Open Access

Heme oxygenase‑1 improves the survival of ischemic skin flaps (Review)

  • Authors:
    • Yinhua Zheng
    • Zhenlan Li
    • Min Yin
    • Xu Gong
  • View Affiliations

  • Published online on: January 26, 2021     https://doi.org/10.3892/mmr.2021.11874
  • Article Number: 235
  • Copyright: © Zheng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Heat shock protein 32 (Hsp32), also known as heme oxygenase‑1 (HO‑1), is an enzyme that exists in microsomes. HO‑1 can be induced by a variety of stimuli, including heavy metals, heat shock, inflammatory stimuli, heme and its derivatives, stress, hypoxia, and biological hormones. HO‑1 is the rate‑limiting enzyme of heme catabolism, which splits heme into biliverdin, carbon monoxide (CO) and iron. The metabolites of HO‑1 have anti‑inflammatory and anti‑oxidant effects, and provide protection to the cardiovascular system and transplanted organs. This review summarizes the biological characteristics of HO‑1 and the functional significance of its products, and specifically elaborates on its protective effect on skin flaps. HO‑1 improves the survival rate of ischemic skin flaps through anti‑inflammatory, anti‑oxidant and vasodilatory effects of enzymatic reaction products. In particular, this review focuses on the role of carbon monoxide (CO), one of the primary metabolites of HO‑1, in flap survival and discusses the feasibility and existing challenges of HO‑1 in flap surgery.

Introduction

In the fields of plastic surgery, hand and foot surgery, and burn treatment, skin flaps are particularly important for the closure of tissue defects. However, the ability to attenuate or even abolish the necrosis that occurs on skin flaps, mainly due to ischemia, remains challenging. Necrosis of the skin flap is not only related to anatomical factors of the skin flap itself (13), but also to some high-risk factors of the patient, such as age and diabetes (47). For example, Las et al (8) reported that diabetes, excessive anesthesia time and postoperative wound infection are risk factors for the failure of free flaps. Bekara et al (5) showed that an age older than 60 years, or the presence of diabetes and arterial diseases are risk factors for complications of lower extremity pedicle perforator flaps. In addition, Sanati-Mehrizy et al (4) reported that smoking and operation time are risk factors for free flap necrosis and de Blacam et al (7) found that venous congestion and increased age are risk factors for pedicle flap failure. Current literature regarding the risk factors of skin flap necrosis mainly focus on free skin flaps and lower limb pedicled skin flaps (815), and treatment methods that have been proposed are based on various mechanisms such as surgical delay, chemical delay, extracorporeal shock wave therapy, local thermal pretreatment, percutaneous neuroelectric pretreatment, cold pretreatment, negative pressure suction, targeted gene therapy, and drug injection (1625). Heat shock proteins (Hsps), which are highly evolutionarily conserved from prokaryotes to human (26), are molecular chaperons that exhibit a variety of biological activities including anti-oxidative, anti-apoptotic and anti-inflammatory effects (27). In recent years, several Hsps have been shown to prevent skin flap necrosis by reducing inflammation, oxidative stress, apoptosis and regulating platelet activity (2832).

Hsp32, a member of the Hsp family also known as heme oxygenase (HO)-1, is the most commonly studied molecule in the HO family of proteins. However, clinical data concerning HO-1 in flap surgery are scarce. This review summarizes the biological characteristics of HO-1 and the functional significance of its products. In particular, this review focuses on the role of carbon monoxide (CO), one of the primary metabolites of HO-1, in flap survival, elaborates on the protective effect of HO-1 on skin flaps (Table I), and discusses the feasibility and existing challenges of HO-1 in flap surgery.

Table I.

Summary of the literature regarding Hsp32 application in the field of flap surgery.

Table I.

Summary of the literature regarding Hsp32 application in the field of flap surgery.

Authors (ref.)YearResearch subjectSkin flap modelStimulusMajor findings (flap necrosis: Control vs. stimulus)
Rücker et al (36)2001RatOsteomyocutaneous flaps of left hindlimbLocal heat-shockStress initiated by local heat shock improved nutrient perfusion of the musculocutaneous flap through HO-1-elicited capillary dilation.
Rücker et al (84)2001RatOsteomyocutaneous flaps of left hindlimbLocal heat-shockLocal heat shock-induced stress increased HO-1 expression in musculocutaneous flaps, which was related to a significant decrease in leukocyte adhesion, ICAM-1 expression and high endothelial permeability. Hsp32 inhibitors completely abolished the anti-inflammatory effect of HO-1. These observations suggest that local heat shock-initiated stress can reduce the inflammatory response of the musculocutaneous flap.
Harder et al (93)2004PigA random-pattern skin flap was collected on both sides of the buttocksLocal heat preconditioningLocal heat preconditioning significantly reduced ischemic flap necrosis by decreasing complications of ischemia-related wound healing (40±8 vs. 7±14%, P<0.01).
Kubulus et al (95)2004MouseAxial-pattern skin flap of the earSurgical delayOverall protection against flap necrosis by chronic ischemic conditioning (delay) was mediated by HO-1, not through an anti-oxidative function but rather through a vasodilatory action, which prevented microvascular perfusion failure (41±3 vs. 16±3%, P<0.05).
Harder et al (34)2005MouseA random-pattern myocutaneous flap was collected on the backLocal heat preconditioningLocal heat preconditioning of myocutaneous tissue markedly increased flap survival by maintaining adequate nutrient perfusion rather than inducing ischemic tolerance (53±5 vs. 4±1%, P<0.001).
Chang et al (37)2005HumanGingival fibroblasts and tissues were obtained from cigarette smokers and non-smokersNicotineThe expression of HO-1 in gingival tissues of smokers was significantly upregulated; nicotine and other components may be a reason for the increased expression of HO-1. Regulation of nicotine-induced HO-1 expression was heavily dependent on the intracellular concentration of GSH.
Kubulus et al (23)2005MouseAxial-pattern skin flap of the earCooling-induced preconditioningCold pretreatment induced significant expression of HO-1, but not Hsp70 protein. HO-1 induced capillary dilation, thereby improving microvascular perfusion.
Harder et al (94)2007MouseAxial-pattern skin flap of the earAgingThe increased susceptibility to ischemic necrosis during aging is more likely due to a loss of vascular reactivity to endogenous HO-1, resulting in a lack of adequate adaptation to chronically ischemic conditions [49±8% (P<0.05) and 42±8% (P<0.05) in aging and adult patients, compared with 31±6% in young patients].
Contaldo et al (38)2007PigA random-pattern skin flap was raised on both sides of the buttocksMPL and local hyperthermiaiNOS and HO-1 were upregulated after MPL and local hyperthermia (P<0.05), but only local hyperthermia significantly reduced the necrosis rate of skin flaps [44% in local hyperthermia, compared with 29% in control (P<0.05)].
Harder et al (92)2008MouseRandom flap on the backChronic ischemiaChronic ischemia-induced endogenous HO-1 protected ischemic tissues through the vasodilatory action of HO-1-associated CO release (73±5 vs. 51±5%, P<0.001).
Schürmann et al (39)2009MouseCaudally pedicled skin flap on the backSurgery of a caudally-based skin flapSkin flap epithelial keratinocytes presented a prominent inflammatory response upon surgery, which was not amplified but rather controlled by invading HO-1 expressing-macrophages in the surviving flap tissues.
Sun et al (96)2013Human cells and ratRandom-pattern skin flaps on backISOISO preconditioning improved survival of skin flaps by upregulating the expression of HIF-1α, HO-1 and VEGF (51.5±5.6 vs. 43.3±6.7%, P<0.05).
Edmunds et al (91)2014Rat and human epidermal keratinocytes (HEKs)A transverse rectus abdominismyocutaneous flapHACompared with the control group, the HO-1 agonist HA failed to improve the survival rate of ischemic flaps (Compared with the control group, skin flap necrosis was more than 30% higher in HO-1 agonist HA group; P=0.002). In vitro studies have shown that HA is cytotoxic to keratinocytes.
Han et al (86)2015RatAbdominal rectangular random flapADSCsAdjuvant treatment with ADSCs significantly increased the survival of a skin flap in the venous ischemia-reperfusion condition. This effect was achieved through the suppression of the inflammatory response and induction of the antioxidative response (68.7±11.9 vs. 48.4±13.6%, P<0.05).
Lin et al (28)2019RatMultiterritory perforator flap on the backGBGB reduced oxidative stress through the activation of Nrf2/HO-1 signaling and enhancement of antioxidant activity (37.03±6.50 vs. 9.17±1.93%, P<0.01).

[i] Hsp, heat shock protein; HO-1, heme oxygenase-1; ICAM-1, intercellular adhesion molecule-1; GSH, glutathione; iNOS, inducible nitric oxide synthase; MPL, ministration of monophosphoryl lipid A; HIF-1α, hypoxia-inducible factor 1α; VEGF, vascular endothelial growth factor; HA, hemearginate; ISO, isoflurane; HA, heme arginate; ADSCs, adipose-derived stem cells; GB, ginkgolide B; Nrf2, nuclear factor erythroid-2-related factor 2.

Biological characteristics of HO-1

The human HO-1 gene, located on chromosome 22q12, is approximately 1414 kb in length and contains four introns and five exons. The length of the (GT)n microsatellite structure is unique to the promoter region of the human HO-1 gene and affects its transcription. A longer (GT)n leads to more GT dinucleotide sequence repeats and decreased HO-1 gene transcription and expression levels (33).

The HO family consists of three members: HO-1, HO-2 and HO-3. HO-1 is the rate-limiting enzyme of heme metabolism (34), but the functions of HO-2 and HO-3 remain elusive. HO-1, with a molecular weight of 32 kDa, is a stress protein that is either not expressed or has exceptionally low expression under normal conditions. However, HO-1 can be induced by a variety of stimuli, including heavy metals, heat shock, inflammatory stimuli, heme and its derivatives, stress, hypoxia, and biological hormones (3539). Increased HO-1 expression under stress reduces protein oxidation and lipid peroxidation, and attenuates cell and blood vessel damage, thus playing a protective role (40,41). For instance, Chen et al (40) reported that stress significantly increases HO-1 expression in the heart, which in turn provides cardioprotection. On the other hand, most metalloporphyrins, including tin protoporphyrin and zinc protoporphyrin, are inhibitors of HO-1 and compete with heme for the HO-1 binding site, thereby inhibiting the biological effects of HO-1 (42,43). The molecular weights of HO-2 and HO-3 are 36 kDa and 33 kDa, respectively (44). It is known that adrenal cortex hormones can induce the expression of HO-2, and it is therefore speculated that HO-2 can regulate functions within the nervous system. HO-3 has 90% primary structure homology with HO-2 but does not have any enzymatic activity. It is speculated that HO-3 regulates heme-dependent functions within cells (45).

HO-1-mediated enzymatic cascades and the functional significance of its products

HO-1 exists in microsomes and is the rate-limiting enzyme for heme catabolism, which splits heme into biliverdin, CO and iron. Biliverdin is further reduced to bilirubin under the action of biliverdin reductase, which is an effective anti-oxidant that protects cells from oxidative stress (46,47). Another metabolite, CO, acts on soluble guanylate cyclase (sGC) to increase the production of cyclic guanosine monophosphate (cGMP), which serves as a second messenger to regulate a wide spectrum of cellular events, including vasodilation (48,49) as well as anti-inflammatory and anti-apoptotic activities (50) (Fig. 1).

Heme

Heme is a pro-oxidant molecule that participates in the formation of oxidative free radicals leading to oxidative damage (51). In a model of heme overload using nude mice, excessive heme in the plasma promotes the production of reactive oxygen species (ROS) and reduces the effectiveness of nitric oxide (NO), thereby affecting the expansion of blood vessels (52). HO-1 degrades heme, and therefore has anti-oxidant activity. Balla et al (51) showed that the combination of heme and ferritin, together with HO-1, prevents oxidative stress. In another study performed in cultured alveolar epithelial cells, HO-1 overexpression was found to lead to a nearly 3-fold increase in ferritin, which was accompanied by a compensatory increase in transferrin receptors, subsequently altering the distribution of iron in the cells and protecting cells from iron toxicity induced by heme degradation (53).

CO anti-inflammatory activity

The anti-inflammatory activity of CO and CO-releasing molecules (CORM) has been well documented in various animal models. Increased HO-1 expression or CO prevents impairment of the blood-brain barrier (BBB), cerebral microvascular congestion and neuroinflammation (54). Mechanistically, this protection is achieved by the binding of CO to hemoglobin to prevent its oxidation and the production of free hemoglobin (54). In addition, a carrageenan-induced inflammation model showed that CO inhibits neutrophil migration and white blood cell adhesion (55). On the other hand, CORM-2 reduces the accumulation of inflammatory cells, the expression of intercellular adhesion molecule-1 (ICAM-1) and the activation of nuclear factor κB (NF-κB) in septic mice (56). In vitro experimental study confirmed that the mechanisms behind the anti-inflammatory effect of CORM-2 are related to the decreased expression of NF-κB-dependent vascular endothelial cell adhesion molecules and secretion of inducible nitric oxide synthase (iNOS) (56). Using an animal model related to human sepsis (cecal ligation and puncture in rodents), CORM-2 or CORM-3 was found to reduce the migration of polymorphonuclear (PMN) leukocytes to purulent tissues, thereby attenuating inflammation and saving animals from succumbing to sepsis (5759). In HO-1-deficient mice, treatment with CORM-2 was found to ameliorate the severity of sepsis associated with Enterococcus faecalis infection (60). CORM-3 was found to have a bactericidal effect on Pseudomonas aeruginosa, thereby reducing the mortality of mice with bacteremia (61). Additionally, CO exerts prominent anti-inflammatory actions in part by reducing the release of tumor necrosis factor α (TNF-α), interleukin (IL)-1, macrophage inflammatory protein (MIP)-1 and IL-6 from activated macrophages (62,63).

Protective effect of CO in cardiovascular diseases

The beneficial effects of CO on the cardiovascular system have been gradually discovered. Due to severe arterial thrombosis, HO-1-deficient mice died within four days following an aortic transplantation; however, treatment with CORM-2 was found to improve the survival rate of HO-1-deficient mice (64). The underlying protective mechanisms are related to the inhibition of platelet aggregation through the activation of guanylate cyclase (65). Endogenous CO binds to and activates sGC, subsequently increasing the concentration of cGMP and inducing a vasodilatory response (50,66). In a mouse model of myocardial infarction caused by coronary artery ligation, intravenous infusion of CORM-3 prior to reperfusion was found to reduce infarct size, fibrillation and tachycardia (67,68). The mechanisms underlying CORM-3-mediated cardiovascular protection involve potassium channels, since a small amount of CORM-3 is lost when mitochondrial ATP-dependent potassium channel inhibitors are used (67,68). Another study also demonstrated that HO-1 promotes angiogenesis through CO (69), and improves the survival of tissues surrounding the blood supply by inhibiting apoptosis of endothelial cells (70).

Preservation of transplanted organs

Many scholars have reported that CO plays an important role in organ transplantation and preservation. CO in a gaseous form or as a CORM can be used as a preservation adjuvant in organ preservation solution. Using a model of allogeneic heart transplantation in rodents, continuous inhalation of CO or CORM can prolong the survival time of the transplanted heart (71,72). The cardioprotective effect of CO gas is achieved by inhibiting platelet aggregation and endothelial cell apoptosis, as well as producing endogenous vasoconstrictors, thereby improving the microcirculation of the graft (71,72). Compared with the untreated control group, rabbit kidneys washed with CORM-3 or CORM-1 and then stored in a cold solution for 24 h showed higher rates of perfusion flow, glomerular filtration, and sodium and glucose reabsorption (73). The above-mentioned mechanisms of renal protection are thought to be related to CO-medicated expansion of blood vessels, inhibition of cell apoptosis and promotion of angiogenesis (73).

Bilirubin and biliverdin

As previously mentioned, HO-1 generates biliverdin, which is then converted to bilirubin. Both bilirubin and biliverdin are effective oxidative inhibitors (74). Human biliverdin reductase (hBVR), a Ser/Thr/Tyr kinase, is activated by insulin and free radicals (75). hBVR is central to the activation as well as the nuclear import and export of extracellular signal-regulated kinase (ERK)1/2 (76). Insulin increases BVR tyrosine phosphorylation and increases glucose uptake when BVR is knocked down by small interfering RNA, suggesting an important function of BVR in insulin signaling (77). HO-1-derived bilirubin is an efficient scavenger of reactive oxygen and nitrogen species (RONS) (78). In macrophages, biliverdin induces the phosphorylation of endothelial NO synthase (eNOS) at Ser1177, which generates eNOS. It is well recognized that NO plays a crucial role in regulating a wide spectrum of vascular functions, including vasorelaxation, inhibition of leukocyte-endothelial adhesion, vascular smooth muscle cell migration and proliferation, and platelet aggregation (79).

In summary, each product generated by HO-1 plays an important role in protecting cells and tissues through a variety of mechanisms.

Protective effects of HO-1 on skin flaps

Anti-inflammatory effect

The survival rate of skin flaps is related to ischemia-reperfusion injury and surgical trauma. Ischemia-reperfusion injury, if not treated in a timely and appropriate manner, may cause unfavorable outcomes for patients. Both surgical trauma and ischemia-reperfusion may initiate inflammatory responses, causing white blood cells to roll and adhere to the capillary vein (8082). In the early reperfusion phase, the accumulation of activated leukocytes and reactive oxygen metabolites aggravates the inflammatory response and ultimately impairs endothelial integrity (83). The initiation of the stress response leads to increased HO-1, which significantly reduces the adhesion of white blood cells to the endothelial surface and lessens the impairment of the integrity of venous endothelium (84). Rücker et al (84) studied whether stress conditioning-induced HO-1 could prevent an inflammatory response in transferred osteomyocutaneous flaps. In all tissues analyzed, control flaps presented with significant leukocyte adherence in postcapillary venules, increased expression of intercellular adhesion molecule-1 (ICAM-1), and disruption of endothelial integrity. In contrast, stress conditioning induced considerable HO-1 expression, which coincided with a significant reduction of leukocyte adherence, ICAM-1 expression, and endothelial hyperpermeability. Correspondingly, the inhibition of HO-1 by tin protoporphyrin IX completely abolished the stress conditioning-induced amelioration of the inflammatory response (84). Hence, the protective effect elicited by stress conditioning is mainly mediated by the induction of HO-1, which reduces oxygen free radicals, ICAM-1, adherence of white blood cells to the endothelial surface, endothelial permeability and macromolecular extravasation, ultimately reducing the overall inflammatory response.

Anti-oxidative effect

Rats undergoing flap reconstruction surgery experience lipid peroxidation and vascular damage, which is related to ischemia-reperfusion injury (85). Ischemia-reperfusion injury involves a complex oxidation process, which is closely related to flap survival (86). Oxidative stress induces an excessive activation of inflammatory processes, which increases ROS. ROS are molecules known to cause tissue damage through multiple mechanisms, including altering the structure and chemistry of proteins, lipids, and nucleic acids. Hence, reducing the release of ROS or removing excessive ROS protects tissues from ischemia-reperfusion injury (87). Lin et al (88) reported that CORM-2 induces HO-1 expression, thereby reducing protein kinase C (PKC)/amino acid-rich tyrosine kinase 2 (Pyk2)-dependent production of ROS. Lin et al (28) showed that ginkgolide B reduced skin flap necrosis and improved the survival of island perforator flaps by inhibiting endoplasmic reticulum stress and oxidative stress. In this study, they further showed that ginkgolide B activated nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Following its activation, Nrf2 is transported to the nucleus, where it binds to antioxidant response elements located in the cis-regulatory sequences of antioxidant-related enzymes and proteins and increases their expression (89).

Anti-apoptosis, vasodilation and angiogenesis

Brouard et al (90) studied the anti-apoptotic potential of HO-1 in cultured endothelial cells and found that CO, the product of heme digested by HO-1, activates the p38 mitogen-activated protein kinase (MAPK) pathway to inhibit endothelial cell apoptosis. But the HO-1 agonist heme arginate (HA) failed to improve the survival rate of ischemic flaps (91). In vitro studies have shown that HA is cytotoxic to keratinocytes (91). Brouard et al (90) also found that CO causes vasodilation, which is part of the mechanism by which HO-1 protects skin flaps from chronic ischemia-induced injury (90). Chronic ischemia was found to increase the expression of HO-1, especially on the first and third days post-operation. Increased HO-1 expression in turn induces arteriole expansion and promotes perfusion, which can maintain sufficient capillary perfusion density in the flap and reduce flap necrosis (92). The inhibition of endogenous HO-1 by tin protoporphyrin IX was found to completely eliminate arteriole expansion and hyperperfusion, resulting in a significant decrease in functional capillary density, a significant increase in cell apoptosis, and skin flap necrosis, but aging can reverse this protective effect (9294). Consistent with the above findings, Kubulus et al (95) reported that delay-associated tissue protection can be obtained by HO-1-mediated attenuation of microcirculatory dysfunction. Increased HO-1 activity protects tissues by biliverdin-associated anti-oxidative actions and/or CO-mediated vasodilation. This is supported by findings that show that trolox treatment after selective blockade of HO-1 reduced necrosis only until day three after flap creation and did not affect the manifestation of microcirculatory disorders. These observations suggested that: i) oxidative stress contributes to the initial development of flap necrosis; ii) anti-oxidative treatment counteracts oxidative stress-induced cell damage; iii) the initial oxidative stress does not define the degree or severity of final flap necrosis; and iv) HO-1 prevents flap necrosis induced by chronic ischemic conditioning (delay) primarily through its vasodilatory action and by improving microvascular perfusion (95). Kubulus et al (23) further showed that cooling induced a marked expression of HO-1 without induction of the Hsp70 protein, which was accompanied by significant improvement in microvascular perfusion. Thus, HO-1 also plays a role in the cooling-mediated amelioration of microcirculation, which results in a significant reduction in final flap necrosis. In another independent study, Sun et al (96) showed that preconditioning with isoflurane promotes hypoxic stabilization and induces the accumulation of hypoxia-inducible factor 1α (HIF-1 α), while HIF-1 αmediates transcriptional activation of the HO-1 gene in response to hypoxia to induce angiogenesis.

However, research has also shown that HO-1 may have dual functions. For example, HO-1 has a dual role in cancer cells. The levels of cellular iron and ROS are the determining factors for the role of HO-1, in which excessive cellular iron and ROS tend to push HO-1 from a protective role to that of a perpetrator. In general, a moderate level of HO-1 activation exerts a cytoprotective effect, while the over-activation of HO-1 becomes cytotoxic due to the excessive increase of labile Fe2+ behind the buffering capacity of ferritin (97). Similarly, HO-1 also has a dual role in nerve cells. Nrf2-dependent activation of HO-1 is generally linked to protective effects in neurons and glial cells, while Nrf2-independent activation of HO-1, which often involves AP-1 or NF-κB, seems to exert neurotoxic effects. Indeed, HO-1 expression is associated with neuronal damage and neurodegeneration, especially in Alzheimer's and Parkinson's disease (98).

The application of CO, the catalytic product of HO-1, within a clinical setting also has the following issues. For example, the safety constraints of CO require clinical dosing to maintain levels of carboxyhemoglobin (CO-Hb) that are under 14%, which is in stark contrast to the preclinical frontrunner studies with protocols that had sustained CO-Hb levels above 20% (99). Also, the pharmacokinetics of CO inhalation protocols varied significantly by species. Rodent models achieve therapeutic levels quickly, whereas the time to steady state in humans is roughly three times longer based on respiratory rate and cardiac output (100).

Similarly, the application of HO-1 or its metabolites to the field of flap surgery will also face the above-mentioned issues. Instead, perhaps it would be possible to: i) use HO-1 inhibitors (such as tin protoporphyrin) to repress its overexpression; ii) use an occasional monitoring system to achieve feedback to control the level of CO in the body; or iii) search for molecules that promote the release of CO or solutions that can dissolve CO, and apply them locally to reduce systemic adverse reactions.

Perspectives

HO-1 was first described in the late 1960s and its functions have been gradually characterized over the following decades, innovative observations have been made over the past 20 years uncovering the cellular cytoprotective capability of HO-1 in a clinical setting. While studies have defined to some degree the tissue protective roles of each of the by-products generated by HO-1 (including CO) and attempted to harness HO-1 to improve CO-delivery for clinical applications, there are still significant limitations that prevent its use at the bed side (99). Previous studies have shown the clinical relevance of HO-1, such as in ischemic stroke (101), skin health (102) and cardiovascular syndromes and co-morbidities (103). However, there are few studies on the application of HO-1 in the field of human flap surgery. How to direct the clinical application of HO-1 and its related products in the field of flap surgery in order to obtain more appreciable outcomes will be the focus of future research.

Acknowledgements

The authors would like to thanks Dr Huanfa Yi from the Second Department of the First Hospital of Jilin University, who provided critical comments and invaluable advice on this manuscript.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

YZ drafted the manuscript. ZL assisted with the literature search. MY assisted with drafting and revising the manuscript. XG conceived and designed the review. YZ and XG are responsible for confirming the authenticity of the raw data. All authors read and approved the final manuscript and agree to be accountable for the accuracy and referencing of the information included in the review.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

Hsp32

heat shock protein 32

HO-1

heme oxygenase-1

CO

carbon monoxide

sGC

soluble guanylate cyclase

cGMP

cyclic guanosine monophosphate

ROS

reactive oxygen species

CORM

carbon monoxide-releasing molecules

BBB

blood-brain barrier

ICAM-1

intercellular adhesion molecule-1

NF-κB

nuclear factor κB

iNOS

inducible nitric oxide synthase

PMN

polymorphonuclear

TNF-α

tumor necrosis factor α

IL

interleukin

MIP

macrophage inflammatory protein

hBVR

human biliverdin reductase

ERK

extracellular signal regulated kinase

RONS

reactive oxygen and nitrogen species

eNOS

endothelial nitric oxide synthase

PKC

protein kinase C

MAPK

mitogen activated protein kinase

HIF-1α

hypoxia-inducible factor 1α

Nrf2

nuclear factor erythroid-2-related factor 2

References

1 

Taylor GI, Corlett RJ, Dhar SC and Ashton MW: The anatomical (angiosome) and clinical territories of cutaneous perforating arteries: Development of the concept and designing safe flaps. Plast Reconstr Surg. 127:1447–1459. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Saint-Cyr M, Wong C, Schaverien M, Mojallal A and Rohrich RJ: The perforasome theory: Vascular anatomy and clinical implications. Plast Reconstr Surg. 124:1529–1544. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Callegari PR, Taylor GI, Caddy CM and Minabe T: An anatomic review of the delay phenomenon: I. Experimental studies. Plast Reconstr Surg. 89:397–407; discussion 417-398. 1992. View Article : Google Scholar : PubMed/NCBI

4 

Sanati-Mehrizy P, Massenburg BB, Rozehnal JM, Ingargiola MJ, Hernandez Rosa J and Taub PJ: Risk factors leading to free flap failure: Analysis from the national surgical quality improvement program database. J Craniofac Surg. 27:1956–1964. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Bekara F, Herlin C, Mojallal A, Sinna R, Ayestaray B, Letois F, Chavoin JP, Garrido I, Grolleau JL and Chaput B: A systematic review and meta-analysis of perforator-pedicled propeller flaps in lower extremity defects: Identification of risk factors for complications. Plast Reconstr Surg. 137:314–331. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Wong AK, Joanna Nguyen T, Peric M, Shahabi A, Vidar EN, Hwang BH, Niknam Leilabadi S, Chan LS and Urata MM: Analysis of risk factors associated with microvascular free flap failure using a multi-institutional database. Microsurgery. 35:6–12. 2015. View Article : Google Scholar : PubMed/NCBI

7 

de Blacam C, Colakoglu S, Ogunleye AA, Nguyen JT, Ibrahim AM, Lin SJ, Kim PS and Lee BT: Risk factors associated with complications in lower-extremity reconstruction with the distally based sural flap: A systematic review and pooled analysis. J Plast Reconstr Aesthet Surg. 67:607–616. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Las DE, de Jong T, Zuidam JM, Verweij NM, Hovius SE and Mureau MA: Identification of independent risk factors for flap failure: A retrospective analysis of 1530 free flaps for breast, head and neck and extremity reconstruction. J Plast Reconstr Aesthet Surg. 69:894–906. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Li B, Chang SM, Du SC, Zhuang L and Hu SJ: Distally based sural adipofascial turnover flap for coverage of complicated wound in the foot and ankle region. Ann Plast Surg. 84:580–587. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Lee ZH, Abdou SA, Ramly EP, Daar DA, Stranix JT, Anzai L, Saadeh PB, Levine JP and Thanik VD: Larger free flap size is associated with increased complications in lower extremity trauma reconstruction. Microsurgery. 40:473–478. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Stranix JT, Lee ZH, Anzai L, Jacoby A, Avraham T, Saadeh PB, Levine JP and Thanik VD: Optimizing venous outflow in reconstruction of Gustilo IIIB lower extremity traumas with soft tissue free flap coverage: Are two veins better than one? Microsurgery. 38:745–751. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Gupta A, Lakhiani C, Lim BH, Aho JM, Goodwin A, Tregaskiss A, Lee M, Scheker L and Saint-Cyr M: Free tissue transfer to the traumatized upper extremity: Risk factors for postoperative complications in 282 cases. J Plast Reconstr Aesthet Surg. 68:1184–1190. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Reece EM, Bonelli MA, Livingston T, Mulligan PS, Rockwood J, Wilson JR, Zoldos J and Champagne L: Factors in free fasciocutaneous flap complications: A logistic regression analysis. Plast Reconstr Surg. 136:54e–58e. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Innocenti M, Menichini G, Baldrighi C, Delcroix L, Vignini L and Tos P: Are there risk factors for complications of perforator-based propeller flaps for lower-extremity reconstruction? Clin Orthop Relat Res. 472:2276–2286. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Dolan RT, Butler JS, Murphy SM and Cronin KJ: Health-related quality of life, surgical and aesthetic outcomes following microvascular free flap reconstructions: An 8-year institutional review. Ann R Coll Surg Engl. 94:43–51. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Ellabban MA, Fattah IOA, Kader GA, Eldeen OS, Mehana AE, Khodeer DM, Hosny H, Elbasiouny MS and Masadeh S: The effects of sildenafil and/or nitroglycerin on random-pattern skin flaps after nicotine application in rats. Sci Rep. 10:32122020. View Article : Google Scholar : PubMed/NCBI

17 

Lee W, Oh W, Oh SM and Yang EJ: Comparative effectiveness of different interventions of perivascular hyaluronidase. Plast Reconstr Surg. 145:957–964. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Giatsidis G, Cheng L, Haddad A, Ji K, Succar J, Lancerotto L, Lujan-Hernandez J, Fiorina P, Matsumine H and Orgill DP: Noninvasive induction of angiogenesis in tissues by external suction: Sequential optimization for use in reconstructive surgery. Angiogenesis. 21:61–78. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Giatsidis G, Cheng L, Facchin F, Haddad A, Lujan-Hernandez J, Lancerotto L, Nabzdyk CG, Matsumine H and Orgill DP: Moderate-intensity intermittent external volume expansion optimizes the soft-tissue response in a murine model. Plast Reconstr Surg. 139:882–890. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Doğan F and Özyazgan İ: Flap preconditioning by electrical stimulation as an alternative to surgical delay: Experimental study. Ann Plast Surg. 75:560–564. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Menevşe GT, TeomanTellioglu A, Altuntas N, Cömert A and Tekdemir I: Polidocanol injection for chemical delay and its effect on the survival of rat dorsal skin flaps. J Plast Reconstr Aesthet Surg. 67:851–856. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Tobalem M, Wettstein R, Pittet-Cuénod B, Vigato E, Machens HG, Lohmeyer JA, Rezaeian F and Harder Y: Local shockwave-induced capillary recruitment improves survival of musculocutaneous flaps. J Surg Res. 184:1196–1204. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Kubulus D, Amon M, Roesken F, Rücker M, Bauer I and Menger MD: Experimental cooling-induced preconditioning attenuates skin flap failure. Br J Surg. 92:1432–1438. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Morris SF and Taylor GI: The time sequence of the delay phenomenon: When is a surgical delay effective? An experimental study. Plast Reconstr Surg. 95:526–533. 1995. View Article : Google Scholar : PubMed/NCBI

25 

Akimoto M, Takeda A, Matsushita O, Inoue J, Sakamoto K, Hattori M, Kounoike N and Uchinuma E: Effects of CB-VEGF-A injection in rat flap models for improved survival. Plast Reconstr Surg. 131:717–725. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Lindquist S and Craig EA: The heat-shock proteins. Annu Rev Genet. 22:631–677. 1988. View Article : Google Scholar : PubMed/NCBI

27 

Miller DJ and Fort PE: Heat shock proteins regulatory role in neurodevelopment. Front Neurosci. 12:8212018. View Article : Google Scholar : PubMed/NCBI

28 

Lin D, Wu H, Zhou Z, Tao Z, Jia T and Gao W: Ginkgolide B improves multiterritory perforator flap survival by inhibiting endoplasmic reticulum stress and oxidative stress. J Invest Surg. 1–7. Dec 23–2019.(Epub ahead of print). View Article : Google Scholar

29 

Kumar D, Jena GR, Ram M, Lingaraju MC, Singh V, Prasad R, Kumawat S, Kant V, Gupta P, Tandan SK and Kumar D: Hemin attenuated oxidative stress and inflammation to improve wound healing in diabetic rats. Naunyn Schmiedebergs Arch Pharmacol. 392:1435–1445. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Kankam HKN, Mehta S and Jain A: Thermal preconditioning for surgery: A systematic review. J Plast Reconstr Aesthet Surg. 73:1645–1664. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Park C, Lee H, Noh JS, Jin CY, Kim GY, Hyun JW, Leem SH and Choi YH: Hemistepsin A protects human keratinocytes against hydrogen peroxide-induced oxidative stress through activation of the Nrf2/HO-1 signaling pathway. Arch Biochem Biophys. 691:1085122020. View Article : Google Scholar : PubMed/NCBI

32 

Jackson JW, Rivera-Marquez GM, Beebe K, Tran AD, Trepel JB, Gestwicki JE, Blagg BSJ, Ohkubo S and Neckers LM: Pharmacologic dissection of the overlapping impact of heat shock protein family members on platelet function. J Thromb Haemost. 18:1197–1209. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Matsunobu T, Satoh Y, Ogawa K and Shiotani A: Heme oxygenase-1 expression in the guinea pig cochlea induced by intense noise stimulation. Acta Otolaryngol Suppl. 18–23. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Harder Y, Amon M, Schramm R, Georgi M, Banic A, Erni D and Menger MD: Heat shock preconditioning reduces ischemic tissue necrosis by heat shock protein (HSP)-32-mediated improvement of the microcirculation rather than induction of ischemic tolerance. Ann Surg. 242:869–878; discussion 878-869. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Tian W, Bonkovsky HL, Shibahara S and Cohen DM: Urea and hypertonicity increase expression of heme oxygenase-1 in murine renal medullary cells. Am J Physiol Renal Physiol. 281:F983–F991. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Rücker M, Schäfer T, Roesken F, Spitzer WJ, Bauer M and Menger MD: Local heat-shock priming-induced improvement in microvascular perfusion in osteomyocutaneous flaps is mediated by heat-shock protein 32. Br J Surg. 88:450–457. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Chang YC, Lai CC, Lin LF, Ni WF and Tsai CH: The up-regulation of heme oxygenase-1 expression in human gingival fibroblasts stimulated with nicotine. J Periodontal Res. 40:252–257. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Contaldo C, Harder Y, Plock J, Banic A, Jakob SM and Erni D: The influence of local and systemic preconditioning on oxygenation, metabolism and survival in critically ischaemic skin flaps in pigs. J Plast Reconstr Aesthet Surg. 60:1182–1192. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Schürmann C, Seitz O, Klein C, Sader R, Pfeilschifter J, Mühl H, Goren I and Frank S: Tight spatial and temporal control in dynamic basal to distal migration of epithelial inflammatory responses and infiltration of cytoprotective macrophages determine healing skin flap transplants in mice. Ann Surg. 249:519–534. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Chen YH, Lin SJ, Lin MW, Tsai HL, Kuo SS, Chen JW, Charng MJ, Wu TC, Chen LC, Ding YA, et al: Microsatellite polymorphism in promoter of heme oxygenase-1 gene is associated with susceptibility to coronary artery disease in type 2 diabetic patients. Hum Genet. 111:1–8. 2002. View Article : Google Scholar : PubMed/NCBI

41 

Schillinger M, Exner M, Mlekusch W, Ahmadi R, Rumpold H, Mannhalter C, Wagner O and Minar E: Heme oxygenase-1 genotype is a vascular anti-inflammatory factor following balloon angioplasty. J Endovasc Ther. 9:385–394. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Whitington PF, Moscioni AD and Gartner LM: The effect of tin (IV)-protoporphyrin-IX on bilirubin production and excretion in the rat. Pediatr Res. 21:487–491. 1987. View Article : Google Scholar : PubMed/NCBI

43 

Vreman HJ, Ekstrand BC and Stevenson DK: Selection of metalloporphyrin heme oxygenase inhibitors based on potency and photoreactivity. Pediatr Res. 33:195–200. 1993. View Article : Google Scholar : PubMed/NCBI

44 

Elbirt KK and Bonkovsky HL: Heme oxygenase: Recent advances in understanding its regulation and role. Proc Assoc Am Physicians. 111:438–447. 1999. View Article : Google Scholar : PubMed/NCBI

45 

Adach W, Błaszczyk M and Olas B: Carbon monoxide and its donors-chemical and biological properties. Chem Biol Interact. 318:1089732020. View Article : Google Scholar : PubMed/NCBI

46 

Sorrenti V: Editorial of special issue ‘protective and detrimental role of heme oxygenase-1’. Int J Mol Sci. 20:47442019. View Article : Google Scholar

47 

Sha JY, Zhou YD, Yang JY, Leng J, Li JH, Hu JN, Liu W, Jiang S, Wang YP, Chen C and Li W: Maltol (3-Hydroxy-2-methyl-4-pyrone) slows d-galactose-induced brain aging process by damping the Nrf2/HO-1-mediated oxidative stress in mice. J Agric Food Chem. 67:10342–10351. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Schallner N, Romão CC, Biermann J, Lagrèze WA, Otterbein LE, Buerkle H, Loop T and Goebel U: Carbon monoxide abrogates ischemic insult to neuronal cells via the soluble guanylate cyclase-cGMP pathway. PLoS One. 8:e606722013. View Article : Google Scholar : PubMed/NCBI

49 

Ndisang JF, Wu L, Zhao W and Wang R: Induction of heme oxygenase-1 and stimulation of cGMP production by hemin in aortic tissues from hypertensive rats. Blood. 101:3893–3900. 2003. View Article : Google Scholar : PubMed/NCBI

50 

Motterlini R and Otterbein LE: The therapeutic potential of carbon monoxide. Nat Rev Drug Discov. 9:728–743. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Balla G, Jacob HS, Balla J, Rosenberg M, Nath K, Apple F, Eaton JW and Vercellotti GM: Ferritin: A cytoprotective antioxidant strategem of endothelium. J Biol Chem. 267:18148–18153. 1992. View Article : Google Scholar : PubMed/NCBI

52 

Vinchi F, De Franceschi L, Ghigo A, Townes T, Cimino J, Silengo L, Hirsch E, Altruda F and Tolosano E: Hemopexin therapy improves cardiovascular function by preventing heme-induced endothelial toxicity in mouse models of hemolytic diseases. Circulation. 127:1317–1329. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Fang R and Aust AE: Induction of ferritin synthesis in human lung epithelial cells treated with crocidolite asbestos. Arch Biochem Biophys. 340:369–375. 1997. View Article : Google Scholar : PubMed/NCBI

54 

Pamplona A, Ferreira A, Balla J, Jeney V, Balla G, Epiphanio S, Chora A, Rodrigues CD, Gregoire IP, Cunha-Rodrigues M, et al: Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimental cerebral malaria. Nat Med. 13:703–710. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Freitas A, Alves-Filho JC, Secco DD, Neto AF, Ferreira SH, Barja-Fidalgo C and Cunha FQ: Heme oxygenase/carbon monoxide-biliverdin pathway down regulates neutrophil rolling, adhesion and migration in acute inflammation. Br J Pharmacol. 149:345–354. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Cepinskas G, Katada K, Bihari A and Potter RF: Carbon monoxide liberated from carbon monoxide-releasing molecule CORM-2 attenuates inflammation in the liver of septic mice. Am J Physiol Gastrointest Liver Physiol. 294:G184–G191. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Mizuguchi S, Stephen J, Bihari R, Markovic N, Suehiro S, Capretta A, Potter RF and Cepinskas G: CORM-3-derived CO modulates polymorphonuclear leukocyte migration across the vascular endothelium by reducing levels of cell surface-bound elastase. Am J Physiol Heart Circ Physiol. 297:H920–H929. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Tsoyi K, Lee TY, Lee YS, Kim HJ, Seo HG, Lee JH and Chang KC: Heme-oxygenase-1 induction and carbon monoxide-releasing molecule inhibit lipopolysaccharide (LPS)-induced high-mobility group box 1 release in vitro and improve survival of mice in LPS- and cecal ligation and puncture-induced sepsis model in vivo. Mol Pharmacol. 76:173–182. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Lancel S, Hassoun SM, Favory R, Decoster B, Motterlini R and Neviere R: Carbon monoxide rescues mice from lethal sepsis by supporting mitochondrial energetic metabolism and activating mitochondrial biogenesis. J Pharmacol Exp Ther. 329:641–648. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Chung SW, Liu X, Macias AA, Baron RM and Perrella MA: Heme oxygenase-1-derived carbon monoxide enhances the host defense response to microbial sepsis in mice. J Clin Invest. 118:239–247. 2008. View Article : Google Scholar : PubMed/NCBI

61 

Desmard M, Davidge KS, Bouvet O, Morin D, Roux D, Foresti R, Ricard JD, Denamur E, Poole RK, Montravers P, et al: A carbon monoxide-releasing molecule (CORM-3) exerts bactericidal activity against pseudomonas aeruginosa and improves survival in an animal model of bacteraemia. FASEB J. 23:1023–1031. 2009. View Article : Google Scholar : PubMed/NCBI

62 

Morse D, Pischke SE, Zhou Z, Davis RJ, Flavell RA, Loop T, Otterbein SL, Otterbein LE and Choi AM: Suppression of inflammatory cytokine production by carbon monoxide involves the JNK pathway and AP-1. J Biol Chem. 278:36993–36998. 2003. View Article : Google Scholar : PubMed/NCBI

63 

Otterbein LE, Bach FH, Alam J, Soares M, Tao Lu H, Wysk M, Davis RJ, Flavell RA and Choi AM: Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway. Nat Med. 6:422–428. 2000. View Article : Google Scholar : PubMed/NCBI

64 

Chen B, Guo L, Fan C, Bolisetty S, Joseph R, Wright MM, Agarwal A and George JF: Carbon monoxide rescues heme oxygenase-1-deficient mice from arterial thrombosis in allogeneic aortic transplantation. Am J Pathol. 175:422–429. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Brüne B and Ullrich V: Inhibition of platelet aggregation by carbon monoxide is mediated by activation of guanylate cyclase. Mol Pharmacol. 32:497–504. 1987.PubMed/NCBI

66 

Wang R: Resurgence of carbon monoxide: An endogenous gaseous vasorelaxing factor. Can J Physiol Pharmacol. 76:1–15. 1998. View Article : Google Scholar : PubMed/NCBI

67 

Varadi J, Lekli I, Juhasz B, Bacskay I, Szabo G, Gesztelyi R, Szendrei L, Varga E, Bak I, Foresti R, et al: Beneficial effects of carbon monoxide-releasing molecules on post-ischemic myocardial recovery. Life Sci. 80:1619–1626. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Guo Y, Stein AB, Wu WJ, Tan W, Zhu X, Li QH, Dawn B, Motterlini R and Bolli R: Administration of a CO-releasing molecule at the time of reperfusion reduces infarct size in vivo. Am J Physiol Heart Circ Physiol. 286:H1649–H1653. 2004. View Article : Google Scholar : PubMed/NCBI

69 

Józkowicz A, Huk I, Nigisch A, Weigel G, Dietrich W, Motterlini R and Dulak J: Heme oxygenase and angiogenic activity of endothelial cells: Stimulation by carbon monoxide and inhibition by tin protoporphyrin-IX. Antioxid Redox Signal. 5:155–162. 2003. View Article : Google Scholar : PubMed/NCBI

70 

Soares MP, Usheva A, Brouard S, Berberat PO, Gunther L, Tobiasch E and Bach FH: Modulation of endothelial cell apoptosis by heme oxygenase-1-derived carbon monoxide. Antioxid Redox Signal. 4:321–329. 2002. View Article : Google Scholar : PubMed/NCBI

71 

Clark JE, Naughton P, Shurey S, Green CJ, Johnson TR, Mann BE, Foresti R and Motterlini R: Cardioprotective actions by a water-soluble carbon monoxide-releasing molecule. Circ Res. 93:e2–e8. 2003. View Article : Google Scholar : PubMed/NCBI

72 

Sato K, Balla J, Otterbein L, Smith RN, Brouard S, Lin Y, Csizmadia E, Sevigny J, Robson SC, Vercellotti G, et al: Carbon monoxide generated by heme oxygenase-1 suppresses the rejection of mouse-to-rat cardiac transplants. J Immunol. 166:4185–4194. 2001. View Article : Google Scholar : PubMed/NCBI

73 

Sandouka A, Fuller BJ, Mann BE, Green CJ, Foresti R and Motterlini R: Treatment with CO-RMs during cold storage improves renal function at reperfusion. Kidney Int. 69:239–247. 2006. View Article : Google Scholar : PubMed/NCBI

74 

Jansen T and Daiber A: Direct antioxidant properties of bilirubin and biliverdin. Is there a role for biliverdin reductase? Front Pharmacol. 3:302012.

75 

Maines MD, Miralem T, Lerner-Marmarosh N, Shen J and Gibbs PE: Human biliverdin reductase, a previously unknown activator of protein kinase C betaII. J Biol Chem. 282:8110–8122. 2007. View Article : Google Scholar : PubMed/NCBI

76 

Gibbs PE, Tudor C and Maines MD: Biliverdin reductase: More than a namesake-the reductase, its peptide fragments, and biliverdin regulate activity of the three classes of protein kinase C. Front Pharmacol. 3:312012. View Article : Google Scholar : PubMed/NCBI

77 

Lerner-Marmarosh N, Shen J, Torno MD, Kravets A, Hu Z and Maines MD: Human biliverdin reductase: A member of the insulin receptor substrate family with serine/threonine/tyrosine kinase activity. Proc Natl Acad Sci USA. 102:7109–7114. 2005. View Article : Google Scholar : PubMed/NCBI

78 

Jansen T, Hortmann M, Oelze M, Opitz B, Steven S, Schell R, Knorr M, Karbach S, Schuhmacher S, Wenzel P, et al: Conversion of biliverdin to bilirubin by biliverdin reductase contributes to endothelial cell protection by heme oxygenase-1-evidence for direct and indirect antioxidant actions of bilirubin. J Mol Cell Cardiol. 49:186–195. 2010. View Article : Google Scholar : PubMed/NCBI

79 

Pae HO, Son Y, Kim NH, Jeong HJ, Chang KC and Chung HT: Role of heme oxygenase in preserving vascular bioactive NO. Nitric Oxide. 23:251–257. 2010. View Article : Google Scholar : PubMed/NCBI

80 

Eppihimer MJ and Granger DN: Ischemia/reperfusion-induced leukocyte-endothelial interactions in postcapillary venules. Shock. 8:16–25. 1997. View Article : Google Scholar : PubMed/NCBI

81 

Kunkel EJ, Jung U, Bullard DC, Norman KE, Wolitzky BA, Vestweber D, Beaudet AL and Ley K: Absence of trauma-induced leukocyte rolling in mice deficient in both P-selectin and intercellular adhesion molecule 1. J Exp Med. 183:57–65. 1996. View Article : Google Scholar : PubMed/NCBI

82 

Menger MD and Vollmar B: Adhesion molecules as determinants of disease: From molecular biology to surgical research. Br J Surg. 83:588–601. 1996. View Article : Google Scholar : PubMed/NCBI

83 

Menger MD, Pelikan S, Steiner D and Messmer K: Microvascular ischemia-reperfusion injury in striated muscle: Significance of ‘reflow paradox’. Am J Physiol. 263:H1901–H1906. 1992.PubMed/NCBI

84 

Rücker M, Schäfer T, Roesken F, Spitzer WJ, Bauer M and Menger MD: Reduction of inflammatory response in composite flap transfer by local stress conditioning-induced heat-shock protein 32. Surgery. 129:292–301. 2001. View Article : Google Scholar : PubMed/NCBI

85 

Taleb S, Moghaddas P, Rahimi Balaei M, Taleb S, Rahimpour S, Abbasi A, Ejtemaei-Mehr S and Dehpour AR: Metformin improves skin flap survival through nitric oxide system. J Surg Res. 192:686–691. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Han HH, Lim YM, Park SW, Lee SJ, Rhie JW and Lee JH: Improved skin flap survival in venous ischemia-reperfusion injury with the use of adipose-derived stem cells. Microsurgery. 35:645–652. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Forrester SJ, Kikuchi DS, Hernandes MS, Xu Q and Griendling KK: Reactive oxygen species in metabolic and inflammatory signaling. Circ Res. 122:877–902. 2018. View Article : Google Scholar : PubMed/NCBI

88 

Lin CC, Hsiao LD, Cho RL and Yang CM: Carbon monoxide releasing molecule-2-upregulated ROS-dependent heme oxygenase-1 axis suppresses lipopolysaccharide-induced airway inflammation. Int J Mol Sci. 20:31572019. View Article : Google Scholar

89 

Shi Y, Liang XC, Zhang H, Sun Q, Wu QL and Qu L: Combination of quercetin, cinnamaldehyde and hirudin protects rat dorsal root ganglion neurons against high glucose-induced injury through Nrf-2/HO-1 activation and NF-κB inhibition. Chin J Integr Med. 23:663–671. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Brouard S, Otterbein LE, Anrather J, Tobiasch E, Bach FH, Choi AM and Soares MP: Carbon monoxide generated by heme oxygenase 1 suppresses endothelial cell apoptosis. J Exp Med. 192:1015–1026. 2000. View Article : Google Scholar : PubMed/NCBI

91 

Edmunds MC, Czopek A, Wigmore SJ and Kluth DC: Paradoxical effects of heme arginate on survival of myocutaneous flaps. Am J Physiol Regul Integr Comp Physiol. 306:R10–R22. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Harder Y, Amon M, Schramm R, Rücker M, Scheuer C, Pittet B, Erni D and Menger MD: Ischemia-induced up-regulation of heme oxygenase-1 protects from apoptotic cell death and tissue necrosis. J Surg Res. 150:293–303. 2008. View Article : Google Scholar : PubMed/NCBI

93 

Harder Y, Contaldo C, Klenk J, Banic A, Jakob SM and Erni D: Improved skin flap survival after local heat preconditioning in pigs. J Surg Res. 119:100–105. 2004. View Article : Google Scholar : PubMed/NCBI

94 

Harder Y, Amon M, Georgi M, Scheuer C, Schramm R, Rücker M, Pittet B, Erni D and Menger MD: Aging is associated with an increased susceptibility to ischaemic necrosis due to microvascular perfusion failure but not a reduction in ischaemic tolerance. Clin Sci (Lond). 112:429–440. 2007. View Article : Google Scholar : PubMed/NCBI

95 

Kubulus D, Roesken F, Amon M, Rücker M, Bauer M, Bauer I and Menger MD: Mechanism of the delay phenomenon: Tissue protection is mediated by heme oxygenase-1. Am J Physiol Heart Circ Physiol. 287:H2332–H2340. 2004. View Article : Google Scholar : PubMed/NCBI

96 

Sun Y, Li QF, Zhang Y, Hu R and Jiang H: Isoflurane preconditioning increases survival of rat skin random-pattern flaps by induction of HIF-1α expression. Cell Physiol Biochem. 31:579–591. 2013. View Article : Google Scholar : PubMed/NCBI

97 

Chiang SK, Chen SE and Chang LC: A dual role of heme oxygenase-1 in cancer cells. Int J Mol Sci. 20:392018. View Article : Google Scholar

98 

Nitti M, Piras S, Brondolo L, Marinari UM, Pronzato MA and Furfaro AL: Heme oxygenase 1 in the nervous system: Does it favor neuronal cell survival or induce neurodegeneration? Int J Mol Sci. 19:22602018. View Article : Google Scholar

99 

Hopper CP, Meinel L, Steiger C and Otterbein LE: Where is the clinical breakthrough of heme oxygenase-1/carbon monoxide therapeutics? Curr Pharm Des. 24:2264–2282. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Steiger C, Hermann C and Meinel L: Localized delivery of carbon monoxide. Eur J Pharm Biopharm. 118:3–12. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Bereczki D Jr, Balla J and Bereczki D: Heme oxygenase-1: Clinical relevance in ischemic stroke. Curr Pharm Des. 24:2229–2235. 2018. View Article : Google Scholar : PubMed/NCBI

102 

Szabo IL, Kenyeres A, Szegedi A and Szollosi AG: Heme oxygenase and the skin in health and disease. Curr Pharm Des. 24:2303–2310. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Haines DD and Tosaki A: Role of heme oxygenases in cardiovascular syndromes and co-morbidities. Curr Pharm Des. 24:2322–2325. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2021
Volume 23 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zheng Y, Li Z, Yin M and Gong X: Heme oxygenase‑1 improves the survival of ischemic skin flaps (Review). Mol Med Rep 23: 235, 2021
APA
Zheng, Y., Li, Z., Yin, M., & Gong, X. (2021). Heme oxygenase‑1 improves the survival of ischemic skin flaps (Review). Molecular Medicine Reports, 23, 235. https://doi.org/10.3892/mmr.2021.11874
MLA
Zheng, Y., Li, Z., Yin, M., Gong, X."Heme oxygenase‑1 improves the survival of ischemic skin flaps (Review)". Molecular Medicine Reports 23.4 (2021): 235.
Chicago
Zheng, Y., Li, Z., Yin, M., Gong, X."Heme oxygenase‑1 improves the survival of ischemic skin flaps (Review)". Molecular Medicine Reports 23, no. 4 (2021): 235. https://doi.org/10.3892/mmr.2021.11874