ArticlePDF Available

Role of angiogenesis and angiogenic factors in acute and chronic wound healing

OAE Publishing Inc.
Plastic and Aesthetic Research
Authors:
  • Srinivas Institute of Medical Sciences and Research Center

Abstract and Figures

Angiogenesis plays a crucial role in wound healing by forming new blood vessels from preexisting vessels by invading the wound clot and organizing in to a microvascular network throughout the granulation tissue. This dynamic process is highly regulated by signals from both serum and the surrounding extracellular matrix (ECM) environment. Vascular endothelial growth factor, angiopoietin, fibroblast growth factor and transforming growth factor beta are amongst the potent angiogenic cytokines in wound angiogenesis. Specific endothelial cell ECM receptors are critical for morphogenetic changes in blood vessels during wound repair. In particular integrin (αvβ3) receptors for fibrin and fibronectin, appear to be required for wound angiogenesis: αvβ3 is focally expressed at the tips of angiogenic capillary sprouts invading the wound clot, and any functional inhibitors of αvβ3 such as monoclonal antibodies, cyclic RGD peptide antagonists and peptidomimetics rapidly inhibit granulation tissue formation. Inspite of clear knowledge about influence of many angiogenic factors on wound healing, little progress has been made in defining the source of these factors, the regulatory events involved in wound angiogenesis and in the clinical use of angiogenic stimulants to promote repair.
This content is subject to copyright. Terms and conditions apply.
Topic: Current Concepts in Wound Healing
© 2015 Plastic and Aesthetic Research | Published by Wolters Kluwer - Medknow 243
INTRODUCTION
Neovascularization or angiogenesis is important for
wound healing as it involves the growth of new capillaries
to form granulation tissue.[1‑4] Three to five days after
tissue injury, new capillaries become visible in the wound
bed as granulation tissue, which acts as a matrix for
proliferating blood vessels, migrating fibroblasts and new
collagen.[5] Impaired granulation is a hallmark of chronic
wounds encountered with diabetes and venous or arterial
insufficiency.
In 1960s, research began in the field of angiogenesis
to determine how new blood vessels enhance solid
tumor growth.[6] Physiologists later discovered that
neovascularization occurs during tissue regeneration.[7]
Proliferating capillaries bring oxygen and micronutrients
to growing tissues and remove catabolic waste products.
These vessels are present in the endothelium that secretes
paracrine factors to promote survival of adjacent cells by
preventing apoptosis or programmed cell death.[8] Because
Role of angiogenesis and angiogenic
factors in acute and chronic wound healing
Thittamaranahalli Muguregowda Honnegowda1, Pramod Kumar1,2,
Echalasara Govindarama Padmanabha Udupa3, Sudesh Kumar4, Udaya Kumar4, Pragna Rao3
1Department of Plastic Surgery and Burns, Kasturba Medical College, Manipal 576104, Karnataka, India.
2Department of Plastic Surgery, King Abdulaziz Specialist Hospital, Sakaka 42421, Al‑Jouf, Saudi Arabia.
3Department of Biochemistry, Kasturba Medical College, Manipal 576104, Karnataka, India.
4Department of Surgery, District Government Hospital, Udupi 576108, Karnataka, India.
Address for correspondence: Dr. Pramod Kumar, Department of Plastic Surgery and Burns, King Abdulaziz Specialist Hospital,
Sakaka 42421, Al‑Jouf, Saudi Arabia. E‑mail: pkumar86@hotmail.com
ABSTRACT
Angiogenesis plays a crucial role in wound healing by forming new blood vessels from preexisting vessels
by invading the wound clot and organizing into a microvascular network throughout the granulation
tissue. This dynamic process is highly regulated by signals from both serum and the surrounding
extracellular matrix environment. Vascular endothelial growth factor, angiopoietin, broblast growth
factor and transforming growth factor‑beta are among the potent angiogenic cytokines in wound
angiogenesis. Specic endothelial cell ECM receptors are critical for morphogenetic changes in blood
vessels during wound repair. In particular integrin (αvβ3) receptors for brin and bronectin, appear to
be required for wound angiogenesis: αvβ3 is focally expressed at the tips of angiogenic capillary sprouts
invading the wound clot, and any functional inhibitors of αvβ3 such as monoclonal antibodies, cyclic
RGD peptide antagonists, and peptidomimetics rapidly inhibit granulation tissue formation. In spite of
clear knowledge about inuence of many angiogenic factors on wound healing, little progress has been
made in dening the source of these factors, the regulatory events involved in wound angiogenesis and
in the clinical use of angiogenic stimulants to promote repair.
Key words:
Angiogenic factors, endothelium, extracellular matrix protein, granulation tissue, wound healing
Access this article online
Quick Response Code:
Website:
www.parjournal.net
DOI:
10.4103/2347-9264.165438
This is an open access article distributed under the terms of the Creative Commons
Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix,
tweak, and build upon the work non-commercially, as long as the author is credited
and the new creations are licensed under the identical terms.
For reprints contact: reprints@medknow.com
How to cite this article: Honnegowda TM, Kumar P, Udupa EG,
Kumar S, Kumar U, Rao P. Role of angiogenesis and angiogenic factors
in acute and chronic wound healing. Plast Aesthet Res 2015;2:243-9.
Received: 20-10-2014; Accepted: 28-01-2015
Plast Aesthet Res || Vol 2 || Issue 5 || Sep 15, 2015244
angiogenesis is required for wound healing, its induction
is beneficial in many clinical situations for achieving
wound closure.
PHYSIOLOGICAL CONTROL OF
ANGIOGENESIS
Angiogenesis plays a critical role in wound healing. By
developing capillary sprouts, which digest endothelial
cells and invade the extracellular matrix (ECM) stroma
after penetrating through the underlying vascular
basement membrane (VBM), and form tube‑like structures
that continue to extend, branch, and form networks.
During angiogenesis capillary advancement in ECM occurs
by endothelial cell proliferation and direction of growth
is guided by chemotaxis from the target region. The
interaction among endothelial cells, angiogenesis factors
and surrounding ECM proteins is temporally and spatially
synchronized.[9,10]
Angiogenesis can be induced in response to injury via
pro‑ and anti‑angiogenic factors present throughout the
body. Pro‑angiogenic factors consist of thrombin, fibrinogen
fragments, thymosin‑β4 and growth factors. Angiogenic
growth factors are stored in platelets and inflammatory
cells that circulate in the bloodstream, and are sequestered
within the ECM. The production of these factors is
regulated by genes expressed in response to hypoxia and
inflammation, such as hypoxia‑inducible factors (HIF) and
cyclooxygenase‑2 (COX‑2).[11‑13] In contrast, angiogenesis
inhibitor factors suppress blood vessel growth.[14,15] Some
inhibitors circulate in the blood stream at low physiological
levels while others are stored in the ECM surrounding
blood vessels. Vascular growth is suppressed when
there is a physiological balance between angiogenesis
stimulators and inhibitors.[15] Immediately following injury,
however, angiogenic stimuli are released into the wound
bed, and a shift occurs in regulators favoring vascular
growth [Figure 1].
THE ANGIOGENESIS CASCADE
Angiogenesis occurs as an orderly cascade of molecular
and cellular events in the wound bed:
1. Endothelial cell surface has receptors to which angiogenic
growth factors bind in preexisting venules (parent vessels);
2. Growth factor‑receptor binding activates signaling
pathways within endothelial cells;
3. Proteolytic enzymes released by activated endothelial cells
dissolve the basement membrane of surrounding parent
vessels;
4. Endothelial cells proliferate and sprout outward through
the basement membrane;
5. Endothelial cells migrate into the wound bed using
integrins (αvβ3, αvβ5 and αvβ1) which are cell surface
adhesion molecules;
6. Matrix metalloproteinases (MMPs) dissolve the surrounding
tissue matrix in the path of sprouting vessels;
7. Vascular sprouts form tubular channels that connect to
form vascular loops;
8. Vascular loops differentiate into afferent (arterial) and
efferent (venous) limbs;
9. New blood vessels mature by recruiting mural cells
(smooth muscle cells and pericytes) to stabilize the
vascular architecture;
10. Blood flow begins in the mature stable vessel.
These complex growth factor‑receptor, cell‑cell and cell‑matrix
interactions characterize the angiogenesis process, regardless
of the stimuli or its location in the body.
THE ANGIOGENESIS MODEL OF
WOUND HEALING
Wound healing occurs in four major overlapping stages:
(1) hemostatic, (2) inflammatory stage, (3) proliferative
stage, and (4) remodeling stage. Although granulation
is assigned to the proliferative stage, angiogenesis is
initiated immediately after tissue injury and is mediated
throughout the wound healing process.
Step 1: Angiogenesis initiation
Basic fibroblast growth factor (bFGF) stored within intact cells
and the ECM is released from damaged tissue.[16] Bleeding and
hemostasis in a wound also initiate angiogenesis. Cellular
receptors for vascular endothelial growth factor (VEGF) are
upregulated by thrombin in the wound.[17] Endothelial cells
exposed to thrombin also release gelatinase A (MMP‑2), which
promotes the local dissolution of basement membrane, a
necessary early step of angiogenesis.[18] Platelets release
multiple growth factors, including platelet‑derived
growth factor (PDGF), VEGF, transforming growth
factor (TGF‑α, TGF‑β), bFGF, platelet‑derived endothelial
cell growth factor and angiopoietin‑1 (Ang‑1). These factors
stimulate endothelial proliferation, migration and tube
formation.[19‑22]
Step 2: Angiogenesis amplication
Macrophages and monocytes release numerous angiogenic
factors, including PDGF, VEGF, Ang‑1, TGFα, bFGF,
interleukin‑8 (IL‑8) and tumor necrosis factor alpha into
the wound bed during the inflammatory phase amplifying
angiogenesis further.[23,24] Several growth factors (PDGF, VEGF
and bFGF) synergize in their ability to vascularize tissues.[25]
Proteases that break down damaged tissue matrix further
release matrix‑bound angiogenic stimulators. Enzymatic
cleavage of fibrin yields fibrin fragment E, which
stimulates angiogenesis directly and also enhances the
Figure 1: Angiogenesis is a balance between stimulators (growth factors)
and inhibitors as shown in this model
Plast Aesthet Res || Vol 2 || Issue 5 || Sep 15, 2015 245
effects of VEGF and bFGF.[26] Expression of the inducible
COX‑2 enzyme during the inflammatory stage of healing
also leads to VEGF production and other promoters of
angiogenesis.[27]
Step 3: Vascular proliferation
Hypoxia is an important driving force for wound
angiogenesis. Expression of gene HIF‑1α, due to hypoxic
gradient between injured and healthy tissue triggers
VEGF production.[24,28] VEGF is present in both wound
tissue and exudate.[28,29] VEGF is also known as vascular
permeability factor since it increases permeability of
capillaries.[30] Hypoxia also leads to endothelial cell
production of nitric oxide (NO). NO promotes vasodilation
and angiogenesis to improve local blood flow.[31]
Step 4: Vascular stabilization
Vascular stabilization is governed by Ang‑1, tyrosine kinase
with immunoglobulin‑like and EGF‑like domains 2 (Tie‑2),
smooth muscle cells and pericytes. Production of PDGF
and recruitment of smooth muscle cells and pericytes to
the newly forming vasculature are regulated by binding
of Ang‑1 to its receptor Tie‑2 on activated endothelial
cells.[32‑34] A PDGF deficiency leads to poorly‑formed
immature blood vessels.[35]
Step 5: Angiogenesis suppression
Angiogenesis is suppressed at the terminal stages of
healing.[36] As tissue hypoxia is restored, and inflammation
subsides, the level of growth factors decline in the wound.
Pericytes which stabilize endothelial cells secrete an
inhibitory form of activated TGF‑β that impedes vascular
proliferation.[34,37,38] A cleavage product of collagen XVIII,
endostatin, is present surrounding the VBM, and it inhibits
wound vascularity.[39,40]
WOUND ANGIOGENIC STIMULATORS
AND INHIBITORS
A number of angiogenic stimulators have been identified
in wound sand others are likely to exist that play an
important role in the repair [Table 1]. The stimulators
in wound fluids are growth factors known to increase
endothelial cell migration and proliferation in vitro.[41]
The FGF comprises of 23 homologous structures that
are small polypeptides with a central core containing
140 amino acids. Acidic FGF and bFGF are the first few
to be discovered and are now designated as FGF‑1 and
FGF‑2, respectively.[42] Both are preferentially involved
in the process of angiogenesis.[43,44] These compounds
are polypeptides of about 18 kDa, single chained and
nonglycosylated. They transmit their signals through
FGF receptor‑4 (FGFR‑4) high‑affinity, protein family of
transmembrane tyrosine kinases (FGFR‑1 to FGFR‑4), that
bind to different FGFs with different affinities. The strong
interactions of FGF‑1 and FGF‑2 with glycosaminoglycans,
such as heparin sulfate present in the ECM,[45] makes the
FGFs stable against thermal, proteolytic denaturation and
limits its diffusibility. Thus, the ECM acts as a reservoir for
pro‑angiogenic factors. Most members of the FGF family
act as a broad spectrum mitogen that stimulates the
proliferation of mesenchymal cells of mesodermal origin,
as well as ectodermal and endodermal cells.
FGF‑1 and FGF‑2 are synthesized by a variety of cell types
including inflammatory cells and dermal fibroblasts that
are involved in angiogenesis and wound healing. When
liberated from ECM, they act on the endothelial cells
in a paracrine manner, or when released by endothelial
cell they act in an autocrine manner promoting cell
proliferation and differentiation. During the formation of
granulation tissue, FGF‑2 promotes cell migration through
surface receptors for integrins, which mediate the binding
of endothelial cells to ECM.[44]
Vascular endothelial growth factor increase vaso‑permeability
by increasing the fenestration and hydraulic conductivity.
This allows leakage of fibrinogen and fibronectin,
which are essential for the formation of the provisional
ECM.[46‑48] The ECM is produced in large quantities by
the epidermis during wound healing.[49] Low oxygen
tension that occurs in tissue hypoxia is a major inducer
of VEGF[50] and its receptors.[51] Thus, cell disruption and
hypoxia appear to be strong initial inducers of potent
angiogenesis factors at the wound site. VEGF family
currently includes VEGF‑A, VEGF‑B, VEGF‑C, VEGF‑D,
VEGF‑E and placental growth factor.[52] VEGF‑A is a
homodimer glycoprotein whose subunits are linked by
2 disulfide bonds. VEGF‑A is synthesized from internal
rearrangements (“alternative splicing”) of mRNA. Thus,
there is the production of 7 isoforms with 121 to 206
amino acids.[53‑55] Among these, the VEGF121, VEGF165,
VEGF189 and VEGF206 are the predominant isoforms.[56]
These isoforms show similar biological activities, but differ
in their binding properties to heparin and ECM.[57]
Vascular endothelial growth factor is a potent vascular
endothelial cell‑specific mitogen that stimulates endothelial
cell proliferation, microvascular permeability and regulates
of several endothelial integrin receptors during sprouting
of new blood vessels.[58] Furthermore, VEGF also acts
as a survival factor for endothelial cells by inducing the
expression of an anti‑apoptotic protein B‑cell lymphoma 2.[59]
TGF‑β stimulates the formation of granulation tissue by
acting as a chemoattractant for neutrophils, macrophages
and fibroblasts. Hence, TGF‑β is an important modulator
of angiogenesis during wound healing by regulating cell
Table 1: Angiogenic stimulators and inhibitors
Stimulators Inhibitors
aFGF (FGF-1) Thrombospondin-1
bFGF (FGF-2) Tissue inhibitors of matrix metalloproteinases
TGF‑α Interferon alpha/beta/gamma
TGF‑β Angiostatin
PGE2 Endostatin
TNF‑α
VEGF
EGF
FGF: Fibroblast growth factor, aFGF: Acidic broblast growth factor,
bFGF: Basic broblast growth factor, TGF‑α: Transforming growth factor‑alpha,
TGF‑β: Transforming growth factor‑beta, VEGF: Vascular endothelial growth
factor, EGF: Endothelial growth factor, PGE2: Prostaglandin E2
Plast Aesthet Res || Vol 2 || Issue 5 || Sep 15, 2015246
proliferation, migration, capillary tube formation and
deposition of ECM.[60,61]
The angiopoietins are members of the VEGF family, which
is largely specific for vascular endothelium. They include a
naturally occurring agonist, Ang‑1, and antagonist, Ang‑2,
both of which act by means of the Tie‑2 receptor. Two
new angiopoietins, Ang‑3 in mice and Ang‑4 in humans,
have been identified, but their function in angiogenesis is
unknown.[62]
Mast cell tryptase, stored in granules of activated mast cells,
is an additional angiogenesis factor that directly degrades
the ECM components or release matrix‑bound growth
factors by its proteolytic activity,[63,64] and acts indirectly
by activating latent matrix metalloproteases. The addition
of tryptase to microvascular endothelial cells cultured on
a basement membrane matrix (matrigel) caused a marked
increase in capillary growth. Furthermore, tryptase can
induce endothelial cell proliferation in a dose‑dependent
manner, whereas specific tryptase inhibitors suppress the
capillary growth.[65]
IMPAIRED ANGIOGENESIS IN CHRONIC
WOUNDS
Angiogenesis is impaired in all chronic wounds leading
to further tissue damage results from chronic hypoxia
and impaired micronutrient delivery. Specific defects have
been identified in diabetic ulcers, venous insufficiency
ulcers and ischemic ulcers.
Diabetic ulcers
Patients with diabetes show abnormal angiogenesis in
various organs. Vasculopathies associated with diabetes
include abnormal blood vessel formation (e.g. retinopathy,
nephropathy) and accelerated atherosclerosis leading
to coronary artery disease, peripheral vascular disease,
and cerebrovascular disease.[65] However, in diabetics,
angiogenesis is decreased[66] resulting in poor formation
of new blood vessels and thus decreased entry of
inflammatory cells and their growth factors. Growth factors
such as FGF‑2 and PDGF, essential for wound healing
have been found to be reduced in experimental diabetic
wounds models.[67‑70] Furthermore, in rat models, topical
administration of high glucose to wounds was shown to
inhibit the normal angiogenic process,[71] suggesting a direct
role for high glucose levels in diminished angiogenesis.
Vascular endothelial growth factor plays an important
role in vascular growth and has been shown to be
deficient in diabetic wounds in experimental and clinical
models.[72] Studies have shown that modulation of
oxidative damage[73] or inhibition of the receptors for
advanced glycation end products[74] improve wound healing
and were associated with the up‑regulation of endogenous
VEGF. Moreover, VEGF administration improves wound
healing in nondiabetic ischemic wounds[75] and blocking
VEGF with neutralizing antibodies impedes tissue repair.[76]
These studies support the notion that VEGF is critical for
repair in impaired healing states and that the addition
of VEGF could have a potential clinical use.[77] In fact,
Galiano et al.[78] found that topical VEGF accelerates wound
healing in a diabetic mouse model.
Weinheimer‑Haus et al.[79] found that low intensity
vibration (LIV) applied vertically at 45 Hz with peak
acceleration of 0.4 g for 30 min a day for 5 days a week
starting on the day of injury in diabetic mice increases
expression of pro‑healing growth factors and chemokines
(insulin‑like growth factor‑1, VEGF and monocyte
chemotactic protein‑1) in wound environment. Though
there was no evidence of a change in the phenotype
of CD11b+ macrophages, however, LIV resulted in
trend toward a less inflammatory phenotype in the
CD11b2 cells which comprised of fibroblasts, endothelial
cells and/or keratinocytes. These findings indicate that
LIV may exert beneficial effects on wound healing by
enhancing angiogenesis and granulation tissue formation,
and these changes are associated with an increase in
pro‑angiogenic growth factors.[79]
Venous insufciency ulcers
Venous insufficiency ulcers or venous stasis ulcers result
from incompetent valves in lower extremity veins, leading
to venous stasis and hypertension that makes the skin
susceptible to ulceration. Pathological findings associated
with venous stasis ulcers include microangiopathy,
fibrin “cuffing” and trapping of leukocytes within the
microvasculature.[80,81]
Chronic venous stasis ulcer patients have elevated levels
of VEGF in their circulation.[82] This may explain the
vascular permeability and increased transudation of serum
fluid in their wounds. Biopsies of these ulcers reveal
microvessels that are surrounded by fibrin cuffs composed
of fibrin and plasma proteins, such as α‑macroglobulin,
thought to compromise gas exchange.[83‑85] Clinical studies
have shown that transcutaneous oxygen tension may be
up to 85% lower in venous stasis ulcers compared with
normal skin regions.[86] VEGF expression is up‑regulated by
hypoxia, which further exacerbates vascular permeability,
formation of pericapillary fibrin cuffs and compromised
gas exchange, which ultimately reduces growth factor
availability in the wound.[87,88] VEGF promotes the
formation tortuous, aberrant glomeruloid‑like vascular
structures found in granulation tissue.[89] Laboratory
animals treated with VEGF form these glomeruloid
vascular structures within 3 days and are characterized
by poor perfusion.[90] In venous ulcers, the persistence of
glomeruloid vessels may interfere with oxygen delivery
and delay healing. In chronic venous stasis ulcers,
high levels of proteases such as neutrophil elastase,
MMPs and urokinase‑type plasminogen activator are
present.[91] Concomitantly, there are decreased levels
of protease inhibitors, such as plasminogen activator
inhibitor‑2. Excessive protease activity may degrade the
growth factors and destroy granulation tissue.
Ischemic ulcers
Peripheral arterial disease (PAD) may result in severe
ischemia.[92] Reduce tissue perfusion due to ischemia
results in progressive tissue hypoxia, ischemia, necrosis
and skin breakdown. In theory, tissue hypoxia should
Plast Aesthet Res || Vol 2 || Issue 5 || Sep 15, 2015 247
initiate angiogenesis via inducing an HIF‑1α and
angiogenic growth factors. In patients with PAD, serum
levels of hepatocyte growth factor are elevated than in
normal subjects.[93] The tissue compromise caused by
severe macrovascular disease, however, may over dominate
the angiogenic response. Inter‑individual differences in the
ability to mount angiogenesis under hypoxic conditions
also exist among patients with atherosclerosis. Such
variations may explain that patients with PAD are unable
to generate adequate collateral circulation and unable to
heal arterial ulcers despite surgical bypass. Therapeutic
growth factors or other methods designed to stimulate
angiogenesis might benefit patients with a defective
angiogenic capacity. VEGF gene transfer[94] or autologous
transplantation of bone marrow‑derived endothelial
progenitor stem cells[95] improved healing of arterial ulcers
in patients.
ANGIOMODULATORY STRATEGIES
Wound angiogenesis represents a realistic model to study
molecular mechanisms involved in the formation and
remodeling of vascular structures. In particular, the repair
of skin defect offers an ideal model to analyze angiogenesis
as it is easy to control and manipulate this process.[96]
Vessel growth is controlled by the local actions of chemical
mediators, the ECM, metabolic gradients and physical
forces. Manipulation of some of these factors is being tried
to improve healing in experimental wounds.[97] Scientists
are working on mathematical models which describe
the role of angiogenesis as observed during (soft tissue)
wound healing. Through this model manipulation of the
capillary tip, macrophage‑derived chemical attractant profile,
extracellular matrix and fibroblast diffusion coefficient may
be analyzed to enhance wound healing.[98]
CONCLUSION
Angiogenesis is a physiological process that is vital for
normal wound healing. A number of factors regulate
wound angiogenesis, including hypoxia, inflammation
and growth factors. The molecular and cellular events in
angiogenesis have been elucidated, and defects in this
process are present in chronic wounds. Based on this
knowledge, new wound healing strategies are emerging
to deliver growth factors to the wound bed. Surgeons
and other wound‑care specialists can use this knowledge
to identify defects and select interventions that may
promote improved wound granulation and healing.
Financial support and sponsorship
Nil.
Conicts of interest
There are no conflicts of interest.
REFERENCES
1. Li WW, Li VW, Tsakayannis D. Angiogenesis therapies. Concepts, clinical
trials, and considerations for new drug development. In: Fan TPD, Kohn EC,
editors. The New Angiotherapy. Totowa: Humana Press; 2002. p. 547‑71.
2. Folkman J. Seminars in medicine of the Beth Israel hospital, Boston. Clinical
applications of research on angiogenesis. N Engl J Med 1995;333:1757‑63.
3. Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J. Free radicals
and antioxidants in normal physiological functions and human disease.
Int J Biochem Cell Biol 2007;39:44‑84.
4. Rees M, Hague S, Oehler MK, Bicknell R. Regulation of endometrial
angiogenesis. Climacteric 1999;2:52‑8.
5. Tonnesen MG, Feng X, Clark RA. Angiogenesis in wound healing. J Investig
Dermatol Symp Proc 2000;5:40‑6.
6. Shah F, Balan P, Weinberg M, Reddy V, Neems R, Feinstein M, Dainauskas J,
Meyer P, Goldin M, Feinstein SB. Contrast‑enhanced ultrasound imaging of
atherosclerotic carotid plaque neovascularization: a new surrogate marker
of atherosclerosis? Vasc Med 2007;12:291‑7.
7. Folkman J. Angiogenesis. In: Braunwald E, Fauci AS, Kasper DL, Hauser SL,
Longo DL, Jameson LJ, editors. Harrison’s Textbook of Internal Medicine.
15th ed. New York: McGraw‑Hill; 2001. p. 517‑30.
8. O’Connor DS, Schechner JS, Adida C, Mesri M, Rothermel AL, Li F, Nath AK,
Pober JS, Altieri DC. Control of apoptosis during angiogenesis by survivin
expression in endothelial cells. Am J Pathol 2000;156:393‑8.
9. Clark RA. Wound repair. Overview and general considerations. In: Clark RAF,
editor. The Molecular and Cellular Biology of Wound Repair. New York:
Plenum; 1996. p. 3‑50.
10. Morgan MR, Humphries MJ, Bass MD. Synergistic control of cell adhesion
by integrins and syndecans. Nat Rev Mol Cell Biol 2007;8:957‑69.
11. Semenza G. Signal transduction to hypoxia‑inducible factor 1. Biochem
Pharmacol 2002;64:993‑8.
12. Majima M, Hayashi I, Muramatsu M, Katada J, Yamashina S, Katori M.
Cyclo‑oxygenase‑2 enhances basic fibroblast growth factor‑induced
angiogenesis through induction of vascular endothelial growth factor in rat
sponge implants. Br J Pharmacol 2000;130:641‑9.
13. Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF
system. Nat Med 2003;9:677‑84.
14. Miles KA. Perfusion CT for the assessment of tumour vascularity: which
protocol? Br J Radiol 2003;76:S36‑42.
15. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell
2011;144:646‑74.
16. Matsuoka H, Sisson TH, Nishiuma T, Simon RH. Plasminogen‑mediated
activation and release of hepatocyte growth factor from extracellular matrix.
Am J Respir Cell Mol Biol 2006;35:705‑13.
17. Tsopanoglou NE, Maragoudakis ME. On the mechanism of thrombin‑induced
angiogenesis. Potentiation of vascular endothelial growth factor activity
on endothelial cells by up‑regulation of its receptors. J Biol Chem
1999;274:23969‑76.
18. Nguyen M, Arkell J, Jackson CJ. Human endothelial gelatinases and
angiogenesis. Int J Biochem Cell Biol 2001;33:960‑70.
19. Hellberg C, Ostman A, Heldin CH. PDGF and vessel maturation. Recent
Results Cancer Res 2010;180:103‑14.
20. Pintucci G, Froum S, Pinnell J, Mignatti P, Rai S, Green D. Trophic effects
of platelets on cultured endothelial cells are mediated by platelet‑associated
fibroblast growth factor‑2 (FGF‑2) and vascular endothelial growth
factor (VEGF). Thromb Haemost 2002;88:834‑42.
21. Li JJ, Huang YQ, Basch R, Karpatkin S. Thrombin induces the release of
angiopoietin‑1 from platelets. Thromb Haemost 2001;85:204‑6.
22. Nath SG, Raveendran R. An insight into the possibilities of broblast growth
factor in periodontal regeneration. J Indian Soc Periodontol 2014;18:289‑92.
23. Yoshida S, Yoshida A, Matsui H, Takada Y, Ishibashi T. Involvement of
macrophage chemotactic protein‑1 and interleukin‑1beta during inammatory
but not basic broblast growth factor‑dependent neovascularization in the
mouse cornea. Lab Invest 2003;83:927‑38.
24. Grimm D, Bauer J, Schoenberger J. Blockade of neoangiogenesis, a new and
promising technique to control the growth of malignant tumors and their
metastases. Curr Vasc Pharmacol 2009;7:347‑57.
25. Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular
microenvironments for morphogenesis in tissue engineering. Nat Biotechnol
2005;23:47‑55.
26. Bootle‑Wilbraham CA, Tazzyman S, Thompson WD, Stirk CM,
Lewis CE. Fibrin fragment E stimulates the proliferation, migration and
differentiation of human microvascular endothelial cells in vitro. Angiogenesis
2001;4:269‑75.
27. Ji K, Tsirka SE. Inammation modulates expression of laminin in the central
nervous system following ischemic injury. J Neuroinammation 2012;9:159.
28. Acker T, Plate KH. Role of hypoxia in tumor angiogenesis‑molecular and
cellular angiogenic crosstalk. Cell Tissue Res 2003;314:145‑55.
29. Howdieshell TR, Webb WL, Sathyanarayana, McNeil PL. Inhibition of
Plast Aesthet Res || Vol 2 || Issue 5 || Sep 15, 2015248
inducible nitric oxide synthase results in reductions in wound vascular
endothelial growth factor expression, granulation tissue formation, and local
perfusion. Surgery 2003;133:528‑37.
30. Leonardi R, Caltabiano M, Pagano M, Pezzuto V, Loreto C, Palestro G.
Detection of vascular endothelial growth factor/vascular permeability factor
in periapical lesions. J Endod 2003;29:180‑3.
31. Smith RS Jr, Gao L, Bledsoe G, Chao L, Chao J. Intermedin is a new angiogenic
growth factor. Am J Physiol Heart Circ Physiol 2009;297:H1040‑7.
32. Inoki I, Shiomi T, Hashimoto G, Enomoto H, Nakamura H, Makino K,
Ikeda E, Takata S, Kobayashi K, Okada Y. Connective tissue growth factor
binds vascular endothelial growth factor (VEGF) and inhibits VEGF‑induced
angiogenesis. FASEB J 2002;16:219‑21.
33. Ma J, Wang Q, Fei T, Han JD, Chen YG. MCP‑1 mediates TGF‑beta‑induced
angiogenesis by stimulating vascular smooth muscle cell migration. Blood
2007;109:987‑94.
34. Korff T, Kimmina S, Martiny‑Baron G, Augustin HG. Blood vessel maturation
in a 3‑dimensional spheroidal coculture model: direct contact with smooth
muscle cells regulates endothelial cell quiescence and abrogates VEGF
responsiveness. FASEB J 2001;15:447‑57.
35. Onimaru M, Yonemitsu Y, Fujii T, Tanii M, Nakano T, Nakagawa K, Kohno R,
Hasegawa M, Nishikawa S, Sueishi K. VEGF‑C regulates lymphangiogenesis
and capillary stability by regulation of PDGF‑B. Am J Physiol Heart Circ Physiol
2009;297:H1685‑96.
36. Kumar I, Staton CA, Cross SS, Reed MW, Brown NJ. Angiogenesis, vascular
endothelial growth factor and its receptors in human surgical wounds. Br J Surg
2009;96:1484‑91.
37. Darland DC, D’Amore PA. TGF beta is required for the formation of
capillary‑like structures in three‑dimensional cocultures of 10T1/2 and
endothelial cells. Angiogenesis 2001;4:11‑20.
38. McCarty MF, Bielenberg DR, Nilsson MB, Gershenwald JE, Barnhill RL,
Ahearne P, Bucana CD, Fidler IJ. Epidermal hyperplasia overlying human
melanoma correlates with tumour depth and angiogenesis. Melanoma Res
2003;13:379‑87.
39. Michaels J 5th, Dobryansky M, Galiano RD, Bhatt KA, Ashinoff R, Ceradini DJ,
Gurtner GC. Topical vascular endothelial growth factor reverses delayed
wound healing secondary to angiogenesis inhibitor administration. Wound
Repair Regen 2005;13:506‑12.
40. Lange‑Asschenfeldt B, Velasco P, Streit M, Hawighorst T, Pike SE, Tosato G,
Detmar M. The angiogenesis inhibitor vasostatin does not impair wound
healing at tumor‑inhibiting doses. J Invest Dermatol 2001;117:1036‑41.
41. Van der Bilt JD, Borel Rinkes IH. Surgery and angiogenesis. Biochim Biophys Acta
2004;1654:95‑104.
42. Hiromatsu Y, Toda S. Mast cells and angiogenesis. Microsc Res Tech
2003;60:64‑9.
43. Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol
2001;2:REVIEWS3005.
44. Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic‑Canic M. Growth
factors and cytokines in wound healing. Wound Repair Regen 2008;16:585‑601.
45. Plum SM, Vu HA, Mercer B, Fogler WE, Fortier AH. Generation of a
specic immunological response to FGF‑2 does not affect wound healing or
reproduction. Immunopharmacol Immunotoxicol 2004;26:29‑41.
46. Nagy JA, Benjamin L, Zeng H, Dvorak AM, Dvorak HF. Vascular
permeability, vascular hyperpermeability and angiogenesis. Angiogenesis
2008;11:109‑19.
47. Breier G, Blum S, Peli J, Groot M, Wild C, Risau W, Reichmann E.
Transforming growth factor‑beta and Ras regulate the VEGF/VEGF‑receptor
system during tumor angiogenesis. Int J Cancer 2002;97:142‑8.
48. Bates DO, Heald RI, Curry FE, Williams B. Vascular endothelial growth factor
increases Rana vascular permeability and compliance by different signalling
pathways. J Physiol 2001;533:263‑72.
49. Failla CM, Odorisio T, Cianfarani F, Schietroma C, Puddu P, Zambruno G.
Placenta growth factor is induced in human keratinocytes during wound
healing. J Invest Dermatol 2000;115:388‑95.
50. Hemmerlein B, Kugler A, Ozisik R, Ringert RH, Radzun HJ, Thelen P. Vascular
endothelial growth factor expression, angiogenesis, and necrosis in renal cell
carcinomas. Virchows Arch 2001;439:645‑52.
51. Zachary I, Gliki G. Signaling transduction mechanisms mediating biological
actions of the vascular endothelial growth factor family. Cardiovasc Res
2001;49:568‑81.
52. Efron PA, Moldawer LL. Cytokines and wound healing: the role of cytokine
and anticytokine therapy in the repair response. J Burn Care Rehabil
2004;25:149‑60.
53. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors.
Nat Med 2003;9:669‑76.
54. Bates DO, Harper SJ. Regulation of vascular permeability by vascular
endothelial growth factors. Vascul Pharmacol 2002;39:225‑37.
55. Ferrara N. Vascular endothelial growth factor: basic science and clinical
progress. Endocr Rev 2004;25:581‑611.
56. Kessler T, Fehrmann F, Bieker R, Berdel WE, Mesters RM. Vascular
endothelial growth factor and its receptor as drug targets in hematological
malignancies. Curr Drug Targets 2007;8:257‑68.
57. Roth D, Piekarek M, Paulsson M, Christ H, Bloch W, Krieg T, Davidson JM,
Eming SA. Plasmin modulates vascular endothelial growth factor‑A‑mediated
angiogenesis during wound repair. Am J Pathol 2006;168:670‑84.
58. Primo L, Seano G, Roca C, Maione F, Gagliardi PA, Sessa R, Martinelli M,
Giraudo E, di Blasio L, Bussolino F. Increased expression of alpha6 integrin
in endothelial cells unveils a proangiogenic role for basement membrane.
Cancer Res 2010;70:5759‑69.
59. Rao X, Zhong J, Zhang S, Zhang Y, Yu Q, Yang P, Wang MH, Fulton DJ, Shi H,
Dong Z, Wang D, Wang CY. Loss of methyl‑CpG‑binding domain protein
2 enhances endothelial angiogenesis and protects mice against hind‑limb
ischemic injury. Circulation 2011;123:2964‑74.
60. Brunner G, Blakytny R. Extracellular regulation of TGF‑beta activity in
wound repair: growth factor latency as a sensor mechanism for injury.
Thromb Haemost 2004;92:253‑61.
61. Verrecchia F, Mauviel A. Transforming growth factor‑beta and brosis. World
J Gastroenterol 2007;13:3056‑62.
62. Tsigkos S, Koutsilieris M, Papapetropoulos A. Angiopoietins in angiogenesis
and beyond. Expert Opin Investig Drugs 2003;12:933‑41.
63. Solovyan VT, Keski‑Oja J. Apoptosis of human endothelial cells is
accompanied by proteolytic processing of latent TGF‑beta binding proteins
and activation of TGF‑beta. Cell Death Differ 2005;12:815‑26.
64. Iddamalgoda A, Le QT, Ito K, Tanaka K, Kojima H, Kido H. Mast cell
tryptase and photoaging: possible involvement in the degradation of extra
cellular matrix and basement membrane proteins. Arch Dermatol Res
2008;300 Suppl 1:S69‑76.
65. Martin A, Komada MR, Sane DC. Abnormal angiogenesis in diabetes mellitus.
Med Res Rev 2003;23:117‑45.
66. Brem H, Jacobs T, Vileikyte L, Weinberger S, Gibber M, Gill K, Tarnovskaya A,
Entero H, Boulton AJ. Wound‑healing protocols for diabetic foot and
pressure ulcers. Surg Technol Int 2003;11:85‑92.
67. Keswani SG, Katz AB, Lim FY, Zoltick P, Radu A, Alaee D, Herlyn M,
Crombleholme TM. Adenoviral mediated gene transfer of PDGF‑B enhances
wound healing in type I and type II diabetic wounds. Wound Repair Regen
2004;12:497‑504.
68. Altavilla D, Saitta A, Cucinotta D, Galeano M, Deodato B, Colonna M,
Torre V, Russo G, Sardella A, Urna G, Campo GM, Cavallari V,
Squadrito G, Squadrito F. Inhibition of lipid peroxidation restores
impaired vascular endothelial growth factor expression and stimulates
wound healing and angiogenesis in the genetically diabetic mouse. Diabetes
2001;50:667‑74.
69. Wicke C, Halliday B, Allen D, Roche NS, Scheuenstuhl H, Spencer MM,
Roberts AB, Hunt TK. Effects of steroids and retinoids on wound healing.
Arch Surg 2000;135:1265‑70.
70. Peplow PV, Baxter GD. Gene expression and release of growth factors during
delayed wound healing: a review of studies in diabetic animals and possible
combined laser phototherapy and growth factor treatment to enhance
healing. Photomed Laser Surg 2012;30:617‑36.
71. Stavrou D. Neovascularisation in wound healing. J Wound Care
2008;17:298‑300, 2.
72. Johnson KE, Wilgus TA. Vascular endothelial growth factor and angiogenesis
in the regulation of cutaneous wound repair. Adv Wound Care (New Rochelle)
2014;3:647‑61.
73. Hoffman M, Monroe DM. Wound healing in haemophilia‑breaking the vicious
cycle. Haemophilia 2010;16 Suppl 3:13‑8.
74. Goova MT, Li J, Kislinger T, Qu W, Lu Y, Bucciarelli LG, Nowygrod S,
Wolf BM, Caliste X, Yan SF, Stern DM, Schmidt AM. Blockade of receptor
for advanced glycation end‑products restores effective wound healing in
diabetic mice. Am J Pathol 2001;159:513‑25.
75. Corral CJ, Siddiqui A, Wu L, Farrell CL, Lyons D, Mustoe TA. Vascular
endothelial growth factor is more important than basic broblastic growth
factor during ischemic wound healing. Arch Surg 1999;134:200‑5.
76. Howdieshell TR, Callaway D, Webb WL, Gaines MD, Procter CD Jr,
Sathyanarayana, Pollock JS, Brock TL, McNeil PL. Antibody neutralization
of vascular endothelial growth factor inhibits wound granulation tissue
formation. J Surg Res 2001;96:173‑82.
77. Lerman OZ, Galiano RD, Armour M, Levine JP, Gurtner GC. Cellular
dysfunction in the diabetic broblast: impairment in migration, vascular
Plast Aesthet Res || Vol 2 || Issue 5 || Sep 15, 2015 249
endothelial growth factor production, and response to hypoxia. Am J Pathol
2003;162:303‑12.
78. Galiano RD, Tepper OM, Pelo CR, Bhatt KA, Callaghan M, Bastidas N,
Bunting S, Steinmetz HG, Gurtner GC. Topical vascular endothelial growth
factor accelerates diabetic wound healing through increased angiogenesis
and by mobilizing and recruiting bone marrow‑derived cells. Am J Pathol
2004;164:1935‑47.
79. Weinheimer‑Haus EM, Judex S, Ennis WJ, Koh TJ. Low‑intensity vibration
improves angiogenesis and wound healing in diabetic mice. PLoS One
2014;9:e91355.
80. Franzeck UK, Haselbach P, Speiser D, Bollinger A. Microangiopathy of
cutaneous blood and lymphatic capillaries in chronic venous insufciency (CVI).
Yale J Biol Med 1993;66:37‑46.
81. Jünger M, Steins A, Hahn M, Häfner HM. Microcirculatory dysfunction in
chronic venous insufciency (CVI). Microcirculation 2000;7:S3‑12.
82. Shoab SS, Scurr JH, Coleridge‑Smith PD. Plasma VEGF as a marker of therapy
in patients with chronic venous disease treated with oral micronised avonoid
fraction‑a pilot study. Eur J Vasc Endovasc Surg 1999;18:334‑8.
83. Oahues N, Philips TJ. Leg ulcers. Curr Probl Dermatol 1995;7:109‑42.
84. Moosa HH, Falanga V, Steed DL, Makaroun MS, Peitzman AB, Eaglstein WH,
Webster MW. Oxygen diffusion in chronic venous ulceration. J Cardiovasc
Surg (Torino) 1987;28:464‑7.
85. Falanga V, Moosa HH, Nemeth AJ, Alstadt SP, Eaglstein WH. Dermal
pericapillary brin in venous disease and venous ulceration. Arch Dermatol
1987;123:620‑3.
86. Weckroth M, Vaheri A, Virolainen S, Saarialho‑Kere U, Jahkola T,
Sirén V. Epithelial tissue‑type plasminogen activator expression, unlike that
of urokinase, its receptor, and plasminogen activator inhibitor‑1, is increased
in chronic venous ulcers. Br J Dermatol 2004;151:1189‑96.
87. Higley HR, Ksander GA, Gerhardt CO, Falanga V. Extravasation of
macromolecules and possible trapping of transforming growth factor‑beta
in venous ulceration. Br J Dermatol 1995;132:79‑85.
88. Trent JT, Falabella A, Eaglstein WH, Kirsner RS. Venous ulcers: pathophysiology
and treatment options. Ostomy Wound Manage 2005;51:38‑54.
89. Lyseng‑Williamson KA, Perry CM. Micronised puried avonoid fraction:
a review of its use in chronic venous insufciency, venous ulcers and
haemorrhoids. Drugs 2003;63:71‑100.
90. Sundberg C, Nagy JA, Brown LF, Feng D, Eckelhoefer IA, Manseau EJ,
Dvorak AM, Dvorak HF. Glomeruloid microvascular proliferation follows
adenoviral vascular permeability factor/vascular endothelial growth factor‑164
gene delivery. Am J Pathol 2001;158:1145‑60.
91. McCarty SM, Cochrane CA, Clegg PD, Percival SL. The role of endogenous
and exogenous enzymes in chronic wounds: a focus on the implications
of aberrant levels of both host and bacterial proteases in wound healing.
Wound Repair Regen 2012;20:125‑36.
92. Weitz JI, Byrne J, Clagett GP, Farkouh ME, Porter JM, Sackett DL,
Strandness DE Jr, Taylor LM. Diagnosis and treatment of chronic arterial
insufficiency of the lower extremities: a critical review. Circulation
1996;94:3026‑49.
93. Konya H, Miuchi M, Satani K, Matsutani S, Tsunoda T, Yano Y, Katsuno T,
Hamaguchi T, Miyagawa J, Namba M. Hepatocyte growth factor, a
biomarker of macroangiopathy in diabetes mellitus. World J Diabetes
2014;5:678‑88.
94. Baumgartner I, Pieczek A, Manor O, Blair R, Kearney M, Walsh K, Isner JM.
Constitutive expression of phVEGF165 after intramuscular gene transfer
promotes collateral vessel development in patients with critical limb ischemia.
Circulation 1998;97:1114‑23.
95. Lawall H, Bramlage P, Amann B. Stem cell and progenitor cell therapy
in peripheral artery disease. A critical appraisal. Thromb Haemost
2010;103:696‑709.
96. Eming SA, Brachvogel B, Odorisio T, Koch M. Regulation of angiogenesis:
wound healing as a model. Prog Histochem Cytochem 2007;42:115‑70.
97. Bauer SM, Bauer RJ, Velazquez OC. Angiogenesis, vasculogenesis,
and induction of healing in chronic wounds. Vasc Endovascular Surg
2005;39:293‑306.
98. Chaplain M, Anderson A. Mathematical modelling of tumour‑induced
angiogenesis: network growth and structure. Cancer Treat Res
2004;117:51‑75.
... TGF-β aligns various cellular events, promoting cell proliferation, differentiation, and extracellular matrix synthesis [10] . VEGF, a key angiogenic factor, stimulates the formation of new blood vessels crucial for nutrient supply to healing tissues [11] . GSK-3β influences diverse cellular pathways, regulating inflammation and cell survival 12 , while MMP-9 facilitates tissue remodeling by degrading extracellular matrix components [13] . ...
... Vascular endothelial growth factor plays a characteristic role is the promotion of angiogenesis. Angiogenesis is significantly elevated during the proliferative phase of wound healing primarily by VEGF 11 . The binding energies of EOCs were highest with VEGF receptor indicating its least affinity towards that receptor. ...
Article
Full-text available
This research explores the multifaceted pharmacological actions of essential oils and its constituents, derived as secondary metabolites from aromatic plants, with a particular focus on their potent wound healing and antibacterial activities, elucidating their significance in therapeutic approach towards infected wounds. An in silico screening was carried out to identify the interaction between the bioactive essential oil contituents (EOC) such as cinnamaldehyde, citral, geraniol, linalool, and p-cymene, docked against various target proteins associated with antibiotic resistance and wound healing, including mec A (PDB ID- 4DK1), nor A (PDB ID- 7LO8), TGF- β1 (PDB ID- 1PY5), TGF- β2 (PDB ID- 1M9Z), VEGF (PDB ID-3QTK), GSK-3β (PDB ID-1Q5K) and MMP-9 (PDB ID-5UE4). The docking was done with AutoDock V 4.0 using five EOCs against seven receptors and the binding energy was gaged. The binding energy of EOCs were observed to be ranging from -5.3 kcal/mol to -2.55 kcal/mol. Notably, all the screened EOCs exhibited favourable binding affinity with GSK-3β, indicating their potential role in the inflammatory phase of wound healing. Additionally, towards antibiotic resistance, all EOC displayed adequate binding affinity with norA, suggesting their potential in modulating multidrug resistant efflux pumps. Compliance with Lipinski's rule, positions these EOC as promising candidates for drug development, particularly in the context of wound healing and antibiotic resistance. This study holds the promise of contributing novel insights to the field of wound care and combating antibiotic resistance, paving the way for innovative approaches in addressing the challenges posed by multi-drug resistant Staphylococcus aureus (MDRSA) infected wounds.
... These cells possess the ability to move toward the site of injury and actively participate in the process of wound healing by undergoing transdifferentiation into various skin cell types. 11,12 Locally transplanted MSCs release paracrine factors (angiogenic and remodeling cytokines), thereby facilitating the wound healing process. [12][13][14] Paracrine secretions serve as the primary mechanism through which hUCMSCs improve the wound microenvironment. ...
... 11,12 Locally transplanted MSCs release paracrine factors (angiogenic and remodeling cytokines), thereby facilitating the wound healing process. [12][13][14] Paracrine secretions serve as the primary mechanism through which hUCMSCs improve the wound microenvironment. These paracrine factors are mainly involved in the recruitment of key regulatory cells (fibroblasts, keratinocytes, macrophages, endothelial cells) which are crucial for wound regeneration. ...
Article
Temporal phases of wound healing and their corresponding healing factors are essential in wound regeneration. Mesenchymal stem cells (MSCs) accelerate wound healing via their paracrine secretions by enhancing cell migration, angiogenesis, and reducing inflammation. This study evaluated the local therapeutic effect of human umbilical cord MSCs (hUCMSCs) in the healing of cold‐induced burn wounds. An in vitro wound (scratch) was developed in rat skin fibroblasts. The culture was maintained in the conditioned medium (CM) which was prepared by inducing an artificial wound in hUCMSCs in a separate experiment. Treated fibroblasts were analyzed for the gene expression profile of healing mediators involved in wound closure. Findings revealed enhanced cell migration and increased levels of healing mediators in the treated fibroblasts relative to the untreated group. Cold‐induced burn wounds were developed in Wistar rats, followed by a single injection of hUCMSCs. Wound healing pattern was examined based on the healing phases: hemostasis/inflammation (Days 1, 3), cell proliferation (Day 7), and remodeling (Day 14). Findings exhibited enhanced wound closure in the treated wound. Gene expression, histological, and immunohistochemical analyses further confirmed enhanced wound regeneration after hUCMSC transplantation. Temporal gene expression profile revealed that the level of corresponding cytokines was substantially increased in the treated wound as compared with the control, indicating improvement in the processes of angiogenesis and remodeling, and a substantial reduction in inflammation. Histology revealed significant collagen formation along with regenerated skin layers and appendages, whereas immunohistochemistry exhibited increased neovascularization during remodeling. Leukocyte infiltration was also suppressed in the treated group. Overall findings demonstrate that a single dose of hUCMSCs enhances wound healing in vivo, and their secreted growth factors accelerate cell migration in vitro.
... The reduction of tissue hypoxia and inflammation results in a decrease in the quantity of growth factors present in the wound. 30 The findings of Johnson and Wilgus 15 , which showed that VEGF protein levels in injured tissue begin to rise one day after injury relative to control intact skin, may also explain this conclusion. VEGF levels are much higher in injured skin three and five days later than in control skin. ...
... They all play various roles in angiogenesis: induction, initiation, cell multiplication, migration, cell stability, wound healing, inflammatory response, and inhibiting angiogenesis. 9,10 In clinical dentistry, endodontic therapies like apexification and apexogenesis, healing of the extraction socket, and tissue regeneration during the post-surgical phase for implant placement cannot be successful without sufficient tissue vascularization. 11 Revascularization of irradiated tissues is also necessary, as it increases the fibroblastic cellular density and reduces the amount of nonviable tissue that needs to be surgically removed. ...
Article
Background: Ozone has become more common in medicine across the globe as an adjuvant treatment method for a variety of illnesses. The combination of ozone and treated human dental pulp stem cell-conditioned media hDPSCs-CM can enhance angiogenesis due to their synergistic action, resulting in increased growth factor expression. Objectives: The aim of the study was to evaluate the angiogenic potential of ozone-treated human dental pulp stem cell-conditioned media (hDPSCs-CM). Materials and Method: HDPSCs were isolated from the extracted tooth. Passaged four cells were characterized with flow cytometry and then exposed to 10μg/mL gaseous Ozone concertation. The conditioned media (CM) were obtained from the treated cells, and growth factor analysis was performed. The functionality of the Ozonated hDPSCs-CM was assessed by the Chick Yolk Sac Membrane (YSM) assay. Results: Ozonated hDPSCs-CM had significantly higher (p<0.01) expression of angiopoietin-2 (Ang-2), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), macrophage colony stimulating factor (M-CSF), and vascular endothelial growth factor (VEGF). The in-ovo YSM assay revealed a notably greater pro-angiogenic potential associated with Ozonated hDPSC-CM
... Angiogenesis followed by blood vessel formation is one of the most important activities that determine the healing of diabetic ulcers [51,52]. Based on the unique angiogenic effects of GExos in both normal and hyperglycemia conditions, the influence of GExos in diabetic wound healing was further evaluated. ...
Article
Full-text available
Reversal of endothelial cell (EC) dysfunction under high-glucose (HG) conditions to achieve angiogenesis has remained a big challenge in diabetic ulcers. Herein, exosomes derived from medicinal plant ginseng (GExos) were shown as excellent nanotherapeutics with biomimetic cell membrane-like structures to be able to efficiently transfer the encapsulated active substances to ECs, resulting in a marked reprogramming of glycolysis by up-regulating anaerobic glycolysis and down-regulating oxidative stress, which further restore the proliferation, migration, and tubule formation abilities of ECs under HG conditions. In vivo, GExos enhance the angiogenesis and nascent vessel network reconstruction in full-thickness diabetic complicated skin ulcer wounds in mice with high biosafety. GExos were shown as promising nanotherapeutics in stimulating glycolysis reprogramming-mediated angiogenesis in diabetic ulcers, possessing wide application potential for reversing hyperglycemic dysangiogenesis and stimulating vascular regeneration.
... Clinically, new capillaries first become visible in the wound bed 3 -5 days after injury, and their appearance is synonymous with granulation tissue, which acts as a matrix for proliferating blood vessels, migrating fibroblasts and new collagen. Impaired granulation is a hallmark of chronic wounds, as encountered in patients with type 2diabetes and venous or arterial insufficiency [1]. A number of researchers have reported, chronic nonhealing wounds are impacted by insufficient angiogenesis; decreased vascularity and capillary density delay wound closure. ...
Article
Full-text available
-Abstract is not required - Keywords: Wound healing, Angiogenesis, Regulating Factors, Nurses, Perspectives Received: 08 January 2019; Reviewed: 19 January 2019; Received: in revised from 25 February 2019; Accepted: 28 February 2019 DOI: 10.35898/ghmj-31548
Chapter
Diabetes is a chronic metabolic disorder that affects approximately 10% of the global population. Unfortunately, diabetic individuals are also at a high risk of developing diabetic foot ulcers (DFUs), with an estimated 19–34% of individuals affected at some point in their lives. These DFUs are a leading cause of lower extremity amputations (LEAs), responsible for 60–70% of all cases, and have a 5-year mortality rate of approximately 49%. The mortality rate associated with DFUs is even more alarming when compared to that of all cancers, which is approximately 31%. The economic burden of DFUs is also staggering, with a global estimated cost of $78.2 billion USD. The incidence of DFUs is further compounded by the fact that 50–60% of cases develop infections, increasing the risk of amputation by 50% compared to patients with uninfected DFUs. This chapter aims to provide a comprehensive overview of infections in diabetes, with a particular focus on infections in DFUs. We will explore the microbiome shift toward pathogenic bacteria in DFU, and how this shift impacts healing outcomes. Additionally, we will examine various factors that make diabetic patients prone to infections, including dysregulations and dysfunctions in the innate immune system. Finally, we will review the conventional, unconventional, and emerging therapeutic options available to address infections in DFUs. By providing an in-depth understanding of the challenges associated with infections in diabetes, this chapter aims to contribute to the development of more effective treatment strategies that can help reduce the burden of DFUs on individuals and society as a whole.
Article
BACKGROUND Vein graft failure following cardiovascular bypass surgery results in significant patient morbidity and cost to the health care system. Vein graft injury can occur during autogenous vein harvest and preparation, as well as after implantation into the arterial system, leading to the development of intimal hyperplasia, vein graft stenosis, and, ultimately, bypass graft failure. Although previous studies have identified maladaptive pathways that occur shortly after implantation, the specific signaling pathways that occur during vein graft preparation are not well defined and may result in a cumulative impact on vein graft failure. We, therefore, aimed to elucidate the response of the vein conduit wall during harvest and following implantation, probing the key maladaptive pathways driving graft failure with the overarching goal of identifying therapeutic targets for biologic intervention to minimize these natural responses to surgical vein graft injury. METHODS Employing a novel approach to investigating vascular pathologies, we harnessed both single-nuclei RNA-sequencing and spatial transcriptomics analyses to profile the genomic effects of vein grafts after harvest and distension, then compared these findings to vein grafts obtained 24 hours after carotid-cartoid vein bypass implantation in a canine model (n=4). RESULTS Spatial transcriptomic analysis of canine cephalic vein after initial conduit harvest and distention revealed significant enrichment of pathways ( P <0.05) involved in the activation of endothelial cells (ECs), fibroblasts, and vascular smooth muscle cells, namely pathways responsible for cellular proliferation and migration and platelet activation across the intimal and medial layers, cytokine signaling within the adventitial layer, and ECM (extracellular matrix) remodeling throughout the vein wall. Subsequent single-nuclei RNA-sequencing analysis supported these findings and further unveiled distinct EC and fibroblast subpopulations with significant upregulation ( P <0.05) of markers related to endothelial injury response and cellular activation of ECs, FBs, and vascular smooth muscle cells. Similarly, in vein grafts obtained 24 hours after arterial bypass, there was an increase in myeloid cell, protomyofibroblast, injury-response EC, and mesenchymal-transitioning EC subpopulations with a concomitant decrease in homeostatic ECs and fibroblasts. Among these markers were genes previously implicated in vein graft injury, including VCAN , FBN1 , and VEGFC , in addition to novel genes of interest such as GLIS3 and EPHA3 . These genes were further noted to be driving the expression of genes implicated in vascular remodeling and graft failure, such as IL-6 , TGFBR1 , SMAD4 , and ADAMTS9. By integrating the spatial transcriptomics and single-nuclei RNA-sequencing data sets, we highlighted the spatial architecture of the vein graft following distension, wherein activated and mesenchymal-transitioning ECs, myeloid cells, and fibroblasts were notably enriched in the intima and media of distended veins. Finally, intercellular communication network analysis unveiled the critical roles of activated ECs, mesenchymal-transitioning ECs, protomyofibroblasts, and vascular smooth muscle cells in upregulating signaling pathways associated with cellular proliferation (MDK, PDGF [platelet-derived growth factor], VEGF), transdifferentiation (Notch), migration (ephrin, semaphorin), ECM remodeling (collagen, laminin, fibronectin), and inflammation (thrombospondin), following distension. CONCLUSIONS Vein conduit harvest and distension elicit a prompt genomic response facilitated by distinct cellular subpopulations heterogeneously distributed throughout the vein wall. This response was found to be further exacerbated following vein graft implantation, resulting in a cascade of maladaptive gene regulatory networks. Together, these results suggest that distension initiates the upregulation of pathological pathways that may ultimately contribute to bypass graft failure and presents potential early targets warranting investigation for targeted therapies. This work highlights the first applications of single-nuclei and spatial transcriptomic analyses to investigate venous pathologies, underscoring the utility of these methodologies and providing a foundation for future investigations.
Article
Full-text available
The ability of cells to adhere to each other and to their surrounding extracellular matrices is essential for a multicellular existence. Adhesion provides physical support for cells, regulates cell positioning and enables microenvironmental sensing. The integrins and the syndecans are two adhesion receptor families that mediate adhesion, but their relative and functional contributions to cell-extracellular matrix interactions remain obscure. Recent advances have highlighted connections between the signalling networks that are controlled by these families of receptors. Here we survey the evidence that synergistic signalling is involved in controlling adhesive function and the regulation of cell behaviour in response to the external environment.
Article
Full-text available
Vascular endothelial growth factor (VEGF) is a strong angiogenic mitogen and plays important roles in angiogenesis under various pathophysiological conditions. The in vivo angiogenic activity of secreted VEGF may be regulated by extracellular inhibitors, because it is also produced in avascular tissues such as the cartilage. To seek the binding inhibitors against VEGF, we screened the chondrocyte cDNA library by a yeast two-hybrid system by using VEGF165 as bait and identified connective tissue growth factor (CTGF) as a candidate. The complex formation of VEGF165 with CTGF was first established by immunoprecipitation from the cells overexpressing both binding partners. A competitive affinity-binding assay also demonstrated that CTGF binds specifically to VEGF165 with two classes of binding sites (Kd = 26 +/- 11 nM and 125 +/- 38 nM). Binding assay using deletion mutants of CTGF indicated that the thrombospondin type-1 repeat (TSP-1) domain of CTGF binds to the exon 7-coded region of VEGF165 and that the COOH-terminal domain preserves the affinity to both VEGF165 and VEGF121. The interaction of VEGF165 with CTGF inhibited the binding of VEGF165 to the endothelial cells and the immobilized KDR/IgG Fc; that is, a recombinant protein for VEGF165 receptor. By in vitro tube formation assay of endothelial cells, full-length CTGF and the deletion mutant possessing the TSP-1 domain inhibited VEGF165-induced angiogenesis significantly in the complex form. This antiangiogenic activity of CTGF was demonstrated further by in vivo angiogenesis assay by using Matrigel injection model in mice. These data demonstrate for the first time that VEGF165 binds to CTGF through a protein-to-protein interaction and suggest that the angiogenic activity of VEGF165 is regulated negatively by CTGF in the extracellular environment.
Article
In addition to their role in primary hemostasis, platelets serve to support and maintain the vascular endothelium. Platelets contain numerous growth factors including the potent angiogenic inducers VEGF and FGF-2. To characterize the function of these two plateletassociated growth factors, the effects of the addition of purified platelets to cultured endothelial cells were examined. The survival and proliferation of endothelial cells were markedly stimulated (2-3-fold and 5-15-fold respectively) by the addition of gel-filtered platelets. Acetylsalicylic acid-treated or lyophilized fixed-platelets were ineffective in supporting endothelial cell proliferation. In Transwell assays, the stimulatory effect of platelets on endothelial cells was preserved, consistent with an effect mediated by secreted factors. The combined inhibition of VEGF and FGF-2 by neutralizing antibodies, in contrast to inhibition of either alone, abrogated both platelet-induced endothelial cell survival and proliferation. FGF-2 isoforms were detected in platelet lysates, as well as in the releasates of agonist-stimulated platelets. Megakaryocytes generated by ex vivo expansion of hematopoietic progenitor cells with kit ligand and thrombopoietin were analyzed for expression of FGF-2. Punctate cytoplasmic staining but no nuclear staining was observed by immunocytochemistry consistent with possible localization of the growth factor to cytoplasmic granules. The addition of platelets to cultured endothelial cells activated extracellular signal-regulated kinase (ERK) in a dose and time-dependent manner. This effect was abrogated by both anti-FGF-2 and anti-VEGF antibody. Since FGF-2 and VEGF are potent angiogenic factors and known endothelial cell survival factors, their release by platelets provides a plausible mechanism for the platelet support of vascular endothelium.
Chapter
In 1971, the year that U.S. President Richard Nixon signed the National Cancer Act declaring the “war on cancer,” Judah Folkman of Harvard Medical School published his seminal hypothesis in the New England Journal of Medicine speculating that “tumors are angiogenesis-dependent” (1). This hypothesis led to the development of the field of angiogenesis research, now being pursued in hundreds of academic, private, and industry laboratories worldwide. During the past three decades, scientists elucidated the fundamental mechanisms underlying angiogenesis and vascular development in a variety of human diseases ranging from cancer to ischemic cardiovascular diseases to blindness and psoriasis, among others. Beginning in the late 1980s, the biopharmaceutical industry began aggressively exploiting this field for creating new therapeutic compounds for modulating new blood-vessel growth in angiogenesis-dependent diseases.
Chapter
Injuries of many types and degrees, ranging from inflammation to frank blood vessel disruption, lead to plasma extravasation and extravascular fibrin clot formation. The clot is composed of crosslinked fibrin and fibronectin and platelets that together entrap plasma water, plasma proteins, and blood cells, primarily erythrocytes. This fibrin gel matrix not only halts bleeding but also establishes a provisional matrix for the influx of inflammatory cells and par-enchymal cells into the injured tissue. With time, this surface clot dessicates to form the familiar scab, which in turn sloughs as wound healing proceeds from below.