Next Article in Journal
Optimal Volume Assessment for Serous Fluid Cytology
Previous Article in Journal
No Impact of Enteral Nutrition on Fecal Short-Chain Fatty Acids in Children with Cerebral Palsy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

The Pharmacological Effect of Hemin in Inflammatory-Related Diseases: A Systematic Review

1
H&TRC—Health and Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal
2
Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(4), 898; https://doi.org/10.3390/biomedicines12040898
Submission received: 7 March 2024 / Revised: 8 April 2024 / Accepted: 12 April 2024 / Published: 18 April 2024
(This article belongs to the Special Issue Molecular Mechanism in Inflammation and Immunity)

Abstract

:
Background: Hemin is clinically used in acute attacks of porphyria; however, recent evidence has also highlighted its capability to stimulate the heme oxygenase enzyme, being associated with cytoprotective, antioxidant, and anti-inflammatory effects. Indeed, current preclinical evidence emphasizes the potential anti-inflammatory role of hemin through its use in animal models of disease. Nevertheless, there is no consensus about the underlying mechanism(s) and the most optimal therapeutic regimens. Therefore, this review aims to summarize, analyze, and discuss the current preclinical evidence concerning the pharmacological effect of hemin. Methods: Following the application of the search expression and the retrieval of the articles, only nonclinical studies in vivo written in English were considered, where the potential anti-inflammatory effect of hemin was evaluated. Results: Forty-nine articles were included according to the eligibility criteria established. The results obtained show the preference of using 30 to 50 mg/kg of hemin, administered intraperitoneally, in both acute and chronic contexts. This drug demonstrates significant anti-inflammatory and antioxidant activities considering its capacity for reducing the expression of proinflammatory and oxidative markers. Conclusions: This review highlighted the significant anti-inflammatory and antioxidant effects of hemin, providing a clearer vision for the medical community about the use of this drug in several human diseases.

Graphical Abstract

1. Introduction

Hemin, also known as ferriprotoporphyrin IX chloride (Figure 1), is an iron-containing metalloporphyrin, commonly used in the treatment of acute attacks of porphyria [1,2]. Theoretically, porphyria is intimately related to a deficient heme biosynthesis pathway, resulting in a lack of heme necessary to produce several hemoproteins. Additionally, it can also result in the accumulation of heme precursors, which are directly and indirectly toxic to the human organism [2,3,4]. Hemin reduces heme deficiency by suppressing delta-aminolaevulinic acid synthase activity. Consecutively, it decreases the concentration of porphyrins and toxic precursors of heme [2,4,5]. In this sense, once the levels of hemoproteins and respiratory pigments become normal and stable, the biological abnormalities observed in patients with porphyria are attenuated [4,5].
Recent evidence has demonstrated that hemin also has the capability of inducing the expression of the heme oxygenase (HO) enzyme, known as a rate-limiting enzyme for heme catabolism, responsible for producing biliverdin, free iron, and carbon monoxide [7,8,9]. Currently, there are three HO isoenzymes identified in mammals, namely HO-1, HO-2, and HO-3, where the first one is usually associated with cytoprotective, anti-inflammatory, and antioxidant activities [7,8,10,11]. Importantly, the induction in HO-1 is intimately related to the capability of hemin to activate the nuclear factor erythroid 2 related factor 2, and, at the same time, to inactivate the repression factor Bach1 [12]. Moreover, preclinical evidence has shown significantly higher values of HO-1 and HO-1 mRNA in inflammatory-related disorders, like, for example, colitis [7,8,11]. The expression of this isoenzyme is usually observed in macrophages; however, it can also be noticed in epithelial cells present in the region where the inflammatory response is occurring [13,14]. Considering the information above, hemin appears as a new potential pharmacological approach in several inflammatory-related human disorders due to its high capacity to stimulate the expression of HO-1.
Currently, an interesting research question for future studies has arisen concerning the existence of several nonclinical studies in vivo that demonstrate the capability of hemin to reduce inflammatory responses. It is essential to understand the underlying mechanism(s) behind the anti-inflammatory and antioxidant effects of this drug to evaluate its potential role in clinical practice. Even though there is already a significant number of nonclinical studies demonstrating the effect of hemin on inflammation, there is an absence of review articles that analyze and criticize the evidence available. In addition, considering the different therapeutic regimens currently described, it is essential to investigate the most optimal ones. Therefore, according to this major gap in the literature, this systematic review aims to summarize, analyze, and discuss the preclinical evidence that is currently available regarding the pharmacological effect of hemin in animal models of disease.

2. Materials and Methods

The present systematic review was developed considering the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA 2020) guideline [15]. Additionally, the protocol of this review was registered in the International Prospective Register of Systematic Reviews—PROSPERO (CRD42023406160) and published in an international research journal [16].

2.1. Eligibility Criteria

The studies will be selected according to the following inclusion criteria: only original articles; studies where a pharmacological treatment with hemin was evaluated; studies that analyzed inflammation and/or oxidative stress; nonclinical studies in vivo with rodents; and articles only published in English. In addition, regarding the exclusion criteria, the following were considered: review articles; expert opinions; book chapters; studies with only in vitro procedures; and experiments that evaluated the treatment with hemin in diseases present in the summary of product characteristics (e.g., porphyria).

2.2. Information Sources and Search Strategy

The biomedical electronic databases used for this highly sensitive search strategy were MEDLINE (Pubmed), Web of Science, and Scopus. The search was only limited by the publication date, namely between 2015 and March 2023. A comprehensive research expression was developed using descriptors related to three terms (hemin, inflammation, and nonclinical study) and their synonyms combined with Boolean operators “AND” and “OR” to identify and select the eligible studies. The search strategies, adapted for each biomedical electronic database, are available in the Supplemental Material.

2.3. Selection Process

Upon the application of the search expression to the respective biomedical electronic databases, the retrieved articles were exported to a Systematic Reviews Web Application (Rayyan QCRI, Rayyan Systems, Cambridge, MA, USA). Following the exclusion of duplicates, the titles and abstracts of the studies were analyzed by two independent reviewers to select the relevant and potentially eligible articles. Afterward, the two independent reviewers assessed the full text of each potentially eligible article, deciding if it was included or not, considering the eligibility criteria established. In these two previous steps, a third element was included in case of discrepancies between the two reviewers to make a final decision.

2.4. Data Collection Process

The data present in the included studies were carefully extracted to the Excel software (Version 2403, Microsoft, Redmond, WA, USA) by the same two independent reviewers. A standardized data extraction document in Excel sheets (Microsoft) was developed to extract the information of interest, which considered article identification (first author’s name and year of publication); hemin-related parameters; disease animal model; and outcomes assessed. All the information of interest was extracted from the text, graphs, and/or tables from each included study.

2.5. Data Items

2.5.1. Population

Only rodent models where the data of interest to be extracted were related to species, strain, gender, and age were considered.

2.5.2. Intervention

Only hemin administration where the parameters of interest were related to dosage, frequency of administration, route of administration, and duration of treatment was considered.

2.5.3. Comparator

No comparator parameters were considered.

2.5.4. Outcomes

The data concerning the outcomes were related to inflammatory and biochemical markers (continuous quantitative measure), macroscopical evaluation (dichotomous measure), and microscopical evaluation (dichotomous measure).

2.5.5. Study Design

Throughout the analysis of the nonclinical studies in vivo, the data of interest to be extracted were related to the disease induced, severity, and chronicity.

2.6. Quality and Risk of Bias Assessment

To evaluate the internal validity of each study, along with the methodological quality, an analysis of the risk of bias through the application of SYRCLE’s risk of bias tool was considered [17]. For each study, the grading of all components present in SYRCLE’s tool was performed considering the following statements: complete (2 points); incomplete (1 point); or not mentioned (0 points). The final grade was decided concerning the average of all individual components, and the studies were assigned into the following categories of quality: high (15–20 points); moderate (8–14 points); or low (0–7 points). This evaluation was performed by the same two independent reviewers. Any disagreements were discussed and arbitrated by a third reviewer.

3. Results

3.1. Study Selection

The application of the search strategies, each one in the respective biomedical electronic database, retrieved a total of 1110 articles, where 606 were identified as duplicates (Figure 2). The analysis of the titles and abstracts from the remaining 504 articles resulted in the exclusion of 441 reports. In this sense, the full text of the 63 potentially eligible studies was critically assessed considering the inclusion and exclusion criteria. After this process, 13 articles were excluded concerning the following justifications: a study that applied a combined treatment of hemin with other active pharmacological substance (n = 1); studies that did not evaluate inflammatory and oxidative stress biomarkers (n = 3); review (n = 1); studies written in Chinese (n = 2); studies with no access for full-text analysis (n = 6), and a publication with retraction order (n = 1). The 49 studies included for qualitative synthesis met all the eligibility criteria, in which the potential anti-inflammation of hemin in rodent models of disease was adequately evaluated (Table 1).

3.2. Study Characteristics

Upon the inclusion of the 49 eligible studies, an extensive analysis was performed in each one to extract the relevant data, namely hemin- and animal-related parameters, along with the biomarkers assessed in the respective experiments (Table 1).

3.3. Risk of Bias Assessment

In the evaluation of the internal validity and methodological quality of each study, considering the application of SYRCLE’s tool for risk of bias assessment, it was possible to observe an absence of low-quality articles (Table 2 and Table S1 (Supplementary Material)). Indeed, 5 articles reached the highest quality level, while the other 44 were categorized as moderate.

4. Discussion

Current preclinical evidence demonstrates a significant anti-inflammatory effect of hemin in different scenarios, such as inflammatory bowel disease [7,8,44,65], kidney dysfunction [23,60], sepsis [40,66,67], arthritis [56,68], pancreatitis [32,53,69], cardiac infarction [46], and airway inflammation [27,29,70]. Considering the long-term toxicity of the current pharmacological treatments applied in chronic inflammatory diseases, it is essential to investigate safer options, either by the development of new molecules or by the process of drug repurposing [71]. Hemin appears as an interesting pharmacological approach in future treatment protocols of chronic inflammatory diseases concerning its efficacy and safety demonstrated in several nonclinical studies in vivo, as previously referred to. Regarding the different treatment protocols applied in preclinical studies, showing high variability in terms of doses, frequency, routes of administration, and duration, it is particularly difficult to understand the most optimal ones. Therefore, it is essential to analyze, summarize, and discuss several therapeutic regimens, providing a well-developed basis for future studies regarding the potential off-label use of hemin in inflammation-related disorders.

4.1. Hemin-Related Parameters

4.1.1. Dose

The studies analyzed demonstrated a high variability concerning the dose of hemin administered, where the most commonly used dose was 30 mg/kg (n = 7), followed by 50 mg/kg (n = 5). Indeed, it is possible to observe a wide range of doses, between 75 µg/kg and 50 mg/kg, which varies considering the route of administration and the animal model chosen. The anti-inflammatory effect of hemin was already possible to observe with a relatively low dose, namely 75 µg/kg, administrated in an animal model of acute pancreatitis, represented by a significant reduction in pathological score in the liver and pancreas, as well as a reduced expression of a proinflammatory cytokine, namely tumor necrosis factor-α (TNF-α) [33]. As expected, when hemin is used at a higher dose, such as 50 mg/kg, significant anti-inflammatory and antioxidant effects are also observed, which translate to reduced concentrations of proinflammatory cytokines (e.g., interleukin (IL)-1β, -6, nuclear factor kappa B (NF-Kb), and TNF-α) and oxidative markers (e.g., malondialdehyde (MDA) and myeloperoxidase (MPO)) [55,56,57,58,59,60]. Importantly, regarding the studies that administered higher doses of hemin, no significant side effects were identified.
In clinical practice, hemin is normally administered at a dose of 3–4 mg/Kg/day in the treatment of acute attacks of porphyria [72]. Therefore, it is possible to consider that this drug achieves its significant pharmacological activity at relatively low doses, concerning both preclinical and clinical settings.
Recently, it was described that HO-1 may influence the enzymatic activity of certain cytochrome P450s (e.g., CYP1A2, CYP2C2, CYP2E1, and CYP3A4). In fact, this can be justified by the fact that HO-1 forms homomeric complexes with these P450s enzymes, affecting their overall activity [73]. Then, the therapeutic regimen (dose, frequency, and duration of treatment) should equate to the potential influence of hemin on the metabolism of other drugs and xenobiotics in general, since it may increase the risk of toxic events.

4.1.2. Frequency of Administration and Duration of Treatment

Regarding the frequency of administration of hemin, it is possible to conclude that the majority of the studies preferred a single-dose treatment (n = 22), followed by daily administrations (n = 11) and every two days (n = 5). Considering the daily treatment, the spectrum of time varies between 2 and 14 days, depending on the disease induced and the chronicity of the therapeutic regimen desired. On the other hand, some studies decided to administer hemin two or three times per week for an extended therapeutic regimen between 8 and 20 weeks. It can be justified by the longer time needed to develop the respective animal model of disease (e.g., emphysema, obesity, steatohepatitis, and liver fibrosis) and the interest in evaluating the potential anti-inflammatory effect of hemin throughout the induction process [18,19,20,50]. Concerning the results obtained in the various studies, it is possible to observe that treatment with hemin demonstrated a significant anti-inflammatory effect in both acute and chronic settings, without showing significant side effects upon its use.
In clinical practice, the standard treatment of acute attacks of porphyria with hemin is through daily intravenous (IV) doses for four consecutive days [72]. However, it can be adapted to each individual, depending on the severity, to a maximum of 14 consecutive daily doses [74].

4.1.3. Route of Administration

Among the different routes of administration considered in the various studies analyzed, the most used was intraperitoneal (IP) (n = 37), followed by subcutaneous (SC) (n = 5) and topical (n = 3). The decision concerning the route of administration should always consider the species of the animal; the substance to be administered; the location where the effect should be noticed; and the total volume [75,76]. Additionally, some studies performed intragastric (n = 1) and pleural (n = 1) injections to make sure that hemin achieved the maximum therapeutic effect on the respective location, namely the stomach and lungs, with a lower rate of absorption and distribution [34,37].
Concerning the different parenteral routes of administration used in laboratory animals, the most commonly observed are IP, SC, and IV. Currently, considering clinical practice, hemin is only administered intravenously [72,74]. In the specific case of rodent models, the IP route is the most desirable one, demonstrating relative ease to perform, quickness, and minimal stress on the animals [75]. In addition, it presents a faster absorption rate in comparison with the SC route and is also preferable in the case of larger volumes. Regarding chronic treatments, the IP route is also recommended since repetitive IV access can be challenging over time [75,76]. Additionally, IV administration is easier to perform in rats compared with mice due to increased vessel diameter and visibility. Furthermore, this route of administration presents a major advantage compared with the IP and SC routes, namely the systemic delivery of substances, because it bypasses the need for solute absorption [77]. However, the unique study analyzed that performed IV administrations was in mice, more specifically, three daily injections [35]. Interestingly, significant anti-inflammatory and wound-healing effects from hemin in a topical application were also proven, revealing a potential route of administration to be considered in future preclinical and clinical studies [62,63,64].

4.2. Animal-Related Parameters

4.2.1. Disease Animal Model

Considering the several studies that evaluated the potential anti-inflammatory and antioxidant effects of hemin, it is possible to emphasize high variability in terms of disease animal models. However, the most commonly used were diabetes mellitus (n = 4), acute lung injury (ALI) (n = 3), colitis (n = 3), and sepsis (n = 3). In a general mode, it is also important to consider versability regarding treatment with hemin, demonstrating a significant anti-inflammatory role in various contexts, such as lungs (e.g., emphysema, airway inflammation, and asthma), kidneys (e.g., acute kidney injury, nephrotoxicity, and renal inflammation), liver (e.g., liver fibrosis, steatohepatitis, and acute liver failure), pancreas (e.g., acute pancreatitis), and cardiovascular system (e.g., myocardial infarction, vascular dysfunction, and restenosis).
Diabetes mellitus is a highly prevalent and complex metabolic disorder characterized by hyperglycemia, which is a physiologically abnormal condition represented by continued elevated blood glucose levels [78,79,80]. One of the major concerns about the progression of this disorder is related to complications in other organs, such as the kidneys, brain, heart, and skin [78]. The pharmacological activity of hemin has been tested in rodent models of diabetes mellitus to evaluate its efficacy and safety in nephropathy and wound-healing abnormalities [26,62,63,64]. Considering diabetes-induced nephropathy, hemin treatment was shown to have significant anti-inflammatory (e.g., reduction in TNF-α), antiapoptotic (e.g., reduction in caspase-3), and antioxidant (e.g., reduction in MDA, MPO, and nitric oxide) activities. In addition, it was also proved to have a beneficial effect in terms of renal morphology by decreasing the microscopical destruction observed [26]. On the other hand, concerning the diabetes-induced wound-healing abnormalities, the topical treatment with hemin also showed significant anti-inflammatory (e.g., reduction in TNF-α, IL-6, allied with the increase of IL-10) and antioxidant (e.g., increase in superoxide dismutase (SOD) and catalase, as well as the reduction in MDA and glutathione peroxidase) effects. Furthermore, it was possible to notice antiapoptotic (e.g., reduction in caspase-3), coagulating (e.g., extension of prothrombin and thrombin times and reduction in fibrinogen), and angiogenic (e.g., increase in vascular endothelial growth factor) properties from this drug. Finally, an acceleration in the process of wound closure and re-epithelialization was also observed [62,63,64]. It is important to note that the pharmacological activity of hemin was mainly related to the capacity to induce the expression of HO-1 and not to altering serum glucose levels [26,62,63,64].
ALI is normally caused by several pathological conditions, namely sepsis, pneumonia, multiple traumas, pulmonary contusion, or aspiration of dangerous gas [51]. Regarding its pathogenesis, ALI is characterized by neutrophil infiltration, accumulation of inflammatory mediators, and hyaline membrane formation [31,51,81]. This disorder is recognized as a major concern worldwide since it presents a high rate of mortality and the underlying mechanisms are not fully understood yet [31,51]. The treatment with hemin showed significant anti-inflammatory (e.g., reduction in NF-kB, caspase-1, IL-1β, -6, and TNF-α) and antioxidant (e.g., reduction in MDA, MPO, and reactive oxygen species, as well and the increase in SOD) effects. In addition, hemin was also capable of reducing the microscopical abnormalities related to the ALI induction and, consequently, the mortality rate [31,51,52].
Inflammatory bowel diseases are characterized as chronic inflammatory disorders in the gastrointestinal tract, which can be divided into two phenotypes, namely ulcerative colitis and Crohn’s disease [44]. Currently, there is no cure for these disorders, and the pharmacological approaches only aim to induce and maintain the remission phase in patients, rather than resolving the underlying mechanisms(s) [7,8,44,82]. Treatment with hemin was revealed to be a promising pharmacological approach in the future since it presented significant anti-inflammatory (e.g., reduction in cyclooxygenase-2, IL-1β, -6, -12, -18, interferon-γ, NF-kB, and TNF-α, allied with an increase in IL-10) and antioxidant (e.g., increase in SOD and catalase, as well as a reduction in MDA, MPO, and nitric oxide) properties [7,8,44]. Furthermore, treatment with hemin also showed the capability of partially reverting the destruction observed in the colon through histopathological analysis [7,44].
Sepsis is described as a major life-threatening organ dysfunction provoked by an exacerbated and dysregulated systemic host response to infection [61,83]. Considering the extensive organ abnormalities observed during this event, there are several complications to be noticed [40,61]. Indeed, concerning the preclinical evidence demonstrated in this review, the treatment with hemin was evaluated for different complications, such as muscle atrophy and intestinal injury [40,41,61]. Regarding the results obtained in these studies, it was possible to identify, once again, significant anti-inflammatory (e.g., reduction in caspase-1, -11, IL-1β, -6, -8, -18, TNF-α, and toll-like receptor-4) and antioxidant (e.g., reduction in MDA and glutamate dehydrogenase, allied with an increase in SOD) effects from hemin [38,39,60]. In addition, a significant reduction in the mortality rate after the treatment with this drug was also observed [40,41], as well as a partial reversion of the sepsis-induced damage observed in different organs, like the intestine, liver, lungs, kidneys, skeletal muscles, and heart [40,41,61].

4.2.2. Species and Strain

The use of laboratory animals, mainly rodents (e.g., rats and mice), is recognized as one of the central strategies for the study of the etiology and pathogenesis of several diseases, as well as for the evaluation of new potential pharmacological and nonpharmacological treatments [77,84,85]. Considering the results from this review, it is possible to conclude that the most widely used animal was the rat (n = 31), followed by mice (n = 18). Indeed, these rodent models present several similarities with humans, regarding gene homology, physiology, anatomy, pathological pathways, and immune system [77,86,87,88]. In addition, they also present some advantages, such as reduced breeding and housing costs, easy handling, relatively small size, short life cycle, and gestation time (19 to 21 days) [77,84,85,86]. Concerning statistical relevance, rodent models are relatively cost-effective, allowing the use of adequate sample sizes for further analysis [84,86].
Besides the high similarity between rodent models, a preference for the use of rats was observed, especially in the scenario of cardiovascular diseases. This preference can be justified by the fact that rats present a higher blood volume when compared with mice, promoting an easier and less stressful collection process. In addition, it is possible to observe more evidence regarding cardiovascular physiology and pathology, as well as disease-linked genome sites, in rats [89]. Nevertheless, the choice of using rats or mice is intimately dependent on researchers’ preference and the disease model presented. Generally, there were no differences regarding the pharmacological activity of hemin between the two rodent models previously discussed.
Regarding the different strains used, it is possible to determine that the most used in rats were Wistar (n= 17) and Sprague Dawley (n = 12), whereas in mice they were C57BL/6 (n = 10) and Balb/c (n = 6). The use of genetically modified strains in a higher proportion in mice (e.g., HO-1flox/flox (n = 1), A2A (n = 1) and A2B knockout (n = 1), DO11.10 (n = 2), and Bas-TRECK (n = 1)) when compared with rats was also observed (e.g., Zucker fatty (n = 1) and Zucker lean (n = 1)).
The decision regarding a specific strain can be relatively difficult and should consider several parameters, such as the objectives of the experiment, the disease animal model, and reproducibility, as well as ethical and financial viability [90]. Theoretically, each strain demonstrates both advantages and disadvantages in comparison with others that are available. Therefore, the final choice should be made after measuring the pros and cons of each strain. Currently, the most commonly used inbred mouse strains in biomedical research are C57BL/6 and BALB/c. Indeed, as inbred strains, each one of them is recognized as being homozygous and genetically stable, presenting consistent phenotypes. Normally, C57BL/6 mice can be useful in several contexts, being recognized as a general multipurpose model. Furthermore, regarding BALB/c mice, they are usually associated with immunology and infectiology research [91,92]. On the other hand, the Wistar and Sprague Dawley strains are the most popular outbred rats used in preclinical research, especially in toxicological and aging studies. The major differences among these species are related to food intake, growth rate, hormonal expression, and tumorigenesis [93].

4.2.3. Gender

The included studies showed a clear preference for using males (n = 35), to the detriment of females (n = 10). In addition, four studies did not mention the gender chosen. The major reason for this preference is related to the hormonal differences observed between males and females. Indeed, normally, a high variability regarding hormonal levels in females is observed throughout all experiments due to the estrous cycle [86,94]. As an example, increased concentrations of progesterone generally reduce an inflammatory response, which can be a problematic bias in the results from certain studies, especially when a potential anti-inflammatory drug is being tested [95,96]. However, the use of females is essential in several disease models, namely in the context of mastitis and endometrial hyperplasia, as presented above in Table 1 [47,55]. On the other hand, there are also major concerns regarding the use of males. In fact, they are significantly more territorial than females, increasing the overall stress, risk of fighting, and, consequently, mortality rate [97]. Therefore, males should be monitored regularly to prevent cases of severe skin lesions, which can induce a threatening infection with a potent inflammatory response, directly influencing the well-being of the animals and the results obtained [77,86].

4.2.4. Age

Regarding the age of the animals used in the included studies, it was possible to notice that rats and mice were between 1–16 and 5–12 weeks old, respectively. Generally, it can be highlighted that the majority of the studies used rodents aged between 6 and 12 weeks old. Nevertheless, some studies did not mention the exact age of the animals, confirming only that they were adults (n = 5).
In normal conditions, only the use of mice and rats beginning at 4 and 5 weeks old should be considered, respectively. In fact, until these ages, rodents have not yet passed the process of weaning and demonstrate an increased dependency on their mothers to survive [86,94]. Only one study used rats aged 1 week old; however, the experiment aimed to evaluate the hepatotoxicity of acetaminophen in neonatal animals. Interestingly, hemin showed significant anti-inflammatory (e.g., reduction in IL-1β, IL-6, and TNF-α) and antioxidant (e.g., reduction in MDA and increase in GSH) effects [57].
Researchers normally prefer rodents aged between 8 and 12 weeks old; however, it is important to consider the normal developmental processes that are ongoing, which can have a significant impact on the results [98]. Indeed, regarding immunology, B- and T-cell responses are usually mature at around 4 and 8 weeks old, respectively [98,99,100]. Furthermore, the production of these two types of cells increases during the first 26 weeks of life [98,101]. On the other hand, considering aged rodents, differences in terms of drug metabolism (e.g., abnormal gene expression of liver enzymes) and a more robust inflammatory response, allied with severe systemic manifestations, when compared with younger animals are normally observed [86,98,102,103]. However, aged rodents appear as useful models in cardiovascular research since they mimic, in most cases, the normal onset and progression of diseases observed in humans (e.g., diabetes mellitus and hypertension) [104]. Therefore, considering the information above, age can be recognized as a determinant factor during experimental research and should be adequate for a study’s objectives since it can directly influence the results obtained.

4.3. Biomarkers Assessed

Biomarkers have major importance during pharmaceutical discovery, considering both clinical and preclinical settings, since they permit the evaluation of the efficacy and safety of several molecules [105,106]. Considering the studies analyzed, it was possible to notice a high variability of biomarkers assessed. However, concerning inflammation, the most commonly observed markers were TNF-α (n = 32), IL-6 (n = 22), IL-1β (n = 20), and NF-kB (n = 10). Furthermore, considering oxidative stress, the most common biomarkers were MDA (n = 20), SOD (n = 12), glutathione (GSH) (n = 12), and MPO (n = 11).
Hemin demonstrated a significant anti-inflammatory effect, which can be justified by its capability to reduce the concentration of proinflammatory cytokines (e.g., IL-1β, -6, and TNF-α), as well as to increase the expression of a well-known anti-inflammatory marker, namely IL-10. In addition, this drug was also capable of decreasing the expression of an important inflammatory transcription factor, that is NF-kB. In normal situations, an inflammatory response is associated with oxidative stress, which plays a pivotal role in the onset and progression of several diseases [107]. In this regard, hemin also showed a significant antioxidant effect, demonstrated by a reduction in oxidative markers (e.g., MDA and MPO), allied with an increase in antioxidant mediators (e.g., SOD and GSH). Considering the scenario of animal models of inflammation, the biomarkers of interest can be divided into several classes (e.g., immune-related effectors, cytokines, chemokines, reactive oxygen and nitrogen species, prostaglandins, cyclooxygenases, acute phase proteins, and transcription factors), highlighting a well-balanced pharmacological effect of hemin concerning the positive results demonstrated above [86].
Finally, it is also important to emphasize that the majority of the studies also assessed the expression of HO-1 (n = 35), which appears to be a proof of concept for the stimulating effect of hemin in this enzyme. In fact, HO-1 is recognized as a stress protein and metabolic enzyme associated with regulatory effects in cellular and tissue homeostatic responses, modulation of the immune system, and host defense, as well as being a mitigator of inflammation [108]. Theoretically, HO-1 reduces the concentration of free heme, which is known to be involved in various human diseases due to its capability of interfering with several pathological responses (e.g., stimulation of toll-like receptor 4-dependent inflammatory response, membrane lipid peroxidation, and ferroptosis) [108]. Therefore, according to the current literature, it is possible to positively correlate the expression of HO-1, stimulated by the administration of hemin, as well as significant anti-inflammatory and antioxidant activities, which is an interesting pharmacological approach to be considered in future clinical practice.
Despite the evidence demonstrated in this systematic review, it is important to consider a major limitation regarding the methodology, which is the absence of clinical studies. However, at this date, no clinical trials are studying the potential anti-inflammatory effect of hemin, and, in a general mode, it is possible to notice a relatively reduced number of clinical studies that focus on this topic. In this sense, we decided to only include nonclinical studies in vivo since they present the most controlled environment possible. On the other hand, considering the specificity of the outcomes evaluated related to inflammation and oxidative stress, potentially interesting studies that demonstrated other pharmacological activities of hemin, such as its cytoprotective role, were not included. At the same time, it would also be interesting to evaluate the other side of hemin, more specifically, its potential toxic effect in certain tissues, such as in the neurological and gastrointestinal systems, since it has shown to be cytotoxic to cultured mouse astrocytes and colonic cells, respectively [109,110]. Nevertheless, this review can be the first step for future studies evaluating different pharmacological effects of hemin, excluding those already present in the summary of product characteristics.

5. Conclusions

This review highlighted an interesting pharmacological effect of hemin in several rodent models of disease, which was based on its significant anti-inflammatory and antioxidant activities. Regarding the use of this drug, most researchers preferred a dose ranging between 30 mg/kg and 50 mg/kg, administered intraperitoneally in most cases. Interestingly, hemin was successfully used in both acute and chronic contexts, demonstrating its pharmacological activity in different therapeutic regimens. In fact, positive results with only a single administration, as well as after several weeks of treatment, were observed. Generally, a significant reduction in proinflammatory (e.g., IL-1β, -6, NF-kB, and TNF-α) and oxidative (e.g., MDA and MPO) markers was noticed. In addition, hemin was also capable of increasing the expression of an anti-inflammatory cytokine, namely IL-10, and antioxidant (e.g., SOD and GSH) mediators. Therefore, regarding the preclinical evidence available about the pharmacological effect of hemin in several rodent models of disease, this drug may arise as a new potential approach in inflammatory-related human disorders, which is essential to evaluate it in a clinical setting.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines12040898/s1, Table S1: Application of the SYRCLE’s tool for assessing the risk of bias in each included study.

Author Contributions

Conceptualization, V.M.; methodology, J.E. and G.C.; software, J.E. and G.C.; validation, V.M.; formal analysis, J.E. and G.C.; investigation, V.M., J.E. and G.C.; data curation, J.E. and G.C.; writing—original draft preparation, J.E.; writing—review and editing, G.C., I.S., P.M. and V.M.; supervision, V.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

There is no additional data created despite the information present in the manuscript and Supplementary Material. Nevertheless, further inquiries can be directed to the corresponding author.

Acknowledgments

The authors from the Health and Technology Research Center gratefully acknowledge the support of the Fundação para a Ciência e a Tecnologia/Ministério da Ciência, Tecnologia e Ensino Superior (UIDB/05608/2020 and UIDP/05608/2020).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bonkowsky, H.L.; Tschudy, D.P.; Collins, A.; Doherty, J.; Bossenmaier, I.; Cardinal, R.; Watson, C.J. Repression of the Overproduction of Porphyrin Precursors in Acute Intermittent Porphyria by Intravenous Infusions of Hematin. Proc. Natl. Acad. Sci. USA 1971, 68, 2725–2729. [Google Scholar] [CrossRef] [PubMed]
  2. Anderson, K.E.; Collins, S. Open-Label Study of Hemin for Acute Porphyria: Clinical Practice Implication. Am. J. Med. 2006, 119, 801.e1–801.e6. [Google Scholar] [CrossRef] [PubMed]
  3. Phillips, J.D. Heme biosynthesis and the porphyrias. Mol. Genet. Metab. 2019, 128, 164–177. [Google Scholar] [CrossRef]
  4. Stölzel, U.; Doss, M.O.; Schuppan, D. Clinical Guide and Update on Porphyrias. Gastroenterology 2019, 157, 365–381.e4. [Google Scholar] [CrossRef] [PubMed]
  5. Gasson, T.; Klein, K. Porphyria. Nurse Pract. 2015, 40, 1–6. [Google Scholar] [CrossRef] [PubMed]
  6. Tahoun, M.; Engeser, M.; Svolacchia, L.; Sander, P.M.; Müller, C.E. Molecular Taphonomy of Heme: Chemical Degradation of Hemin under Presumed Fossilization Conditions. Molecules 2023, 28, 4887. [Google Scholar] [CrossRef]
  7. Mateus, V.; Rocha, J.; Mota-Filipe, H.; Sepodes, B.; Pinto, R. Hemin reduces inflammation associated with TNBS-induced colitis. Clin. Exp. Gastroenterol. 2018, 11, 325–334. [Google Scholar] [CrossRef]
  8. Silva, I.; Correia, R.; Pinto, R.; Mateus, V. Hemin Ameliorates the Inflammatory Activity in the Inflammatory Bowel Disease: A Non-Clinical Study in Rodents. Biomedicines 2022, 10, 2025. [Google Scholar] [CrossRef] [PubMed]
  9. Naito, Y.; Takagi, T.; Uchiyama, K.; Yoshikawa, T. Heme oxygenase-1: A novel therapeutic target for gastrointestinal diseases. J. Clin. Biochem. Nutr. 2011, 48, 126–133. [Google Scholar] [CrossRef]
  10. Abraham, N.G.; Kappas, A. Pharmacological and Clinical Aspects of Heme Oxygenase. Pharmacol. Rev. 2008, 60, 79–127. [Google Scholar] [CrossRef]
  11. Campbell, N.K.; Fitzgerald, H.K.; Dunne, A. Regulation of inflammation by the antioxidant haem oxygenase 1. Nat. Rev. Immunol. 2021, 21, 411–425. [Google Scholar] [CrossRef] [PubMed]
  12. Kasai, S.; Mimura, J.; Ozaki, T.; Itoh, K. Emerging Regulatory Role of Nrf2 in Iron, Heme, and Hemoglobin Metabolism in Physiology and Disease. Front. Vet. Sci. 2018, 5, 242. [Google Scholar] [CrossRef] [PubMed]
  13. Nitti, M.; Ivaldo, C.; Traverso, N.; Furfaro, A.L. Clinical Significance of Heme Oxygenase 1 in Tumor Progression. Antioxidants 2021, 10, 789. [Google Scholar] [CrossRef]
  14. Vijayan, V.; Wagener, F.A.D.T.G.; Immenschuh, S. The macrophage heme-heme oxygenase-1 system and its role in inflammation. Biochem. Pharmacol. 2018, 153, 159–167. [Google Scholar] [CrossRef] [PubMed]
  15. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. PLoS Med. 2021, 18, e1003583. [Google Scholar] [CrossRef] [PubMed]
  16. Estarreja, J.; Caldeira, G.; Silva, I.; Mendes, P.; Mateus, V. The Pharmacological Effect of Hemin in Inflammatory-Related Diseases: Protocol for a Systematic Review. JMIR Res. Protoc. 2023, 12, e48368. [Google Scholar] [CrossRef] [PubMed]
  17. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed]
  18. Wei, J.; Fan, G.; Zhao, H.; Li, J. Heme oxygenase-1 attenuates inflammation and oxidative damage in a rat model of smoke-induced emphysema. Int. J. Mol. Med. 2015, 36, 1384–1392. [Google Scholar] [CrossRef] [PubMed]
  19. Du, J.; Ren, W.; Zhang, Q.; Fu, N.; Han, F.; Cui, P.; Li, W.; Kong, L.; Zhao, S.; Wang, R.; et al. Heme Oxygenase-1 Suppresses Wnt Signaling Pathway in Nonalcoholic Steatohepatitis-Related Liver Fibrosis. BioMed Res. Int. 2020, 2020, 4910601. [Google Scholar] [CrossRef]
  20. Wang, R.-Q.; Mi, H.-M.; Li, H.; Zhao, S.-X.; Jia, Y.-H.; Nan, Y.-M. Modulation of IKKβ/NF-κB and TGF-β1/Smad via Fuzheng Huayu recipe involves in prevention of nutritional steatohepatitis and fibrosis in mice. Iran. J. Basic Med. Sci. 2015, 18, 404–411. [Google Scholar]
  21. Tang, J.; Li, L.; Li, C.-M.; Wu, J.; Sun, Y.; Wang, G.-L. Upregulation of HO-1 with Haemin Alleviates LPS-Stimulated Pro-inflammatory Responses Through Downregulation of p38 Signalling Pathways in Rat Liver. Scand. J. Immunol. 2015, 82, 443–451. [Google Scholar] [CrossRef] [PubMed]
  22. Li, L.; Tang, J.; Sun, Y.; Wu, J.; Yu, P.; Wang, G. Upregulation of HO-1 Attenuates LPS-Stimulated Proinflammatory Responses Through Downregulation of p38 Signaling Pathways in Rat Ovary. Inflammation 2015, 38, 1085–1092. [Google Scholar] [CrossRef] [PubMed]
  23. El-Ashmawy, N.E.; Al-Ashmawy, G.M.; Farag, A.A.; Ibrahim, A.O. Hemin versus erythropoietin: Possible role in Nrf2/HO-1 signaling pathway in rats with nephrotoxicity. Biomed. Pharmacother. 2022, 156, 113971. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, Z.; Zhu, W.; Ren, H.-Z.; Zhao, X.; Wang, S.; Ma, H.-C.; Shi, X.-L. Mesenchymal stem cells increase expression of heme oxygenase-1 leading to anti-inflammatory activity in treatment of acute liver failure. Stem Cell Res. Ther. 2017, 8, 70. [Google Scholar] [CrossRef] [PubMed]
  25. Heeba, G.H.; Hamza, A.A.; Hassanin, S.O. Induction of heme oxygenase-1 with hemin alleviates cisplatin-induced reproductive toxicity in male rats and enhances its cytotoxicity in prostate cancer cell line. Toxicol. Lett. 2016, 264, 38–50. [Google Scholar] [CrossRef] [PubMed]
  26. Ali, M.A.M.; Heeba, G.H.; El-Sheikh, A.A.K. Modulation of heme oxygenase-1 expression and activity affects streptozotocin-induced diabetic nephropathy in rats. Fundam. Clin. Pharmacol. 2017, 31, 546–557. [Google Scholar] [CrossRef] [PubMed]
  27. Lin, X.L.; Lv, J.J.; Lv, J.; Di, C.X.; Zhang, Y.J.; Zhou, T.; Liu, J.L.; Xia, Z.W. Heme oxygenase-1 directly binds STAT 3 to control the generation of pathogenic Th17 cells during neutrophilic airway inflammation. Allergy 2017, 72, 1972–1987. [Google Scholar] [CrossRef]
  28. Lin, X.; Lv, J.; Ge, D.; Bai, H.; Yang, Y.; Wu, J. Heme oxygenase-1 alleviates eosinophilic inflammation by inhibiting STAT3-SOCS3 signaling. Pediatr. Pulmonol. 2020, 55, 1440–1447. [Google Scholar] [CrossRef] [PubMed]
  29. Zhong, W.; Di, C.; Lv, J.; Zhang, Y.; Lin, X.; Yuan, Y.; Lv, J.; Xia, Z. Heme oxygenase-1 inhibits basophil maturation and activation but promotes its apoptosis in T helper type 2-mediated allergic airway inflammation. Immunology 2016, 147, 321–337. [Google Scholar] [CrossRef]
  30. Konrad, F.M.; Zwergel, C.; Ngamsri, K.-C.; Reutershan, J. Anti-inflammatory Effects of Heme Oxygenase-1 Depend on Adenosine A2A- and A2B-Receptor Signaling in Acute Pulmonary Inflammation. Front. Immunol. 2017, 8, 1874. [Google Scholar] [CrossRef]
  31. Konrad, F.M.; Knausberg, U.; Höne, R.; Ngamsri, K.-C.; Reutershan, J. Tissue heme oxygenase-1 exerts anti-inflammatory effects on LPS-induced pulmonary inflammation. Mucosal Immunol. 2016, 9, 98–111. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, F.; Sun, Y.-H.; Fan, K.-L.; Dong, X.-B.; Han, N.; Zhao, H.; Kong, L. Protective effects of heme oxygenase-1 against severe acute pancreatitis via inhibition of tumor necrosis factor-α and augmentation of interleukin-10. BMC Gastroenterol. 2017, 17, 100. [Google Scholar] [CrossRef] [PubMed]
  33. Fan, K.-L.; Li, M.-F.; Chen, Y.-G. Effects of heme oxygenase-1 on cytokines and histological changes of pancreas and liver in rats with severe acute pancreatitis. Int. J. Clin. Exp. Pathol. 2016, 9, 7156–7163. [Google Scholar]
  34. Iwata, M.; Inoue, T.; Asai, Y.; Hori, K.; Fujiwara, M.; Matsuo, S.; Tsuchida, W.; Suzuki, S. The protective role of localized nitric oxide production during inflammation may be mediated by the heme oxygenase-1/20carbon monoxide pathway. Biochem. Biophys. Rep. 2020, 23, 100790. [Google Scholar] [CrossRef]
  35. Wang, C.; Zhang, Y.; Han, L.; Guo, L.; Zhong, H.; Wang, J. Hemin ameliorates influenza pneumonia by attenuating lung injury and regulating the immune response. Int. J. Antimicrob. Agents 2017, 49, 45–52. [Google Scholar] [CrossRef] [PubMed]
  36. Chaves, H.V.V.; do Val, D.R.; Ribeiro, K.A.; Lemos, J.C.; Souza, R.B.; Gomes, F.I.F.; da Cunha, R.M.S.; Pinto, V.P.T.; Filho, G.C.; de Souza, M.H.L.P.; et al. Heme oxygenase-1/biliverdin/carbon monoxide pathway downregulates hypernociception in rats by a mechanism dependent on cGMP/ATP-sensitive K+ channels. Inflamm. Res. 2018, 67, 407–422. [Google Scholar] [CrossRef]
  37. Magierowski, M.; Magierowska, K.; Hubalewska-Mazgaj, M.; Sliwowski, Z.; Ginter, G.; Pajdo, R.; Chmura, A.; Kwiecien, S.; Brzozowski, T. Carbon monoxide released from its pharmacological donor, tricarbonyldichlororuthenium (II) dimer, accelerates the healing of pre-existing gastric ulcers. Br. J. Pharmacol. 2017, 174, 3654–3668. [Google Scholar] [CrossRef] [PubMed]
  38. Sun, G.; Lu, Y.; Zhao, L.; Xia, W.; Zhang, H.; Wang, L.; Zhang, L.; Wen, A. Hemin impairs resolution of inflammation via microRNA-144-3p-dependent downregulation of ALX/FPR2. Transfusion 2019, 59, 196–206. [Google Scholar] [CrossRef]
  39. Rossi, M.; Delbauve, S.; Roumeguère, T.; Wespes, E.; Leo, O.; Flamand, V.; Moine, A.L.; Hougardy, J.-M. HO-1 mitigates acute kidney injury and subsequent kidney-lung cross-talk. Free Radic. Res. 2019, 53, 1035–1043. [Google Scholar] [CrossRef]
  40. Wang, H.; Zhang, S.; Zhao, H.; Qin, H.; Zhang, J.; Dong, J.; Zhang, H.; Liu, X.; Zhao, Z.; Zhao, Y.; et al. Carbon Monoxide Inhibits the Expression of Proteins Associated with Intestinal Mucosal Pyroptosis in a Rat Model of Sepsis Induced by Cecal Ligation and Puncture. Med. Sci. Monit. 2020, 26, e920668. [Google Scholar] [CrossRef]
  41. Park, J.-S.; Choi, H.-S.; Yim, S.-Y.; Lee, S.-M. Heme Oxygenase-1 Protects the Liver from Septic Injury by Modulating TLR4-Mediated Mitochondrial Quality Control in Mice. Shock 2018, 50, 209–218. [Google Scholar] [CrossRef] [PubMed]
  42. Wedn, A.M.; El-Gowilly, S.M.; El-Mas, M.M. The α7-nAChR/heme oxygenase-1/carbon monoxide pathway mediates the nicotine counteraction of renal inflammation and vasoconstrictor hyporeactivity in endotoxic male rats. Inflamm. Res. 2020, 69, 217–231. [Google Scholar] [CrossRef] [PubMed]
  43. Khan, A.; Jamwal, S.; Bijjem, K.R.V.; Prakash, A.; Kumar, P. Neuroprotective effect of hemeoxygenase-1/glycogen synthase kinase-3β modulators in 3-nitropropionic acid-induced neurotoxicity in rats. Neuroscience 2015, 287, 66–77. [Google Scholar] [CrossRef]
  44. Wu, Y.; Wu, B.; Zhang, Z.; Lu, H.; Fan, C.; Qi, Q.; Gao, Y.; Li, H.; Feng, C.; Zuo, J.; et al. Heme protects intestinal mucosal barrier in DSS-induced colitis through regulating macrophage polarization in both HO-1-dependent and HO-1-independent way. FASEB J. 2020, 34, 8028–8043. [Google Scholar] [CrossRef]
  45. Aycan-Ustyol, E.; Kabasakal, M.; Bekpinar, S.; Alp-Yıldırım, F.I.; Tepe, O.; Giris, M.; Ozluk, Y.; Unlucerci, Y.; Uydes-Dogan, B.S.; Uysal, M. Vascular function and arginine and dimethylarginines in gentamicin-induced renal failure: A possible effect of heme oxygenase 1 inducer hemin. Can. J. Physiol. Pharmacol. 2017, 95, 1406–1413. [Google Scholar] [CrossRef]
  46. Refaie, M.M.M.; Rifaai, R.A.; Bayoumi, A.M.A.; Shehata, S. Cardioprotective effect of hemin in isoprenaline-induced myocardial infarction: Role of ATP-sensitive potassium channel and endothelial nitric oxide synthase. Fundam. Clin. Pharmacol. 2020, 34, 302–312. [Google Scholar] [CrossRef] [PubMed]
  47. Ali, F.F.; Abdelzaher, W.Y.; Ibrahim, R.A.; Elroby Ali, D.M. Amelioration of estrogen-induced endometrial hyperplasia in female rats by hemin via heme-oxygenase-1 expression, suppression of iNOS, p38 MAPK, and Ki67. Can. J. Physiol. Pharmacol. 2019, 97, 1159–1168. [Google Scholar] [CrossRef] [PubMed]
  48. Bayo Jimenez, M.T.; Frenis, K.; Kröller-Schön, S.; Kuntic, M.; Stamm, P.; Kvandová, M.; Oelze, M.; Li, H.; Steven, S.; Münzel, T.; et al. Noise-Induced Vascular Dysfunction, Oxidative Stress, and Inflammation Are Improved by Pharmacological Modulation of the NRF2/HO-1 Axis. Antioxidants 2021, 10, 625. [Google Scholar] [CrossRef] [PubMed]
  49. Dai, Y.; Chen, X.; Yu, J.; Cheng, X.; Ciao, Y.; Tang, F.; Li, Y.; Wan, S.; Su, W.; Liang, D. Hemin Promotes Corneal Allograft Survival Through the Suppression of Macrophage Recruitment and Activation. Investig. Opthalmology Vis. Sci. 2018, 59, 3952–3962. [Google Scholar] [CrossRef]
  50. Ndisang, J.F.; Tiwari, S. Featured Article: Induction of heme oxygenase with hemin improves pericardial adipocyte morphology and function in obese Zucker rats by enhancing proteins of regeneration. Exp. Biol. Med. 2015, 240, 45–57. [Google Scholar] [CrossRef]
  51. Cheng, X.; Yin, M.; Sun, X.; Zhang, Z.; Yao, X.; Liu, H.; Xia, H. Hemin attenuated lps-induced acute lung injury in mice via protecting pulmonary epithelial barrier and regulating ho-1/nlrp3-mediated pyroptosis. Shock 2023, 59, 744–753. [Google Scholar] [CrossRef] [PubMed]
  52. Chi, X.; Guo, N.; Yao, W.; Jin, Y.; Gao, W.; Cai, J.; Hei, Z. Induction of heme oxygenase-1 by hemin protects lung against orthotopic autologous liver transplantation-induced acute lung injury in rats. J. Transl. Med. 2016, 14, 35. [Google Scholar] [CrossRef] [PubMed]
  53. Aziz, N.M.; Kamel, M.Y.; Rifaai, R.A. Effects of hemin, a heme oxygenase-1 inducer in L-arginine-induced acute pancreatitis and associated lung injury in adult male albino rats. Endocr. Regul. 2017, 51, 20–30. [Google Scholar] [CrossRef] [PubMed]
  54. Chi, X.; Yao, W.; Xia, H.; Jin, Y.; Li, X.; Cai, J.; Hei, Z. Elevation of HO-1 Expression Mitigates Intestinal Ischemia-Reperfusion Injury and Restores Tight Junction Function in a Rat Liver Transplantation Model. Oxid. Med. Cell. Longev. 2015, 2015, 986075. [Google Scholar] [CrossRef] [PubMed]
  55. Xiaoyu, H.; Si, H.; Li, S.; Wang, W.; Guo, J.; Li, Y.; Cao, Y.; Fu, Y.; Zhang, N. Induction of heme oxygenas-1 attenuates NLRP3 inflammasome activation in lipopolysaccharide-induced mastitis in mice. Int. Immunopharmacol. 2017, 52, 185–190. [Google Scholar] [CrossRef]
  56. Sun, B.; Li, G.; Guo, L.; Yin, N.; Huang, H.; Wu, X.; Huang, R.; Feng, M. Once-monthly hemin suppresses inflammatory and autoreactive CD4+ T cell responses to robustly ameliorate experimental rheumatoid arthritis. iScience 2021, 24, 103101. [Google Scholar] [CrossRef]
  57. Rofaeil, R.R.; Welson, N.N.; Fawzy, M.A.; Ahmed, A.F.; Atta, M.; Bahaa El-deen, M.A.; Abdelzaher, W.Y. The IL-6/HO-1/STAT3 signaling pathway is implicated in the amelioration of acetaminophen-induced hepatic toxicity: A neonatal rat model. Hum. Exp. Toxicol. 2023, 42, 9603271231151376. [Google Scholar] [CrossRef]
  58. Abdel-Raheem, I.T.; Omran, G.A.E.-H. Induction of heme oxygenase-1 attenuates chemotherapy-induced pulmonary toxicity in rats: A possible link between heme oxygenase-1 and NF-κB. Pak. J. Pharm. Sci. 2016, 29, 685–694. [Google Scholar] [PubMed]
  59. Ni, L.; Wang, Z.; Yang, G.; Li, T.; Liu, X.; Liu, C. Heme oxygenase-1 alleviates cigarette smoke-induced restenosis after vascular angioplasty by attenuating inflammation in rat model. Toxicol. Lett. 2016, 245, 99–105. [Google Scholar] [CrossRef]
  60. Guo, S.; Yu, M.; Fang, Q.; Zhang, L.; You, C.; Wang, X.; Liu, Y.; Han, C. Heme oxygenase-1 induction mitigates burn-associated early acute kidney injury via the TLR4 signaling pathway. Burns 2022, 48, 156–167. [Google Scholar] [CrossRef]
  61. Yu, X.; Han, W.; Wang, C.; Sui, D.; Bian, J.; Bo, L.; Deng, X. Upregulation of Heme Oxygenase-1 by Hemin Alleviates Sepsis-Induced Muscle Wasting in Mice. Oxid. Med. Cell. Longev. 2018, 2018, 8927104. [Google Scholar] [CrossRef] [PubMed]
  62. Ahanger, A.A.; Prawez, S.; Shakoor, A.; Ahmad, W.; Khan, A.M.; Kumar, D. Topical application of ‘Hemin’ promotes wound healing in Streptozotocin-induced diabetic rats. Vet. Arh. 2021, 91, 287–296. [Google Scholar] [CrossRef]
  63. Kumar, D.; Jena, G.T.; Ram, M.; Lingaraju, M.; Cholenahalli Singh, V.; Prasad, R.; Kumawat, S.; Kant, V.; Gupta, P.; Tandan, S.K.; et al. Hemin attenuated oxidative stress and inflammation to improve wound healing in diabetic rats. Naunyn. Schmiedebergs. Arch. Pharmacol. 2019, 392, 1435–1445. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, Q.-Y.; Wang, G.-G.; Li, W.; Jiang, Y.-X.; Lu, X.-H.; Zhou, P.-P. Heme Oxygenase-1 Promotes Delayed Wound Healing in Diabetic Rats. J. Diabetes Res. 2016, 2016, 9726503. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, L.; Zhang, Y.; Zhong, W.; Di, C.; Lin, X.; Xia, Z. Heme Oxygenase-1 Ameliorates Dextran Sulfate Sodium-induced Acute Murine Colitis by Regulating Th17/Treg Cell Balance. J. Biol. Chem. 2014, 289, 26847–26858. [Google Scholar] [CrossRef] [PubMed]
  66. Fei, D.; Meng, X.; Zhao, M.; Kang, K.; Tan, G.; Pan, S.; Luo, Y.; Liu, W.; Nan, C.; Jiang, H.; et al. Enhanced induction of heme oxygenase-1 suppresses thrombus formation and affects the protein C system in sepsis. Transl. Res. 2012, 159, 99–109. [Google Scholar] [CrossRef] [PubMed]
  67. Bortscher, S.; Chang, J.; Vilz, T.O.; Schäfer, N.; Sommer, N.; Wehner, S.; Kalff, J.C.; Overhaus, M. Hemin induction of HO-1 protects against LPS-induced septic ileus. J. Surg. Res. 2012, 178, 866–873. [Google Scholar] [CrossRef] [PubMed]
  68. Devesa, I.; Ferrándiz, M.L.; Busserolles, J.; Alcaraz, M.J. Effects of heme oxygenase-1 inducers on established rat adjuvant arthritis. Cell. Mol. Biol. 2005, 51, 479–485. [Google Scholar] [PubMed]
  69. Zhang, J.; Jiang, Y.; Li, H.; Wang, J.; Li, C.; Zhang, D. Elevation of HO-1 expression protects the intestinal mucosal barrier in severe acute pancreatitis via inhibition of the MLCK/p-MLC signaling pathway. Exp. Cell Res. 2023, 424, 113508. [Google Scholar] [CrossRef]
  70. Zhang, Y.; Zhang, L.; Wu, J.; Di, C.; Xia, Z. Heme Oxygenase-1 Exerts a Protective Role in Ovalbumin-induced Neutrophilic Airway Inflammation by Inhibiting Th17 Cell-mediated Immune Response. J. Biol. Chem. 2013, 288, 34612–34626. [Google Scholar] [CrossRef]
  71. Placha, D.; Jampilek, J. Chronic Inflammatory Diseases, Anti-Inflammatory Agents and Their Delivery Nanosystems. Pharmaceutics 2021, 13, 64. [Google Scholar] [CrossRef] [PubMed]
  72. Desnick, R.J.; Balwani, M.; Anderson, K.E. Inherited Porphyrias. In Emery and Rimoin’s Principles and Practice of Medical Genetics; Elsevier: Amsterdam, The Netherlands, 2013; pp. 1–32. [Google Scholar]
  73. Connick, J.P.; Reed, J.R.; Cawley, G.F.; Backes, W.L. Heme oxygenase-1 affects cytochrome P450 function through the formation of heteromeric complexes: Interactions between CYP1A2 and heme oxygenase-1. J. Biol. Chem. 2021, 296, 100030. [Google Scholar] [CrossRef] [PubMed]
  74. Basetty, V.; Deruiter, J.; Pathak, S.; Dua, K.; Dhanasekaran, M. Advanced drug delivery systems targeting to improve therapeutic outcomes in porphyria. In Drug Delivery Systems for Metabolic Disorders; Elsevier: Amsterdam, The Netherlands, 2022; pp. 65–76. [Google Scholar]
  75. Al Shoyaib, A.; Archie, S.R.; Karamyan, V.T. Intraperitoneal Route of Drug Administration: Should it Be Used in Experimental Animal Studies? Pharm. Res. 2020, 37, 12. [Google Scholar] [CrossRef] [PubMed]
  76. Turner, P.V.; Brabb, T.; Pekow, C.; Vasbinder, M.A. Administration of substances to laboratory animals: Routes of administration and factors to consider. J. Am. Assoc. Lab. Anim. Sci. 2011, 50, 600–613. [Google Scholar] [PubMed]
  77. Pritchett-Corning, K.R.; Hashway, S.; Suckow, M.A. The Laboratory Mouse, 3rd ed.; CRC Press: Boca Raton, FL, USA, 2023; pp. 157–208. [Google Scholar] [CrossRef]
  78. Banday, M.Z.; Sameer, A.S.; Nissar, S. Pathophysiology of diabetes: An overview. Avicenna J. Med. 2020, 10, 174–188. [Google Scholar] [CrossRef] [PubMed]
  79. American Diabetes Association. Diagnosis and Classification of Diabetes Mellitus. Diabetes Care 2014, 37, S81–S90. [Google Scholar] [CrossRef] [PubMed]
  80. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef] [PubMed]
  81. Matthay, M.A.; Ware, L.B.; Zimmerman, G.A. The acute respiratory distress syndrome. J. Clin. Investig. 2012, 122, 2731–2740. [Google Scholar] [CrossRef]
  82. Silva, I.; Solas, J.; Pinto, R.; Mateus, V. Chronic Experimental Model of TNBS-Induced Colitis to Study Inflammatory Bowel Disease. Int. J. Mol. Sci. 2022, 23, 4739. [Google Scholar] [CrossRef]
  83. Seymour, C.W.; Liu, V.X.; Iwashyna, T.J.; Brunkhorst, F.M.; Rea, T.D.; Scherag, A.; Rubenfeld, G.; Kahn, J.M.; Shankar-Hari, M.; Singer, M.; et al. Assessment of Clinical Criteria for Sepsis. JAMA 2016, 315, 762. [Google Scholar] [CrossRef]
  84. Bryda, E.C. The Mighty Mouse: The impact of rodents on advances in biomedical research. Mo. Med. 2013, 110, 207–211. [Google Scholar] [PubMed]
  85. Voelkl, B.; Vogt, L.; Sena, E.S.; Würbel, H. Reproducibility of preclinical animal research improves with heterogeneity of study samples. PLoS Biol. 2018, 16, e2003693. [Google Scholar] [CrossRef]
  86. Silva, I.; Alípio, C.; Pinto, R.; Mateus, V. Potential anti-inflammatory effect of erythropoietin in non-clinical studies in vivo: A systematic review. Biomed. Pharmacother. 2021, 139, 111558. [Google Scholar] [CrossRef]
  87. McGonigle, P.; Ruggeri, B. Animal models of human disease: Challenges in enabling translation. Biochem. Pharmacol. 2014, 87, 162–171. [Google Scholar] [CrossRef] [PubMed]
  88. Waterson, R.H.; Lindblad-Toh, K.; Birney, E.; Rogers, J.; Abril, J.F.; Agarwala, P.; Agarwala, R.; Ainscough, R.; Alexandersson, M.; An, P.; et al. Initial sequencing and comparative analysis of the mouse genome. Nature 2002, 420, 520–562. [Google Scholar] [CrossRef]
  89. Dillmann, W.H. The rat as a model for cardiovascular disease. Drug Discov. Today Dis. Model. 2008, 5, 173–178. [Google Scholar] [CrossRef]
  90. Dietrich, M.R.; Ankeny, R.A.; Crowe, N.; Green, S.; Leonelli, S. How to choose your research organism. Stud. Hist. Philos. Sci. Part C Stud. Hist. Philos. Biol. Biomed. Sci. 2020, 80, 101227. [Google Scholar] [CrossRef]
  91. How to Choose the Right Mouse Models and Genetic Modification Method for Your Research. Available online: https://apac.cyagen.com/community/technical-bulletin/c57bl6-and-balbc-mouse.htm (accessed on 19 December 2023).
  92. C57BL/6 Mice. Available online: https://www.criver.com/products-services/find-model/c57bl6-mouse?region=3616 (accessed on 19 December 2023).
  93. Garg, R.; Heinzle, E.; Noor, F. Hepatocytes of Wistar and Sprague Dawley rats differ significantly in their central metabolism. J. Cell. Biochem. 2018, 119, 909–917. [Google Scholar] [CrossRef] [PubMed]
  94. Andrade, A.; Pinto, S.C.; de Oliveira, R.S. Animais de Laboratório: Criação e Experimentação; Editora FIOCRUZ: Rio de Janeiro, Brazil, 2006. [Google Scholar] [CrossRef]
  95. Hall, O.J.; Klein, S.L. Progesterone-based compounds affect immune responses and susceptibility to infections at diverse mucosal sites. Mucosal Immunol. 2017, 10, 1097–1107. [Google Scholar] [CrossRef]
  96. Fedotcheva, T.A.; Fedotcheva, N.I.; Shimanovsky, N.L. Progesterone as an Anti-Inflammatory Drug and Immunomodulator: New Aspects in Hormonal Regulation of the Inflammation. Biomolecules 2022, 12, 1299. [Google Scholar] [CrossRef]
  97. Adams, D.B.; Cowan, C.W.; Marshall, M.E.; Stark, J. Competitive and territorial fighting: Two types of offense in the rat. Physiol. Behav. 1994, 55, 247–254. [Google Scholar] [CrossRef] [PubMed]
  98. Jackson, S.J.; Andrews, N.; Ball, D.; Bellantuono, I.; Gray, J.; Hachoumi, L.; Holmes, A.; Latcham, J.; Petrie, A.; Potter, P.; et al. Does age matter? The impact of rodent age on study outcomes. Lab. Anim. 2017, 51, 160–169. [Google Scholar] [CrossRef] [PubMed]
  99. Ghia, P.; ten Boekel, E.; Rolink, A.G.; Melchers, F. B-cell development: A comparison between mouse and man. Immunol. Today. 1998, 19, 480–485. [Google Scholar] [CrossRef] [PubMed]
  100. Holladay, S.D.; Smialowicz, R.J. Development of the murine and human immune system: Differential effects of immunotoxicants depend on time of exposure. Environ. Health Perspect. 2000, 108, 463–473. [Google Scholar] [CrossRef] [PubMed]
  101. Kincade, P.W. Formation of B Lymphocytes in Fetal and Adult Life. Adv. Imm. 1981, 31, 177–245. [Google Scholar] [CrossRef]
  102. Liu, F.; McCullough, L.D. Interactions between age, sex, and hormones in experimental ischemic stroke. Neurochem. Int. 2012, 61, 1255–1265. [Google Scholar] [CrossRef] [PubMed]
  103. Gomez, C.R.; Hirano, S.; Cutro, B.T.; Birjandi, S.; Baila, H.; Nomellini, V.; Kovacs, E.J. Advanced age exacerbates the pulmonary inflammatory response after lipopolysaccharide exposure. Crit. Care Med. 2007, 35, 246–251. [Google Scholar] [CrossRef] [PubMed]
  104. Jia, T.; Wang, C.; Han, Z.; Wang, X.; Ding, M.; Wang, Q. Experimental Rodent Models of Cardiovascular Diseases. Front. Cardiovasc. Med. 2020, 7, 588075. [Google Scholar] [CrossRef] [PubMed]
  105. Bodaghi, A.; Fattahi, N.; Ramazani, A. Biomarkers: Promising and valuable tools towards diagnosis, prognosis and treatment of COVID-19 and other diseases. Heliyon 2023, 9, e13323. [Google Scholar] [CrossRef]
  106. Sauer, J.-M.; Porter, A.C. Preclinical biomarker qualification. Exp. Biol. Med. 2018, 243, 222–227. [Google Scholar] [CrossRef]
  107. Khansari, N.; Shakiba, Y.; Mahmoudi, M. Chronic Inflammation and Oxidative Stress as a Major Cause of Age- Related Diseases and Cancer. Recent Pat. Inflamm. Allergy Drug Discov. 2009, 3, 73–80. [Google Scholar] [CrossRef] [PubMed]
  108. Ryter, S.W. Heme Oxygenase-1: An Anti-Inflammatory Effector in Cardiovascular, Lung, and Related Metabolic Disorders. Antioxidants 2022, 11, 555. [Google Scholar] [CrossRef] [PubMed]
  109. Gamage, S.M.K.; Cheng, T.; Lee, K.T.-W.; Dissabandara, L.; Lam, A.K.-Y.; Gopalan, V. Hemin, a major heme molecule, induced cellular and genetic alterations in normal colonic and colon cancer cells. Pathol. Res. Pract. 2021, 224, 153530. [Google Scholar] [CrossRef] [PubMed]
  110. Owen, J.E.; Bishop, G.M.; Robinson, S.R. Uptake and Toxicity of Hemin and Iron in Cultured Mouse Astrocytes. Neurochem Res. 2016, 41, 298–306. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structure of hemin (adapted from Tahoun et al., 2023) [6].
Figure 1. Structure of hemin (adapted from Tahoun et al., 2023) [6].
Biomedicines 12 00898 g001
Figure 2. PRISMA flow diagram representing the selection process.
Figure 2. PRISMA flow diagram representing the selection process.
Biomedicines 12 00898 g002
Table 1. Qualitative synthesis of the included studies.
Table 1. Qualitative synthesis of the included studies.
Hemin-Related ParametersAnimal-Related ParametersBiomarkers AssessedReference
DoseFrequencyRoute of AdministrationDurationDisease Animal ModelSpecieStrainGenderAge
20 µmol/kg3× weekSC20 weeksEmphysemaRatWistarMale6 weeksIL-8, IL-10, IL-17, Macrophages, MCP-1, MDA, MIP-2α, neutrophils, SOD, GSH, TNF-α[18]
30 µmol/kgIP8 weeksLiver fibrosisMiceC57BL/6NDHO-1, NFAT5, ROS, TGF-β1[19]
Steatohepatitis and fibrosis8 weeksHO-1, MCP-1, NF-kB, TGF-β1[20]
Single dose-LPS-stimulated inflammatory responseRatSprague Dawley10–11 weeksHO-1, IL-1β, IL-6, LDH, MDA, SOD[21]
WistarFemale[22]
40 µmol/kgDaily8 daysNephrotoxicityAlbinoMale Adult GSH, HO-1, IL-6, MDA [23]
Single dose- Acute liver failureSprague Dawley6–7 weeksHO-1, IL-1β, IL-6, MDA, MPO, NF-kB p65, Nrf2, TNF-α[24]
SCReproductive toxicityWistarAdultCAT, HO-1, GSH, iNOS, MDA, NO, TNF-α[25]
ND30 daysDiabetesNDCAT, GSH, HO-1, MDA, MPO, SOD, TNF-α[26]
75 µmol/kgDailyIP2 daysNeutrophilic airway inflammationMiceBALB/c and DO11.10 transgenicFemale8–10 weeksHO-1, IL-6, IL-10, IL-17A, JAK1, JAK2, STAT3, T-cells[27]
Variable (days −2, −1, 12, 13, 21, 22, and 25)27 daysEosinophilic asthmaBALB/cNDNDGATA3, HO-1, IL-4, IL-5, IL-17A, IL-17F, eosinophils, lymphocytes, macrophages, neutrophils, RORγt, SOCS3, STAT3[28]
27 daysAllergic airway inflammationBALB/c, DO11.10, and Bas-TRECK transgenic6–8 weeksBasophils, eosinophils, IL-4, T-cells[29]
80 µmol/kgSingle dose- Acute pulmonary inflammationCD1 (WT and A2A gene-deficient) and C57BL/6 (WT and A2B gene-deficient)Male8–12 weeksHO-1, IL-6, TNF-α[30]
8 × 104 µmol/kgAcute lung injuryC57BL/6 (WT and HO-1flox/flox)HO-1, IL-6, NF-kB p65, NF-kB p52, TNF-α[31]
7.5 × 10−2 mg/kgAcute pancreatitisRatSprague DawleyNDHO-1, IL-10, TNF-α[32]
6–7 weeks[33]
0.1, 0.3, or 1.0 mg/kgPIPleurisyWistarNDCAT, GPx, GST, IL-1β, iNOS, MCP-1, MPO, NOx, SOD, TNF-α[34]
0.1, 0.3, 1, or 3 mg/kgDailyIV3 daysInfluenza pneumoniaMiceBALB/cFemale5–6 weeksIFN-γ, IL-6, IL-10, HO-1, MCP-1, TNF-α[35]
1, 3, or 10 mg/kgSingle doseSC-Temporomandibular joint arthritisRat WistarMaleNDHO-1, IL-1β, MPO, TNF-α, White blood cells[36]
5 mg/kgDailyIG9 daysChronic gastric ulcers8–9 weeksCOX-2, HO-1, HO-2, IL-1β, iNOS, Nrf2, TNF-α[37]
Single doseIP-PeritonitisMiceC57BL/66–8 weeksIL-6, Leukocytes, macrophages, polymorphonuclear cells, TNF-α[38]
Renal ischemia–reperfusion injuryND8–12 weeksHO-1, IL-1β, IL-6, Neutrophils, nitrotyrosine, peroxidase, TNF-α[39]
5 or 10 mg/kgDaily14 daysExperimental colitisCD-1Female6 weeksIL-10, TNF-α[8]
4 daysMale6–10 weeksIL-1β, IL-10, MPO, TNF-α[7]
8 mg/kgSingle dose-SepsisRatSprague Dawley8–10 weeksCaspase-1, Caspase-11, GSDMD, HMGB1, IL-1β, IL-18, TNF-α[40]
10 mg/kgSepsisMiceC57BL/6NDGDH, HO-1, IL-1β, IL-6, LDH, MDA, SOD2, TLR4[41]
NDRenal inflammation and vasoconstrictor dysfunctionRatWistar10–13 weeksHO-1, IL-1β, iNOS, NF-kB [42]
10 or 30 mg/kgDailyIP14 daysNeurotoxicityNDCAT, GSH, HO-1, IL-1β, TNF-α[43]
20 mg/kgEvery 2 days7 daysExperimental colitisMiceC57BL/6Female6–8 weeks4-HNE, IFN-γ, IL-1β, IL-6, IL-12, iNOS, Ly6C, macrophages, MDA, MPO, NO, NF-kB p-p65, STAT1, STAT6, TNF-α[44]
14 daysRenal failureRatWistarMaleND4-HNE, GSH, GSH-Px, HO-1, MPO, SOD[45]
25 mg/kgDaily5 daysMyocardial infarctionNDeNOS, GSH, HO-1, LDH, MDA, TNF-α[46]
3× weekNDEndometrial hyperplasiaFemale9–10 weeksGSH, IL-1β, iNOS, MDA, NOx, SOD[47]
Every 2 days6 daysVascular dysfunctionMiceC57BL/6Male 6–12 weeks3-nitrotyrosine, 4-HNE, eNOS, Hmox1, macrophages, HO-1, IL-6, ROS[48]
30 mg/kg30 daysPenetrating keratoplastyRatWistarFemale6–8 weeksHO-1, iNOS, macrophages, MCP-1, MIP-1α, TNF-α[49]
2× week8 weeksObesityZucker fatty and Zucker leanMale12 weeks8-isoprostane, HO-1, IL-1β, IL-6, macrophages, nitrotyrosine, TGF-β1/2/3, TNF-α[50]
Single dose-Acute lung injuryMiceBALB/cFemaleNDCaspase-1, IL-1β, MDA, MPO, HO-1, ROS[51]
RatSprague DawleyMaleHO-1, IL-6, MDA, MPO, NF-kB, Nrf2, SOD, TNF-α[52]
Acute pancreatitis16 weeksHO-1, MDA, NO, TNF-α[53]
Liver transplantationWistarNDHO-1, IL-6, MDA, NF-kB, Nrf2, SOD, TNF-α[54]
NDMastitisMiceBALB/cFemale8–12 weeksIL-1β, MPO, NLRP3, procaspase-1, ROS, TXNIP[55]
40 mg/kg2× weekIP14 daysRheumatoid arthritis Rat Sprague DawleyMale8–10 weeksIFN-γ, IL-1β, IL-6, T-cells, TNF-α[56]
40 or 90 mg/kg28 days
40 mg/kg + 120 mg/kg2 daily adm + 1 admIP + SC28 or 41 days
50 mg/kgDailyIP14 daysHepatic toxicityN.D.1 weekGSH, HO-1, IL-1β, IL-6, MDA, NOx, STAT3, TNF-α [57]
14 daysPulmonary toxicityWistarFemaleAdultGSH, HO-1, IL-6, LDH, MDA, MPO, NF-kB, NO, SOD[58]
Every 2 days21 daysRestenosisSprague DawleyMaleNDCD45, HO-1, IL-1β, IL-6, MCP-1, NF-kB, TNF-α [59]
Single dose-Major burn modelAdultHO-1, IL-1β, IL-6, MDA, NF-kB p65, TLR4, TNF-α[60]
Acute kidney injurySprague Dawley (WT and PINK1 knockout)8 weeksCaspase-1, HO-1, IL-1β, IL-6, MDA, NGAL, SOD, TNF-α[61]
SepsisMiceC57BL/6HO-1, IL-6, MDA, SOD, TNF-α
0.5%DailyTopical13 daysDiabetesRatWistarAdultIL-10, TNF-α[62]
2× day19 daysCAT, GPx, GSH, HO-1, IL-10, MDA, SOD, TNF-α[63]
10%N.D.21 daysSprague DawleyNDHO-1, IL-6, MDA, SOD, TNF-α[64]
4-HNE, 4-hydroxynonenal; CAT, catalase; COX-2, cyclooxygenase-2; eNOS, endothelial nitric oxide synthase; GATA3, GATA3 binding protein; GDH, glutamate dehydrogenase; GPx, glutathione peroxidase; GSDMD, gasdermin D; GSH, glutathione; GSH-Px, glutathione peroxidase; GST, glutathione S-transferase; HMGB1, high-mobility group box 1; Hmox1, heme oxygenase 1 gene; HO, heme oxygenase; IFN-γ, interferon-γ; IG, intragastric; IL, interleukin; iNOS, inducible nitric oxide synthase; IP, intraperitoneal; IV, intravenous; JAK, janus kinase; LDH, lactate dehydrogenase; LPS, lipopolysaccharides; Ly6C, lymphocyte antigen 6 family member C; MCP-1, monocyte chemoattractant protein-1; MDA, malondialdehyde; MIP, macrophage inflammatory protein; MPO, myeloperoxidase; ND, not defined; NFAT5, nuclear factor of activated T-cells 5; NF-kB, nuclear factor kappa B; NLRP3, NOD-like receptor pyrin domain-containing protein 3; NO, nitric oxide; NOx, total nitrites; Nrf2, Nuclear factor erythroid 2 related factor 2; PI, pleural injection; RORγt, retinoic-acid-receptor-related orphan nuclear receptor gamma; ROS, reactive oxygen species; SC, subcutaneous; SOCS3, suppressor of cytokine signaling 3; SOD, superoxide dismutase; STAT, signal transducer and activator of transcription; TGF-β, transforming growth factor-β; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor-α; TXNIP, thioredoxin-interacting protein.
Table 2. Risk of bias assessment in the included studies.
Table 2. Risk of bias assessment in the included studies.
Final GradeReferences
High
(15–20) points
[21,22,32,37,46]
Moderate
(8–14) points
[7,8,18,19,20,23,24,25,26,27,28,29,30,31,33,34,35,36,38,39,40,41,42,43,44,45,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Estarreja, J.; Caldeira, G.; Silva, I.; Mendes, P.; Mateus, V. The Pharmacological Effect of Hemin in Inflammatory-Related Diseases: A Systematic Review. Biomedicines 2024, 12, 898. https://doi.org/10.3390/biomedicines12040898

AMA Style

Estarreja J, Caldeira G, Silva I, Mendes P, Mateus V. The Pharmacological Effect of Hemin in Inflammatory-Related Diseases: A Systematic Review. Biomedicines. 2024; 12(4):898. https://doi.org/10.3390/biomedicines12040898

Chicago/Turabian Style

Estarreja, João, Gonçalo Caldeira, Inês Silva, Priscila Mendes, and Vanessa Mateus. 2024. "The Pharmacological Effect of Hemin in Inflammatory-Related Diseases: A Systematic Review" Biomedicines 12, no. 4: 898. https://doi.org/10.3390/biomedicines12040898

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop