management". Home Healthcare Nurse. 22 (9): 586–90. doi:10.1097/00004045-200409000-00003. PMID   15359168.
  61. Iconomou TG, Zuker RM, Michelow BJ (1993). "Management of major penetrating glass injuries to the upper extremities in children and adolescents". Microsurgery. 14 (2): 91–6. doi:10.1002/micr.1920140202. PMID   8469109. S2CID   25492817.
  62. "Nerve injury". Johns Hopkins Medicine. The Johns Hopkins University, The Johns Hopkins Hospital, and Johns Hopkins Health System. Archived from the original on 27 September 2016. Retrieved 2 October 2016.
  63. Brem H, Tomic-Canic M (May 2007). "Cellular and molecular basis of wound healing in diabetes". The Journal of Clinical Investigation. 117 (5): 1219–22. doi:10.1172/jci32169. PMC   1857239 . PMID   17476353.
  64. 1 2 3 4 5 6 7 8 Guo S, Dipietro LA (March 2010). "Factors affecting wound healing". Journal of Dental Research. 89 (3): 219–29. doi:10.1177/0022034509359125. PMC   2903966 . PMID   20139336.
  65. Arnold M, Barbul A (June 2006). "Nutrition and wound healing". Plastic and Reconstructive Surgery. 117 (7 Suppl): 42S–58S. doi:10.1097/01.prs.0000225432.17501.6c. PMID   16799374. S2CID   8658373.
  66. Wong LS, Green HM, Feugate JE, Yadav M, Nothnagel EA, Martins-Green M (April 2004). "Effects of "second-hand" smoke on structure and function of fibroblasts, cells that are critical for tissue repair and remodeling". BMC Cell Biology. 5 (1): 13. doi: 10.1186/1471-2121-5-13 . PMC   400727 . PMID   15066202.
  67. Gosain A, DiPietro LA (March 2004). "Aging and wound healing". World Journal of Surgery. 28 (3): 321–6. doi:10.1007/s00268-003-7397-6. PMID   14961191. S2CID   28491255.
  68. Swift ME, Burns AL, Gray KL, DiPietro LA (November 2001). "Age-related alterations in the inflammatory response to dermal injury". The Journal of Investigative Dermatology. 117 (5): 1027–35. doi: 10.1046/j.0022-202x.2001.01539.x . PMID   11710909.
  69. Alfredo Palomino I.; Leighton Estrada R.; Javier Valeriano M.; Sergio Luque M. (September 24, 2019). "Mathematical modeling of injury healing process under the action of an active pharmaceutical ingredient (API)" [Modelamiento matemático del proceso de sanación de heridas bajo la acción de un ingrediente farmacéuticamente activo (IFA).]. Selecciones Matemáticas (in American English and Spanish). 6 (2): 283–288. doi: 10.17268/sel.mat.2019.02.14 . ISSN   2411-1783. OCLC   8469127433. Archived from the original on July 22, 2020. Retrieved August 29, 2020.
  70. Krasilnikova, O. A.; Baranovskii, D. S.; Lyundup, A. V.; Shegay, P. V.; Kaprin, A. D.; Klabukov, I. D. (2022-04-27). "Stem and Somatic Cell Monotherapy for the Treatment of Diabetic Foot Ulcers: Review of Clinical Studies and Mechanisms of Action". Stem Cell Reviews and Reports. 18 (6): 1974–1985. doi:10.1007/s12015-022-10379-z. ISSN   2629-3277. PMID   35476187. S2CID   248402820.
  71. 1 2 3 4 Min S, Wang SW, Orr W (2006). "Graphic general pathology: 2.3 Incomplete regeneration". Pathology. pathol.med.stu.edu.cn. Archived from the original on 2013-11-10. Retrieved 2012-12-07. The new tissue is not the same as the tissue that was lost. After the repair process has been completed, there is a loss in the structure or function of the injured tissue. In this type of repair, it is common that granulation tissue (stromal connective tissue) proliferates to fill the defect created by the necrotic cells. The necrotic cells are then replaced by scar tissue.
  72. 1 2 3 4 Min S, Wang SW, Orr W (2006). "Graphic general pathology: 2.2 complete regeneration". Pathology. pathol.med.stu.edu.cn. Archived from the original on 2012-12-07. Retrieved 2012-12-07. (1) Complete regeneration: The new tissue is the same as the tissue that was lost. After the repair process has been completed, the structure and function of the injured tissue are completely normal
  73. 1 2 3 Min S, Wang SW, Orr W (2006). "Graphic general pathology: 2.2 complete regeneration". Pathology. pathol.med.stu.edu.cn. Archived from the original on 2012-12-07. Retrieved 2013-11-10. After the repair process has been completed, the structure and function of the injured tissue are completely normal. This type of regeneration is common in physiological situations. Examples of physiological regeneration are the continual replacement of cells of the skin and repair of the endometrium after menstruation. Complete regeneration can occur in pathological situations in tissues that have good regenerative capacity.
  74. Yannas IV, Lee E, Orgill DP, Skrabut EM, Murphy GF (February 1989). "Synthesis and characterization of a model extracellular matrix that induces partial regeneration of adult mammalian skin". Proceedings of the National Academy of Sciences of the United States of America. 86 (3): 933–7. Bibcode:1989PNAS...86..933Y. doi: 10.1073/pnas.86.3.933 . JSTOR   33315. PMC   286593 . PMID   2915988.
  75. O'Leary R, Ponnambalam S, Wood EJ (September 2003). "Pioglitazone-induced myofibroblast cell death: implications for cutaneous scarring". The British Journal of Dermatology. 149 (3): 665–7. doi:10.1046/j.1365-2133.2003.05501.x. PMID   14511015. S2CID   45852269.
  76. Tong M, Tuk B, Hekking IM, Vermeij M, Barritault D, van Neck JW (2009). "Stimulated neovascularization, inflammation resolution and collagen maturation in healing rat cutaneous wounds by a heparan sulfate glycosaminoglycan mimetic, OTR4120". Wound Repair and Regeneration. 17 (6): 840–52. doi:10.1111/j.1524-475X.2009.00548.x. PMID   19903305. S2CID   17262546.
  77. Barritault D, Caruelle JP (March 2006). "[Regenerating agents (RGTAs): a new therapeutic approach]" [Regenerating agents (RGTAs): a new therapeutic approach]. Annales Pharmaceutiques Françaises (in French). 64 (2): 135–44. doi:10.1016/S0003-4509(06)75306-8. PMID   16568015.
  78. Van Neck et al, Heparan sulfate proteoglycan mimetics thrive tissue regeneration: an overview. In Intech book under the working title "Tissue Regeneration", ISBN   978-953-307-876-2 is scheduled for on line publication on Nov 26, 2011"[ page needed ]
  79. eurekalert.org staff (3 June 2015). "Scientist at LIMR leads study demonstrating drug-induced tissue regeneration". eurekalert.org. Lankenau Institute for Medical Research (LIMR). Archived from the original on 4 July 2015. Retrieved 3 July 2015.
  80. Zhang Y, Strehin I, Bedelbaeva K, Gourevitch D, Clark L, Leferovich J, Messersmith PB, Heber-Katz E. Drug-induced regeneration in adult mice. Sci Transl Med. 2015;290.
  81. Rush, J. (2005). Spiritual tattoo: a cultural history of tattooing, piercing, scarification, branding, and implants, Frog Ltd.
  82. Brown BC, McKenna SP, Siddhi K, McGrouther DA, Bayat A (September 2008). "The hidden cost of skin scars: quality of life after skin scarring". Journal of Plastic, Reconstructive & Aesthetic Surgery. 61 (9): 1049–58. doi:10.1016/j.bjps.2008.03.020. PMID   18617450.
  83. Bayat A, McGrouther DA, Ferguson MW (January 2003). "Skin scarring". BMJ. 326 (7380): 88–92. doi:10.1136/bmj.326.7380.88. PMC   1125033 . PMID   12521975.
  84. Clark, R. (1996). The molecular and cellular biology of wound repair, Springer Us.
  85. Tonnesen MG, Feng X, Clark RA (December 2000). "Angiogenesis in wound healing". The Journal of Investigative Dermatology. Symposium Proceedings. 5 (1): 40–6. doi: 10.1046/j.1087-0024.2000.00014.x . PMID   11147674.
  86. 1 2 Ferguson MW, Whitby DJ, Shah M, Armstrong J, Siebert JW, Longaker MT (April 1996). "Scar formation: the spectral nature of fetal and adult wound repair". Plastic and Reconstructive Surgery. 97 (4): 854–60. doi:10.1097/00006534-199604000-00029. PMID   8628785.
  87. Brockes JP, Kumar A, Velloso CP (2001). "Regeneration as an evolutionary variable". Journal of Anatomy. 199 (Pt 1–2): 3–11. doi:10.1046/j.1469-7580.2001.19910003.x. PMC   1594962 . PMID   11523827.
  88. 1 2 "Scarless Healing". Star. Christchurch, New Zealand. 1906-07-07. pp. Page 4. Archived from the original on 2013-10-08. Retrieved 2013-07-02.
  89. 1 2 "Scarless Healing". Marlborough Express, Volume XXXIX, Issue 160. paperspast.natlib.govt.nz. 1906-07-12. pp. Page 1. Archived from the original on 2013-10-08. Retrieved 2013-07-02.
  90. 1 2 "A Wonderful New Surgery". Reading Eagle. 1906-07-06. pp. Page 6. Archived from the original on 2016-03-12. Retrieved 2013-07-02.
  91. Karin M, Clevers H (January 2016). "Reparative inflammation takes charge of tissue regeneration". Nature. 529 (7586): 307–15. Bibcode:2016Natur.529..307K. doi:10.1038/nature17039. PMC   5228603 . PMID   26791721.
  92. Vlahopoulos SA (August 2017). "Aberrant control of NF-κB in cancer permits transcriptional and phenotypic plasticity, to curtail dependence on host tissue: molecular mode". Cancer Biology & Medicine. 14 (3): 254–270. doi:10.20892/j.issn.2095-3941.2017.0029. PMC   5570602 . PMID   28884042.
  93. J Drugs Dermatol. 2019;18(1):9-16.
  94. 1 2 Zeng R, Lin C, Lin Z, Chen H, Lu W, Lin C, Li H (November 2018). "Approaches to cutaneous wound healing: basics and future directions". Cell and Tissue Research. 374 (2): 217–232. doi:10.1007/s00441-018-2830-1. PMID   29637308. S2CID   4727434.
  95. 1 2 McLain NE, Moore ZE, Avsar P, et al. (Cochrane Wounds Group) (March 2021). "Wound cleansing for treating venous leg ulcers". The Cochrane Database of Systematic Reviews. 2021 (3): CD011675. doi:10.1002/14651858.CD011675.pub2. PMC   8092712 . PMID   33734426.
  96. Cumming BD, McElwain DL, Upton Z (January 2010). "A mathematical model of wound healing and subsequent scarring". Journal of the Royal Society, Interface. 7 (42): 19–34. doi:10.1098/rsif.2008.0536. PMC   2839370 . PMID   19324672.
  97. Gurtner GC, Werner S, Barrandon Y, Longaker MT (May 2008). "Wound repair and regeneration". Nature. 453 (7193): 314–21. Bibcode:2008Natur.453..314G. doi:10.1038/nature07039. PMID   18480812. S2CID   205213660.
  98. Gurtner GC, Dauskardt RH, Wong VW, Bhatt KA, Wu K, Vial IN, Padois K, Korman JM, Longaker MT (August 2011). "Improving cutaneous scar formation by controlling the mechanical environment: large animal and phase I studies". Annals of Surgery. 254 (2): 217–25. doi:10.1097/SLA.0b013e318220b159. PMID   21606834. S2CID   21111114.
  99. Kuhl E, Steinmann P (June 2004). "Computational modeling of healing: an application of the material force method". Biomechanics and Modeling in Mechanobiology. 2 (4): 187–203. doi:10.1007/s10237-003-0034-3. PMID   14872320. S2CID   8070456.
  100. Rouillard AD, Holmes JW (September 2012). "Mechanical regulation of fibroblast migration and collagen remodelling in healing myocardial infarcts". The Journal of Physiology. 590 (18): 4585–602. doi:10.1113/jphysiol.2012.229484. PMC   3477759 . PMID   22495588.
  101. 1 2 3 4 Velnar T, Bailey T, Smrkolj V (2009-10-01). "The wound healing process: an overview of the cellular and molecular mechanisms". The Journal of International Medical Research. 37 (5): 1528–42. doi: 10.1177/147323000903700531 . PMID   19930861. S2CID   2496871.
  102. 1 2 Armitage J, Lockwood S (2011-10-01). "Skin incisions and wound closure". Surgery (Oxford). Wound Management. 29 (10): 496–501. doi:10.1016/j.mpsur.2011.06.022.
  103. Toon CD, Lusuku C, Ramamoorthy R, Davidson BR, Gurusamy KS, et al. (Cochrane Wounds Group) (September 2015). "Early versus delayed dressing removal after primary closure of clean and clean-contaminated surgical wounds". The Cochrane Database of Systematic Reviews. 2015 (9): CD010259. doi:10.1002/14651858.CD010259.pub3. PMC   7087443 . PMID   26331392.
  104. Norman G, Dumville JC, Mohapatra DP, Owens GL, Crosbie EJ (March 2016). "Antibiotics and antiseptics for surgical wounds healing by secondary intention". The Cochrane Database of Systematic Reviews. 3 (4): CD011712. doi:10.1002/14651858.CD011712.pub2. PMC   6599835 . PMID   27021482.
  105. Vermeulen H, Ubbink D, Goossens A, de Vos R, Legemate D, et al. (Cochrane Wounds Group) (2004-01-26). "Dressings and topical agents for surgical wounds healing by secondary intention". The Cochrane Database of Systematic Reviews. 2012 (2): CD003554. doi:10.1002/14651858.CD003554.pub2. PMC   8407283 . PMID   15106207.
  106. Dumville JC, Owens GL, Crosbie EJ, Peinemann F, Liu Z, et al. (Cochrane Wounds Group) (June 2015). "Negative pressure wound therapy for treating surgical wounds healing by secondary intention". The Cochrane Database of Systematic Reviews (6): CD011278. doi:10.1002/14651858.CD011278.pub2. PMID   26042534.
  107. Table 3-1 in: Mitchell RS, Kumar V, Abbas AK, Nelson F (2007). Robbins Basic Pathology (8th ed.). Philadelphia: Saunders. ISBN   978-1-4160-2973-1.
  108. Stejskalová A, Almquist BD (July 2017). "Using biomaterials to rewire the process of wound repair". Biomaterials Science. 5 (8): 1421–1434. doi:10.1039/c7bm00295e. PMC   5576529 . PMID   28692083.
  109. Vyas KS, Vasconez HC. Wound Healing: Biologics, Skin Substitutes, Biomembranes and Scaffolds Archived 2017-04-13 at the Wayback Machine . Healthcare. 2014; 2(3):356-400.

Related Research Articles

A growth factor is a naturally occurring substance capable of stimulating cell proliferation, wound healing, and occasionally cellular differentiation. Usually it is a secreted protein or a steroid hormone. Growth factors are important for regulating a variety of cellular processes.

<span class="mw-page-title-main">Scar</span> Area of fibrous tissue that replaces normal skin after an injury

A scar is an area of fibrous tissue that replaces normal skin after an injury. Scars result from the biological process of wound repair in the skin, as well as in other organs, and tissues of the body. Thus, scarring is a natural part of the healing process. With the exception of very minor lesions, every wound results in some degree of scarring. An exception to this are animals with complete regeneration, which regrow tissue without scar formation.

<span class="mw-page-title-main">Fibronectin</span> Protein involved in cell adhesion, cell growth, cell migration and differentiation

Fibronectin is a high-molecular weight glycoprotein of the extracellular matrix that binds to membrane-spanning receptor proteins called integrins. It is approved for marketing as a topical solution in India by Central Drugs Standard Control organization in 2020 under the brand name FIBREGA for chronic wounds. Fibronectin also binds to other extracellular matrix proteins such as collagen, fibrin, and heparan sulfate proteoglycans.

<span class="mw-page-title-main">Extracellular matrix</span> Network of proteins and molecules outside cells that provides structural support for cells

In biology, the extracellular matrix (ECM), also called intercellular matrix (ICM), is a network consisting of extracellular macromolecules and minerals, such as collagen, enzymes, glycoproteins and hydroxyapatite that provide structural and biochemical support to surrounding cells. Because multicellularity evolved independently in different multicellular lineages, the composition of ECM varies between multicellular structures; however, cell adhesion, cell-to-cell communication and differentiation are common functions of the ECM.

<span class="mw-page-title-main">Healing</span> Process of the restoration of health

With physical trauma or disease suffered by an organism, healing involves the repairing of damaged tissue(s), organs and the biological system as a whole and resumption of (normal) functioning. Medicine includes the process by which the cells in the body regenerate and repair to reduce the size of a damaged or necrotic area and replace it with new living tissue. The replacement can happen in two ways: by regeneration in which the necrotic cells are replaced by new cells that form "like" tissue as was originally there; or by repair in which injured tissue is replaced with scar tissue. Most organs will heal using a mixture of both mechanisms.

Haptotaxis is the directional motility or outgrowth of cells, e.g. in the case of axonal outgrowth, usually up a gradient of cellular adhesion sites or substrate-bound chemoattractants. These gradients are naturally present in the extracellular matrix (ECM) of the body during processes such as angiogenesis or artificially present in biomaterials where gradients are established by altering the concentration of adhesion sites on a polymer substrate.

Granulation tissue is new connective tissue and microscopic blood vessels that form on the surfaces of a wound during the healing process. Granulation tissue typically grows from the base of a wound and is able to fill wounds of almost any size. Examples of granulation tissue can be seen in pyogenic granulomas and pulp polyps. Its histological appearance is characterized by proliferation of fibroblasts and thin-walled, delicate capillaries (angiogenesis), and infiltrated inflammatory cells in a loose extracellular matrix.

Protection from mechanical injury, chemical hazards, and bacterial invasion is provided by the skin because the epidermis is relatively thick and covered with keratin. Secretions from sebaceous glands and sweat glands also benefit this protective barrier. In the event of an injury that damages the skin's protective barrier, the body triggers a response called wound healing. After hemostasis, inflammation white blood cells, including phagocytic macrophages arrive at the injury site. Once the invading microorganisms have been brought under control, the skin proceeds to heal itself. The ability of the skin to heal even after considerable damage has occurred is due to the presence of stem cells in the dermis and cells in the stratum basale of the epidermis, all of which can generate new tissue.

A chronic wound is a wound that does not heal in an orderly set of stages and in a predictable amount of time the way most wounds do; wounds that do not heal within three months are often considered chronic. Chronic wounds seem to be detained in one or more of the phases of wound healing. For example, chronic wounds often remain in the inflammatory stage for too long. To overcome that stage and jump-start the healing process, a number of factors need to be addressed such as bacterial burden, necrotic tissue, and moisture balance of the whole wound. In acute wounds, there is a precise balance between production and degradation of molecules such as collagen; in chronic wounds this balance is lost and degradation plays too large a role.

<span class="mw-page-title-main">Myofibroblast</span>

A myofibroblast is a cell phenotype that was first described as being in a state between a fibroblast and a smooth muscle cell.

<span class="mw-page-title-main">CYR61</span> Protein-coding gene in the species Homo sapiens

Cysteine-rich angiogenic inducer 61 (CYR61) or CCN family member 1 (CCN1), is a matricellular protein that in humans is encoded by the CYR61 gene.

<span class="mw-page-title-main">Dermatopontin</span> Protein-coding gene in the species Homo sapiens

Dermatopontin also known as tyrosine-rich acidic matrix protein (TRAMP) is a protein that in humans is encoded by the DPT gene. Dermatopontin is a 22-kDa protein of the noncollagenous extracellular matrix (ECM) estimated to comprise 12 mg/kg of wet dermis weight. To date, homologues have been identified in five different mammals and 12 different invertebrates with multiple functions. In vertebrates, the primary function of dermatopontin is a structural component of the ECM, cell adhesion, modulation of TGF-β activity and cellular quiescence). It also has pathological involvement in heart attacks and decreased expression in leiomyoma and fibrosis. In invertebrate, dermatopontin homologue plays a role in hemagglutination, cell-cell aggregation, and expression during parasite infection.

<span class="mw-page-title-main">Desmoplasia</span> Growth of fibrous or connective tissue

In medicine, desmoplasia is the growth of fibrous connective tissue. It is also called a desmoplastic reaction to emphasize that it is secondary to an insult. Desmoplasia may occur around a neoplasm, causing dense fibrosis around the tumor, or scar tissue (adhesions) within the abdomen after abdominal surgery.

Acellular dermis is a type of biomaterial derived from processing human or animal tissues to remove cells and retain portions of the extracellular matrix (ECM). These materials are typically cell-free, distinguishing them from classical allografts and xenografts, can be integrated or incorporated into the body, and have been FDA approved for human use for more than 10 years in a wide range of clinical indications.

The dermal equivalent, also known as dermal replacement or neodermis, is an in vitro model of the dermal layer of skin. There is no specific way of forming a dermal equivalent, however the first dermal equivalent was constructed by seeding dermal fibroblasts into a collagen gel. This gel may then be allowed to contract as a model of wound contraction. This collagen gel contraction assay may be used to screen for treatments which promote or inhibit contraction and thus affect the development of a scar. Other cell types may be incorporated into the dermal equivalent to increase the complexity of the model. For example, keratinocytes may be seeded on the surface to create a skin equivalent, or macrophages may be incorporated to model the inflammatory phase of wound healing.

Dermal fibroblasts are cells within the dermis layer of skin which are responsible for generating connective tissue and allowing the skin to recover from injury. Using organelles, dermal fibroblasts generate and maintain the connective tissue which unites separate cell layers. Furthermore, these dermal fibroblasts produce the protein molecules including laminin and fibronectin which comprise the extracellular matrix. By creating the extracellular matrix between the dermis and epidermis, fibroblasts allow the epithelial cells of the epidermis to affix the matrix, thereby allowing the epidermal cells to effectively join together to form the top layer of the skin.

<span class="mw-page-title-main">Heparan sulfate analogue</span>

Heparan sulfate analogues are polymers engineered to mimic several properties of heparan sulfates. They can be constituted with a backbone of polysaccharides, such as poly glucose or glucuronates or a polyester such as co polymers of lactic or malic acid to which sulfates, sulfonate or carboxyl groups are added in controlled amounts and location. They have a molecular weight that can range from a few thousand to several hundred thousand Dalton. Heparan sulfates can sequester growth factors (GFs) and cytokines in the extracellular matrix (ECM) thereby protecting them from degradation. This ensures local presence of these signaling proteins to fulfill their function in the ECM which contributes to the preservation of anatomical form and function. Heparan sulfates bind to matrix proteins on specific sites called "heparan sulfate binding sites" on ECM macromolecules like collagen, fibronectin and laminin, to form a scaffold surrounding the cells and to protect ECM proteins and growth factors from proteolytic degradation by steric hindrance. However, at any site of inflammation, so also in wound areas, heparan sulfates are degraded, mainly by heparanases giving free access to protease to degrade the ECM and a subsequent loss of GFs and cytokines that disrupts the normal tissue homeostasis. Heparan sulfate analogues obtain many of the characteristics of heparan sulfates including the ability to sequester GFs and bind and protect matrix proteins. However, heparan sulfate analogues are resistant to enzymatic degradation. This way they strengthen the healing potential of the wound bed by repositioning GFs and cytokines back into the ECM.

<span class="mw-page-title-main">Diabetic foot ulcer</span> Medical condition

Diabetic foot ulcer is a breakdown of the skin and sometimes deeper tissues of the foot that leads to sore formation. It may occur due to a variety of mechanisms. It is thought to occur due to abnormal pressure or mechanical stress chronically applied to the foot, usually with concomitant predisposing conditions such as peripheral sensory neuropathy, peripheral motor neuropathy, autonomic neuropathy or peripheral arterial disease. It is a major complication of diabetes mellitus, and it is a type of diabetic foot disease. Secondary complications to the ulcer, such as infection of the skin or subcutaneous tissue, bone infection, gangrene or sepsis are possible, often leading to amputation.

Scar free healing is the process by which significant injuries can heal without permanent damage to the tissue the injury has affected. In most healing, scars form due to the fibrosis and wound contraction, however in scar free healing, tissue is completely regenerated. During the 1990s, published research on the subject increased; it is a relatively recent term in the literature. Scar free healing occurs in foetal life but the ability progressively diminishes into adulthood. In other animals such as amphibians, however, tissue regeneration occurs, for example as skin regeneration in the adult axolotl.

Immune system contribution to regeneration of tissues generally involves specific cellular components, transcription of a wide variety of genes, morphogenesis, epithelia renewal and proliferation of damaged cell types. However, current knowledge reveals more and more studies about immune system influence that cannot be omitted. As the immune system exhibits inhibitory or inflammatory functions during regeneration, the therapies are focused on either stopping these processes or control the immune cells setting in a regenerative way, suggesting that interplay between damaged tissue and immune system response must be well-balanced. Recent studies provide evidence that immune components are required not only after body injury but also in homeostasis or senescent cells replacement.