Next Article in Journal
Association Study of CACNA1D, KCNJ11, KCNQ1, and CACNA1E Single-Nucleotide Polymorphisms with Type 2 Diabetes Mellitus
Previous Article in Journal
The Contribution of Mast Cells to the Regulation of Elastic Fiber Tensometry in the Skin Dermis of Children with Marfan Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Heme Oxygenase-1 and Prostate Cancer: Function, Regulation, and Implication in Cancer Therapy

1
Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
2
Department of Clinical, Diagnostic, and Therapeutic Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
3
Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(17), 9195; https://doi.org/10.3390/ijms25179195 (registering DOI)
Submission received: 13 July 2024 / Revised: 15 August 2024 / Accepted: 23 August 2024 / Published: 24 August 2024
(This article belongs to the Section Molecular Oncology)

Abstract

:
Prostate cancer (PC) is a significant cause of mortality in men worldwide, hence the need for a comprehensive understanding of the molecular mechanisms underlying its progression and resistance to treatment. Heme oxygenase-1 (HO-1), an inducible enzyme involved in heme catabolism, has emerged as a critical player in cancer biology, including PC. This review explores the multifaceted role of HO-1 in PC, encompassing its function, regulation, and implications in cancer therapy. HO-1 influences cell proliferation, anti-apoptotic pathways, angiogenesis, and the tumor microenvironment, thereby influencing tumor growth and metastasis. HO-1 has also been associated with therapy resistance, affecting response to standard treatments. Moreover, HO-1 plays a significant role in immune modulation, affecting the tumor immune microenvironment and potentially influencing therapy outcomes. Understanding the intricate balance of HO-1 in PC is vital for developing effective therapeutic strategies. This review further explores the potential of targeting HO-1 as a therapeutic approach, highlighting challenges and opportunities. Additionally, clinical implications are discussed, focusing on the prognostic value of HO-1 expression and the development of novel combined therapies to augment PC sensitivity to standard treatment strategies. Ultimately, unraveling the complexities of HO-1 in PC biology will provide critical insights into personalized treatment approaches for PC patients.

1. Introduction to PC

PC is the leading cause of cancer-related deaths among men worldwide [1]. The incidence of PC varies across populations, with higher rates reported in North America, Europe, and Australia, while lower rates are observed in Asian countries [2,3]. PC poses significant challenges due to its heterogeneity and variable clinical outcomes despite advancements in screening techniques and treatment options [4].
The prostate gland is a small, walnut-shaped organ located just below the bladder and in front of the rectum in males. The primary function of the prostate is to provide nutrients to sperm [5]. Oxidative stress and inflammation within the prostate microenvironment may lead to increased cell growth, known as benign prostate hyperplasia (BPH), or genetic and epigenetic alterations in prostate epithelial cells that can generate carcinomas [6].
Several risk factors have been identified to influence PC development, including age, ethnicity, family history, and genetics [2,7]. The likelihood of developing PC increases with age, with most cases being diagnosed in men over 65 [7,8]. Ethnicity also plays a role, with higher incidence rates observed in certain populations [9]. Family history and genetic mutations further elevate the risk [9]. Additional modifiable factors can include diet, physical activity, smoking, and specific medications [10].
Prostate-specific antigen (PSA) is an enzyme that is primarily found in semen, where its primary function is to liquify semen, facilitating sperm mobility [5]. Although predominantly present in semen, a small amount of PSA circulates in the blood. The measurement of PSA levels in the blood is commonly used as a screening tool for PC [11] (Figure 1). Elevated PSA levels can indicate the presence of PC, although high levels can also result from other prostate conditions, such as BPH or prostatitis, which is an inflammation of the prostate [11,12]. PSA screening has been crucial for early detection, leading to an increase in the diagnosis of localized disease [13].
The Gleason score, which was first described by Gleason [14], is a critical tool used in evaluating the prognosis of PC. It grades cancer based on its microscopic appearance and is a strong predictor of disease aggressiveness [15] (Figure 1). Specifically, the Gleason score is the combination of the cellular grade of the most common and second most common histologic appearances within the malignancy. Cellular grade in PC ranges from 3 to 5 with Gleason score thus ranging from 6–10; with higher scores indicating more aggressive cancer and a poorer prognosis [16]. This scoring system helps guide treatment decisions and provides insights into the potential progression of the disease.
Current treatment strategies for PC are primarily determined by the patient’s risk group, which is assessed based on tumor stage, Gleason score, and PSA levels [17]. For localized disease, treatment options include radical prostatectomy and radiation therapy [17,18]. Androgen deprivation therapy (ADT), targeting the androgen receptor (AR) signaling pathway, is the mainstay of treatment for advanced PC. While initially effective, most patients eventually develop resistance to ADT, leading to the development of castration-resistant PC (CRPC) [19]. Therapeutic approaches for CRPC include novel AR-targeted agents, chemotherapy, immunotherapy, and radiopharmaceuticals [20] (Figure 1).
Despite the high 5-year survival rate of early-diagnosed PC, 97%, there remains a critical need to develop more effective therapeutic strategies due to several factors [1]. Firstly, advanced and metastatic PC has a significantly worse prognosis compared to early-stage disease, 32%, with current therapies often proving less effective [21]. Additionally, many patients with advanced PC develop resistance to standard treatments, including ADT, chemotherapy, and radiation, necessitating new approaches to overcome this resistance. Furthermore, existing treatments can have significant side effects that impact patients’ quality of life, highlighting the need for therapies that are both more effective and less damaging [22]. Moreover, even after successful initial treatment, there is a risk of recurrence, which underscores the importance of developing strategies to prevent relapse [23,24]. Finally, enhancing early detection methods to better distinguish between aggressive and indolent forms of PC can help ensure prompt and appropriate treatment [25,26]. These advancements are crucial for improving long-term outcomes and the overall well-being of patients.
To address these challenges, it is imperative to unravel the complex molecular and cellular pathways involved in PC development and progression. This review aims to shed light on a critical molecular effector, HO-1, a key enzyme in the heme degradation pathway that has emerged as a driver in PC progression and therapy resistance. We will elucidate the intricate interplay of genetic and epigenetic alterations, signaling pathways, tumor microenvironment, and therapy-induced changes that shape PC biology. By examining the existing literature and recent advancements, we hope to contribute to a deeper understanding of the molecular complexity of PC and identify potential avenues for developing more effective treatment strategies.

2. Introduction to HO-1

HO-1 stands as a pivotal and dynamic player in cellular stress response and therapy resistance, captivating the attention of researchers across various disciplines [27]. HO-1 is a member of the heme oxygenase family and is primarily localized in the endoplasmic reticulum, mitochondria, and plasma membrane [27,28] (Figure 2). Unlike its constitutively expressed counterpart, heme oxygenase-2 (HO-2), HO-1 is an inducible enzyme. Additionally, a third isoform, HO-3, has been identified in rats; however, it lacks catalytic activity [29]. Various cells within the prostate tumor microenvironment and immune cells express HO-1, such as macrophages, T cells, and dendritic cells [30,31,32]. HO-1 is induced in response to various stressors, including oxidative stress, inflammation, heavy metals, certain cytokines, and hypoxia [33]. These stimuli trigger the activation of specific signaling pathways and transcription factors, leading to the upregulation of HO-1 expression as a protective measure to combat cellular stress.
Central to the function of HO-1 is its role in the heme degradation pathway, which is critical for cellular homeostasis (Figure 2). Heme, an iron-protoporphyrin IX complex, is an essential component in various biological processes, including oxygen storage and transfer via myoglobin and hemoglobin, electron transfer through cytochromes, and signal transduction via heme-regulated inhibitors [34,35]. However, free heme is highly toxic and can contribute to the generation of harmful ROS through the Fenton reaction, necessitating its breakdown by heme oxygenases [34,35]. HO-1 enzymatically degrades heme, producing equimolar amounts of carbon monoxide (CO), biliverdin, and free iron (Fe2+) (Figure 2).
CO, a well-known gaseous signaling molecule, demonstrates various effects in cellular physiology, including vasodilation, anti-inflammatory actions, and modulation of apoptosis and cell proliferation [36]. Biliverdin, in turn, is rapidly converted into bilirubin, a potent antioxidant molecule known for its protective role against oxidative damage [37]. Finally, the release of free iron presents both challenges and opportunities. In contrast to bilirubin, free iron can fuel the Fenton reaction, leading to harmful reactive oxygen species (ROS) generation; it also plays a crucial role in essential cellular processes [38]. The dynamic interplay of these bioactive molecules governs diverse cellular responses to stress (Figure 2). Different studies show that CO can trigger upregulation of HO-1 by upregulating Nrf2-dependent HO-1 expression via PERK activation [39]. Furthermore, studies indicate that HO exhibits a higher affinity for binding O2 compared to other heme proteins, enabling it to effectively differentiate between CO and O2 [40]. It has been shown that the active site of HO is specifically adapted to strongly favor O2 binding over CO [40,41,42].
Emerging evidence suggests that HO-1 can translocate to the nucleus under certain stress conditions, where it may regulate gene expression [43,44] (Figure 2). Normally, HO-1 is anchored to the lumen of the smooth endoplasmic reticulum (sER) by its C-terminus. However, under stress, proteolytic cleavage leads to the truncation of the C-terminus, releasing a significant HO-1 fragment containing the N-terminus into the cytosol [45]. This fragment can migrate to the nucleus in response to specific signals, with the “nuclear shuttling sequence (NSS)” playing a critical role in facilitating HO-1’s nuclear import [45]. Once in the nucleus, HO-1 interacts with nuclear proteins that regulate transcription factors such as AP1, STAT1, STAT3, among others [46,47]. Additionally, HO-1 functions as a DNA-binding protein at specific DNA binding motifs, with three potential DNA-binding domains identified within its structure [48]. Through its interaction with DNA, HO-1 may regulate the expression of various pro-tumorigenic genes, promoting cancer cell survival, proliferation, and metastasis, independent of its enzymatic activity (Figure 2).
Within the nucleus, HO-1 interact directly with components of the DNA damage response (DDR) pathway [49,50]. Given the critical role of the DDR in maintaining genomic stability, HO-1’s involvement suggests it may help cancer cells withstand genotoxic stress, potentially leading to resistance against treatments like chemotherapy and radiation, which rely on inducing DNA damage to kill cancer cells. Additionally, studies suggest that HO-1 might interact with or influence the stability of G-quadruplexes, which are non-canonical nucleic acid structures formed in guanine-rich regions of DNA and RNA [50]. These structures play key roles in the regulation of gene expression, DNA replication, and genome stability. By stabilizing or destabilizing G-quadruplexes, nuclear HO-1 might modulate the transcription of genes involved in cell proliferation and survival, thereby influencing cancer growth. This nuclear activity of HO-1 adds another layer to its role in cancer biology, suggesting that its function extends beyond cryoprotection and into the modulation of tumor-promoting pathways. The overexpression of the nuclear HO-1 has been shown to correlate with PC aggressiveness and metastasis [32].
The induction of HO-1 reflects a cellular strategy aimed at diminishing oxidative stress and inflammation, which are pivotal factors in numerous pathological conditions, including cancer. Moreover, the broad functions of HO-1 extend beyond cellular protection, encompassing immunomodulation, tissue repair, and angiogenesis, further emphasizing the significance of this enzyme in PC development.

3. Regulation of HO-1 in PC

HO-1 has been shown to be induced in multiple cancers, including PC [51,52,53,54,55], and its elevated expression is closely associated with poor patient prognosis [44]. Regulating HO-1 expression is essential for maintaining cellular homeostasis, and dysregulation of this process has been implicated in various diseases, including cancer. In PC, the expression of HO-1 is intricately regulated by numerous signaling pathways, transcription factors, and external stimuli that impact HO-1 induction in PC cells (Figure 3).
One significant pathway involved in HO-1 induction is the nuclear factor erythroid 2-related factor 2 (NRF2) pathway, which plays the main role in inducing HO-1 expression in response to oxidative stress [56,57] (Figure 3). Under normal conditions, NRF2 is sequestered in the cytoplasm through its association with Kelch-like ECH-associated protein 1 (KEAP1). Upon exposure to oxidative stress, NRF2 dissociates from KEAP1, translocates to the nucleus, and binds to antioxidant response elements (AREs) within the HO-1 promoter region, leading to its transcriptional activation [56,58] (Figure 3). However, an alternative “hinge and latch” model has been proposed, wherein NRF2 is not merely released from KEAP1 but rather, its nuclear translocation requires de novo synthesis of NRF2 [59,60]. This model posits that even under oxidative stress, some KEAP1-bound NRF2 is degraded, and the newly synthesized NRF2 is what accumulates in the nucleus to drive the expression of target genes [59,60]. Additionally, BTB and CNC homology 1 (BACH1), a heme sensor, acts as a major transcription repressor of HO-1 by competing with NRF2 for ARE binding under normal conditions. Elevated heme levels cause BACH1 to dissociate from AREs, permitting HO-1 expression [57,61,62].
Another pathway involved in HO-1 induction is hypoxia-inducible factor 1 α (HIF-1α) pathway [63]. Under hypoxic conditions, HIF-1α escapes proteasomal degradation, translocates to the nucleus, and dimerizes with HIF-1β, forming the HIF-1 complex, which binds to hypoxia-response elements (HREs) within the HO-1 promoter to initiate transcriptional upregulation [64,65] (Figure 3). Hypoxia is a common feature of solid tumors, including PC, resulting in HIF-1-mediated induction of HO-1, contributing to tumor adaptation and angiogenesis [63,66].
In addition to these pathways, the regulation of HO-1 expression is also influenced by non-coding RNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) [67]. Recent studies have demonstrated that miRNAs can modulate HO-1 expression in specific cell types. For instance, miR-377 and miR-217 have been shown to decrease HO-1 levels in endothelial cells by directly targeting the 3′ untranslated region (UTR) of HO-1 mRNA [68]. Similarly, miR-378 has been shown to suppress HO-1 levels in lung cancer cells [69]. lncRNAs also contribute to cancer progression by regulating gene expression [70]. They can act as molecular sponges for miRNAs or directly interact with transcription factors and chromatin-modifying complexes to influence HO-1 transcription [67]. These non-coding RNAs play crucial roles in the progression of PC [71,72], though their specific impact on HO-1 expression in PC requires further investigation. This adds an additional layer of complexity to the regulation of HO-1, emphasizing their potential role in the progression and therapeutic resistance of PC.
Inflammation, a hallmark of cancer, also promotes HO-1 expression via the nuclear factor-kappa B (NF-ƙB) pathway, activated by various cytokines, growth factors, and inflammatory stimuli [73,74]. The active NF-ƙB complex translocates to the nucleus and binds directly to the HO-1 promoter, activating its transcription (Figure 3). Another pro-inflammatory transcription factor, activator protein 1 (AP1), has an identified binding site within the HMOX1 promoter, and HO-1 upregulation has been correlated with both NF- ƙB and AP1 expression [75,76] (Figure 3). Chronic inflammation in the prostate microenvironment can create a pro-survival niche for tumor cells and inducing HO-1 may contribute to this process.
While several direct transcriptional regulators control HO-1 expression, many other inducers activate transcription via intermediate signaling pathways. Key protein kinase pathways associated with cellular stress, such as the mitogen-activated protein kinases (MAPKs), including ERK, JNK, and notably p38, are involved in regulating HO-1 [75,77] (Figure 3). Additionally, the phosphatidylinositol 3-kinase (PI3K)–AKT pathway influences HO-1 expression in response to cytokines and prostaglandins, with IL-10 upregulation mediated via PI3K and STAT3 [78]. AMP-activated protein kinase (AMPK) also modulates HO-1 by interacting with its downstream effectors and NRF2 [79]. The exact process through which the protein kinase signaling pathway triggers HO-1 expression is a complex interplay involving NRF2, AP-1, and HIF-1 transcription factors [57]. Furthermore, our lab has identified STAT1 as a potential regulator of HO-1 expression. Inhibiting STAT1 with fludarabine, a specific STAT1 inhibitor, has been shown to reduce HO-1 expression in various PC cell lines [80].
ROS and oxidative stress are common occurrences in cancer cells. As an antioxidant enzyme, HO-1 is upregulated in response to increased oxidative stress to help counteract ROS-induced cellular damage and support cancer cell survival and proliferation [33]. This upregulation of HO-1 is not typically a direct induction but occurs through various signaling pathways, such as NRF2 and NF-ƙB signaling pathways [56,81]. Through these pathways, HO-1 is upregulated in a coordinated response to oxidative stress, enabling the cell to mitigate ROS-induced damage and enhance survival and proliferation in the context of cancer.
Understanding the regulation of HO-1 expression in PC is crucial for unraveling its role in disease pathogenesis and therapy response. Targeting the pathways and factors involved in HO-1 induction may hold promise as a therapeutic strategy for PC treatment and warrants further investigation. Moreover, elucidating the interplay between inflammation, oxidative stress, hypoxia, and cancer-related factors in HO-1 regulation may provide valuable insights into the mechanisms underlying disease progression and therapeutic resistance in PC.

4. Role of HO-1 in PC Progression: Hypoxia

Understanding the fundamental biological events in prostate carcinogenesis is crucial to reducing PC mortality. Hypoxia, or low oxygen levels, is an early and significant event in prostate carcinogenesis, linked to aggressive cancer phenotype [33,82]. It plays a significant role in PC growth, metastasis, and the progression to hormone-refractory stages [82,83]. Hypoxia drives genetic, metabolic, and proteomic changes that enhance glycolysis, angiogenesis, stem cell-like properties, and the selection of resistant cancer clones [84,85]. Consequently, tumor hypoxia and related biomarkers are associated with poor prognosis, treatment failure, and disease relapse [85]. Targeting hypoxia-induced signaling pathways and their biological effects could offer effective strategies for preventing and treating PC.
Several signaling pathways are activated in the hypoxic microenvironment of PC, leading to the induction of HO-1. This enzyme plays a crucial role in helping cells adapt to hypoxic stress and mitigating the activation of proinflammatory pathways [86]. HO-1 overexpression is linked to improved cell survival under hypoxic conditions, highlighting its protective function and its promising therapeutic significance for PC treatment [87].
Under hypoxic conditions, the HIF-1α pathway is activated, leading to upregulating in HO-1 expression [88,89]. In response to this stress, HO-1 undergoes proteolytic cleavage, resulting in its nuclear translocation, where it modulates key transcription factors, facilitating the expression of critical genes that promote cell proliferation and invasion, enhancing tumorigenesis, and activating survival genes [45,90] (Figure 4). The induction of HO-1 under hypoxia helps cancer cells to survive and adapt to the low-oxygen environment by enhancing their antioxidative capacity and promoting angiogenesis.
There is a bidirectional relationship between HO-1 and HIF-1α. While HIF-1α directly upregulates HO-1 expression under hypoxic conditions, HO-1 can also stabilize HIF-1α, creating a feedback loop that amplifies the hypoxic response. HO-1’s byproducts, particularly CO, can inhibit prolyl hydroxylases (PHDs), the enzymes responsible for HIF-1α degradation under normoxic conditions [91,92,93]. By inhibiting PHDs, HO-1 prevents the degradation of HIF-1α, leading to its accumulation and sustained activity, which further promotes the transcription of hypoxia-responsive genes.
The role of HO-1 in the hypoxic response has significant implications for therapy resistance in PC (Figure 4). Data from our lab and others has demonstrated that hypoxia-induced HO-1 upregulation contributes to the resistance of cancer cells to conventional therapies, such as radiotherapy and chemotherapy, which often rely on oxygen-dependent mechanisms to induce cell death [80,93,94]. By enhancing the antioxidant defenses of cancer cells and promoting survival pathways, HO-1 helps to protect PC cells from the cytotoxic effects of these treatments, thereby contributing to therapy resistance and disease progression.
Hypoxic regions are commonly found in various cancers, including PC, rendering these tumors resistant to therapies. Hypoxia induces a range of responses in solid tumors, such as gene expression alteration, apoptosis suppression, EMT stimulation, and enhanced angiogenesis [95,96]. Moreover, Hypoxia also enhances clonal selection leading to increased invasion and metastasis, which poses a significant challenge to conventional therapies and contributes to cancer cell resistance against ADT, radiotherapy, and chemotherapy [95,97,98] (Figure 4).
In addition, hypoxic cancer stem cells (CSC) are characterized with high expression levels of Adenosine-triphosphate binding cassette transporters (ABC transporters) [99]. These transporters constitute a highly efficient efflux system that confers chemoresistance in cancer cells [100]. Activation of these ABC transporters by HIFs upon hypoxia conditions diminishes the effect of chemotherapy agents, thereby exacerbating chemoresistance [99].
In conclusion, the upregulation of HO-1 in response to hypoxia plays a crucial role in PC progression by enhancing cell survival, promoting angiogenesis, and contributing to therapy resistance. Therefore, employing HO-1 inhibitors alongside standard therapies could overcome resistance and improve treatment outcomes for PC patients.

5. The Interplay of HO-1, Oxidative Stress, and PC

Carcinogenesis is a complex process involving genetic alterations and dysregulated cell proliferation [65]. Oxidative stress, characterized by an imbalance between ROS production and antioxidant defense mechanisms, has been implicated in cancer initiation and progression [101,102]. Chronic oxidative stress leads to DNA damage, inflammation, and cellular dysfunction, contributing to carcinogenesis [103,104,105]. Enzymatic antioxidants such as superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPx), along with non-enzymatic antioxidants like glutathione (GSH), play crucial roles in neutralizing ROS and maintaining cellular redox homeostasis [106,107,108,109].
HO-1 is a stress-inducible enzyme activated by various stimuli including oxidants, cytokines, and heavy metals [110]. While HO-1 exhibits cytoprotective effects in normal cells by mitigating oxidative stress and inflammation [111,112], its overexpression is observed in several cancer types, including PC, where it contributes to tumor cell survival and resistance to therapy [33,55,113,114]. Understanding the mechanisms underlying HO-1-mediated redox regulation in cancer cells is critical for developing effective therapeutic strategies. Combinatorial approaches targeting HO-1 alongside conventional chemotherapy or radiotherapy hold promise for overcoming treatment resistance in PC [113].
HO -1 serves as the rate-controlling enzyme in cellular heme catabolism, catalyzing the breakdown of heme moieties into equimolar amounts of CO, Fe, and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase [115,116] (Figure 2). HO-1’s cellular protective effects stem from its ability to suppress inflammation and oxidative stress [117]. Bilirubin, a byproduct of heme degradation, exhibits antioxidant properties and likely contributes to the mammalian body’s redox balance by scavenging ROS [118,119,120]. Moreover, CO exerts physiological functions, including anti-inflammatory and anti-apoptotic effects mediated through the mitogen-activated protein kinase (MAPK) pathway, as well as vasodilatory effects [121,122]. Free iron generated by HO-1 activity is sequestered by ferritin, thereby preventing hydroxyl radical formation via the Fenton reaction [123].
Notably, numerous studies, including investigations conducted in our laboratory, have elucidated the multifaceted roles of HO-1 in cellular processes such as growth regulation, angiogenesis, response to bacterial infection, and modulation of macrophage polarization towards anti-inflammatory (M2) or pro-inflammatory (M1) phenotypes [124,125,126]. The antioxidant properties of HO-1 enable cancer cells to evade ROS-induced DNA damage and apoptosis (Figure 4), thereby conferring resistance to chemotherapy and radiotherapy [113,114]. Consequently, the combined administration of chemotherapy and/or radiation therapy with HO-1-inhibiting agents holds promise for overcoming therapeutic resistance in cancer treatment.
Clinical studies have reported that HO-1 expression is increased in PC patients, which correlates with increased serum PSA, resistance to therapy, and poor clinical outcomes [127,128,129,130]. It has been shown that overexpression of HO-1 correlates with PC growth, aggressiveness, and metastasis [50,127,131]. However, conflicting opinions have emerged on whether HO-1 provides growth advantages and metastasis to PC cells or promotes cell death [132]. Several concerns should be addressed. The available literature suggests that the effects of HO-1 depend on the amount of ROS. Low ROS levels enhance cell survival, while high levels promote cell death [133,134] (Figure 4). Under oxidative stress, increased ROS levels enhance HO-1 expression, thereby protecting PC cells by mitigating the deleterious effects of ROS, resulting in metastasis and resistance to cancer treatments [135]. However, HO-1 inhibition in PC tilts the balance towards ROS overproduction, promoting cell death and inhibiting PC proliferation and metastasis [136,137] (Figure 4). Thus, targeting HO-1 pathways may serve as a novel therapeutic strategy in PC therapy.

6. HO-1 and PC Metastasis

HO-1 plays a critical role in the metastatic progression of PC. Metastasis, the spread of cancer cells from the primary tumor to distant organs, is a significant cause of morbidity and mortality among PC patients [138,139,140]. Understanding the underlying mechanisms of metastasis in PC is a critical aspect to improving cancer prognosis. As mentioned previously, HO-1 is notably upregulated in response to various stressors, including hypoxia, which is common in the tumor microenvironment [141]. This upregulation is essential not only for the survival of cancer cells under stress but also for their metastatic capabilities [142].
HO-1 enzymatic byproduct, such as biliverdin and CO, possess pro-angiogenic properties [27,44,143]. These catabolic products contribute to forming new blood vessels by activating VEGF, a key regulator of angiogenesis, which is vital for supplying nutrients and oxygen to growing tumors [144,145]. By driving angiogenesis, HO-1 supports tumor expansion and enables cancer cells to enter the bloodstream and metastasize to other parts of the body [146,147,148] (Figure 4).
Furthermore, HO-1 enhances the invasive and migratory abilities of PC cells by modulating the expression of matrix metalloproteinases (MMPs), which degrade the extracellular matrix and facilitate cancer cell invasion [149,150]. Additionally, HO-1 influences the epithelial-mesenchymal transition (EMT), a process where epithelial cells acquire mesenchymal properties, increasing their motility and invasiveness [151,152] (Figure 4). Studies, including those from our lab, have demonstrated that inhibiting HO-1 can downregulate the expression of EMT markers and attenuate PC cell migration [31,80].
Furthermore, aggressive prostate tumors often induce supportive changes in adjacent benign tissues, one of which is the increased expression of HO-1 [32]. HO-1 is predominantly produced by macrophages that accumulate in tumor regions and at the tumor-invasive front [153,154]. This is evident in both rat models and human clinical samples. In rat prostate tumors, small metastatic tumors were more effective at attracting HO-1+ macrophages compared to larger non-metastatic ones [32]. Similarly, in human samples, high-grade tumors and bone metastases showed significant accumulation of HO-1+ macrophages at invasive fronts, which correlated with bone metastases presence and lower AR expression [32]. This suggests that HO-1+ macrophages play a crucial role in the metastatic progression of PC.
Recent studies suggest that hypoxic cancer cells, including PC cells, release more exosomes loaded with bioactive molecules such as proteins, lipids, and nucleic acids [155,156]. These exosomes can alter the tumor microenvironment and promote metastasis [157,158]. Further supporting this, studies indicate that exosomal long noncoding RNA (lncRNA) HOXD-AS1 is upregulated in CRPC and serum exosomes from metastatic PC patients [156]. Exosomal HOXD-AS1 promotes metastasis by modulating the miR-361-5p/FOXM1 axis, enhancing cancer cell invasiveness. Additionally, serum exosomal HOXD-AS1 levels correlate with higher Gleason scores and worse clinical outcomes, highlighting its potential as a biomarker for detecting and treating metastatic PC [156]. HO-1 upregulation has been linked to increased exosome production, further facilitating the metastatic process.
Additionally, the hypoxic response promotes the expression of genes involved in angiogenesis, invasion, and survival under low oxygen conditions [159]. Furthermore, the immunomodulating capabilities of HO-1 in PC, which will be described later in detail, exert anti-inflammatory effects, which can suppress anti-tumor immunity [159]. This immune modulation creates a more favorable environment for cancer cells to thrive and spread.
The role of HO-1 in PC metastasis underscores its potential as a therapeutic target. Inhibiting HO-1 could disrupt the various pathways it influences, thereby impairing the metastatic capabilities of cancer cells. This approach could be particularly beneficial when combined with conventional therapies, which may be less effective against metastatic disease due to the protective effects conferred by HO-1.

7. HO-1 and Therapy Resistance in PC

HO-1 has emerged as a significant player in therapy resistance mechanisms observed in PC [27,33,159,160], encompassing resistance to ADT, radiation therapy, and chemotherapy. Resistance to ADT is a pivotal challenge in the treatment landscape of PC [161]. As the primary treatment for PC, ADT aims to suppress the AR signaling pathway, thereby inhibiting the growth and proliferation of PC cells [162]. However, despite the initial efficacy of traditional treatment options, many patients eventually develop resistance, leading to the progression of PC into CRPC. The emergence of CRPC poses significant clinical hurdles and necessitates a deeper understanding of the underlying resistance mechanisms [163,164].
While growing evidence suggests that HO-1 may be involved in resistance to ADT in PC, the specific role of HO-1 in ADT resistance is still an area of active research and ongoing investigation [33]. Several studies have reported elevated levels of HO-1 in CRPC, and its upregulation has been implicated in several resistance mechanisms [159,165,166]. One such mechanism involves the anti-inflammatory products of heme degradation that protect the cancer cell against the oxidative damage caused by therapies [142,159]. Additionally, HO-1-mediated induction of anti-apoptotic pathways and suppression of apoptotic signaling contribute to enhanced survival of PC cells under androgen-deprived conditions [143]. These observations underscore the critical need for further research to elucidate the precise molecular mechanisms through which HO-1 contributes to ADT resistance and to clarify its impact on AR expression, the principal driver of PC progression.
Following ADT, resistance to radiation therapy also presents a significant challenge in PC management. Radiation therapy is commonly employed for localized and locally advanced PC, aiming to eradicate cancer cells through targeted radiation-induced DNA damage [113,167]. However, resistance to radiation therapy often develops, leading to treatment failure and disease recurrence [168]. In the context of radiation resistance, HO-1’s role extends to multiple facets of cellular response to radiation-induced stress. HO-1 upregulation has been associated with enhanced cellular antioxidant defenses, which can mitigate the cytotoxic effects of radiation-induced oxidative stress [168]. Additionally, HO-1 may promote DNA damage repair mechanisms enabling cancer cells to survive and proliferate despite radiation exposure [169]. Furthermore, HO-1 inhibition enhanced the radiosensitivity of human non-small lung cancer [170].
Chemotherapy resistance further complicates PC management, particularly in the advanced stages of the disease [169]. HO-1’s multifaceted role in chemotherapy resistance encompasses mechanisms such as enhanced antioxidant defenses, modulation of apoptotic pathways, and promotion of pro-survival signaling pathways [44,159,160]. Clinical evidence highlights the possible correlation between HO-1 expression and PC growth, aggressiveness, metastasized tumor, resistance to therapy, and poor clinical outcome [159]. Furthermore, research conducted in our lab has shown that inhibiting HO-1 enhances the chemosensitivity of PC cells by modulating several interconnected pathways related to apoptosis, including increased ROS production, disruption of the glutathione cycle, and modulation of the STAT1 pathway, in addition to attenuating cell migration [80]. Understanding the complex interplay between HO-1 and therapy resistance mechanisms is crucial for developing effective treatment strategies to improve treatment outcomes in PC.
Moreover, HO-1’s immunomodulatory properties may further exacerbate resistance to therapies. By promoting immune evasion and dampening anti-tumor immune responses, HO-1 creates a microenvironment conducive to cancer cell survival and proliferation despite oxidative stress caused by different therapies [171].
The role of HO-1 in therapy resistance underscores its potential as a therapeutic target to enhance treatment efficacy and overcome acquired resistance. Our lab previously showed that inhibiting HO-1 suppressed cell proliferation and increased the efficacy of chemotherapy treatment in pancreatic ductal adenocarcinoma and PC [80,126]. Therefore, strategies aimed at inhibiting HO-1 activity or disrupting its downstream signaling pathways hold promise for improving therapy outcomes and prolonging PC patient’s survival.

8. Immunomodulation by HO-1 in PC

Overexpression of HO-1 in cancer cells, including PC, has been shown to promote tumor progression through multiple mechanisms, including the modulation and enforcing the immune suppressive characteristics of the TME. Both malignant tumor cells and stromal cells within the TME can express HO-1 [172,173,174]. The catabolites of heme degradation can also diffuse outside of the cell, modulating the wider TME and influencing cellular functionality and biological processes that promote tumor progression, such as facilitating angiogenesis and metastasis, as well as fostering anti-inflammatory and immune-suppressive environments [172].
Tumor-associated macrophages (TAMs) represent the most abundant stromal cell type in the TME and the primary source of HO-1 in murine models and humans [52,173]. TAMs respond to the TME and adapt a variety of phenotypes, including pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes, each with distinct gene expression and cytokine profiles [175]. HO-1 upregulation has been associated with the M2 phenotype, which is the tumor-promoting macrophage phenotype, suggesting that HO-1 may contribute to macrophage polarization [176,177,178].
Other immune cells in the TME, such as dendritic cells (DC), CD4+ T cells, CD8+ T cells, and Foxp3+ regulatory T cells (Treg), have been shown to express HO-1, which can influence their activity and suppress their immunogenicity [30]. Since the catabolites of heme degradation diffuse to the extracellular matrix, they may also affect other immune cells that don’t express HO-1, influencing their polarization and recruitment.
Oncogenes in cancer cells or genetic mutations in the HMOX1 promotor, the gene encoding HO-1, can drive the constitutive expression of HO-1, leading to potent immunomodulating anti-tumor immune responses. The immunomodulating role of HO-1 supports its consideration as an innate immune checkpoint [52]. Blocking immune checkpoints, which are regulatory proteins that suppress T-cell activity and immune responses, has resulted in an unprecedented clinical response in cancer patients [179,180]. In parallel comparison, inhibition of HO-1 displayed superior immunological control of tumor growth in a murine mammary adenocarcinoma model compared to anti-PD1 immunotherapy [52]. This observation underscores the potential of targeting HO-1 as a novel immunotherapy approach.
Heme degradation products, such as bilirubin and CO, have significant immunomodulatory effects [30]. The upregulation of HO-1 in PC has been correlated with advanced disease stages, cancer progression, and metastasis. Specifically, HO-1-expressing macrophages have been implicated in contributing to bone metastasis [32]. Furthermore, inhibiting HO-1 has been shown to enhance the efficacy of certain cancer treatments in PC, suggesting that HO-1 inhibition may improve the overall immune response against PC and increase the effectiveness of other therapies [129]. Although the collective evidence underscores the role of HO-1 in immunomodulation within PC, the precise function of HO-1 in PC remains incompletely understood and continues to be an active area of research.
In conclusion, HO-1-induced immunomodulation may contribute to immune evasion and therapy resistance in PC. By suppressing immune-mediated anti-tumor responses, HO-1 creates an immunosuppressive microenvironment that fosters cancer cell survival and proliferation. Understanding the complex interplay between HO-1 and therapy resistance mechanisms is crucial for developing effective strategies to overcome resistance and improve treatment outcomes in PC.

9. Targeting HO-1 as a Therapeutic Strategy

HO-1 has emerged as a promising therapeutic target in various diseases due to its inducible nature. This inducibility allows for targeted inhibition using metalloporphyrins (MPs), compounds structurally similar to heme but with a metal ion, such as tin or zinc, replacing the iron in heme [135]. MPs bind to HO-1 with a higher affinity than heme, inhibiting its function. These elements can modulate HO-1 activity, providing potential avenues for therapeutic intervention [181]. In addition to MPs, non-porphyrin-based inhibitors are also being explored. These include small molecule inhibitors like OB-24, as well as RNA interference techniques such as small interfering RNA (siRNA) and short hairpin RNA (shRNA) [129,135,182,183,184,185].
The strategy of HO-1 inhibition first emerged as a promising therapeutic approach in the late 1980s [186]. Research has shown that overexpression of HO-1 is strongly correlated with elevated bilirubin levels, leading to neonatal hyperbilirubinemia [187]. Based on these findings, studies have investigated how HO-1 inhibition can reduce bilirubin levels in newborns and subsequently mitigate hyperbilirubinemia [188]. Furthermore, HO-1 inhibition has shown therapeutic potential across a broad spectrum of diseases, including M. Tuberculosis (TB) and Alzheimer’s disease [189,190,191,192]. However, the function of HO-1 in PC still needs to be fully elucidated.
As mentioned previously, HO-1 exhibits anti-inflammatory effects by reducing pro-inflammatory mediators, positioning it as an attractive target in PC, where inflammation significantly contributes to disease progression [193,194]. Hypoxia plays a significant role in PC progression and metastasis activating the expression of multiple pathways that mediate adaptation including HO-1 [82,87]. Moreover, HO-1 promotes tumor growth by inducing angiogenesis, primarily through the expression of angiogenic factors such as VEGF [175,176]. Additionally, HO-1 facilitates PC metastasis, leading to advanced disease stages and compromising the efficacy of localized treatments. HO-1 modulates the tumor microenvironment by fostering pro-inflammatory niches supporting PC survival. Importantly, HO-1 also confers resistance to ADT, radiotherapy, and chemotherapy by activating anti-apoptotic pathways, enhancing cellular antioxidant defenses, and promoting DNA repair mechanisms.
Interestingly, both induction and inhibition of HO-1 have shown anticancer effects in PC models [55,132,141]. HO-1 effects in PC depend on whether the cells are normal or malignant and the level of HO-1 activity. Evidence suggests HO-1 has dual roles in PC, influenced by cellular iron and ROS levels [33]. Low to moderate levels of ROS reinforce HO-1’s protective role by promoting anti-apoptotic pathways, thereby facilitating PC progression and enhancing cancer cell survival, treatment resistance, and metastasis [159]. Targeting HO-1 in PC can mitigate these protective effects by elevating ROS levels in cancer cells, suppressing cellular survival, and increasing tumor cell sensitivity to therapies. These excessive ROS levels trigger programmed cell death pathways, which in turn suppress tumor growth.
Clinical studies have shown increased HO-1 expression in PC patients, correlating with higher Gleason grades, elevated PSA, therapy resistance, and poor clinical outcomes [131,195]. Consequently, regulating HO-1 expression represents a promising avenue in the landscape of PC treatment, and HO-1 could also be a valuable biomarker for PC progression. In normal cells, HO-1 induction provides cytoprotection by neutralizing oxidative stress, thereby inhibiting PC growth. Thus, HO-1 represents a potential target for therapeutic prevention by enhancing its protective functions in normal cells while also serving as a treatment strategy by inhibiting PC progression and angiogenesis.
Targeting HO-1 could revolutionize various stages of PC treatment. For newly diagnosed patients undergoing RT or radical prostatectomy, an HO-1 inhibitor might enhance the efficacy of these treatments by reducing therapy resistance, potentially leading to better patient outcomes and higher cure rates. In patients with de novo metastatic disease, inhibiting HO-1 could limit the cancer’s ability to spread further and enhance the effectiveness of systemic therapies, offering new hope in combating advanced stages of the disease. Even in patients with relapsed PC, where treatment options become limited and less effective, HO-1 inhibition could provide a new avenue to re-sensitize cancer cells to existing therapies. These multifaceted roles highlight the potential of targeting HO-1 as a therapeutic strategy in PC.

10. Future Perspectives and Conclusions

Targeting HO-1 or its downstream effectors may offer promising avenues for enhancing the efficacy of existing therapies and overcoming resistance in PC treatment. The clinical application of HO-1 inhibition is still in its early stages, with ongoing research focusing on optimizing their efficacy and minimizing side effects. Future studies are needed to better understand the molecular pathways regulated by HO-1 and to identify biomarkers for selecting patients who might benefit the most from HO-1-targeted therapies. Additionally, exploring the role of HO-1 in combination with standard treatment modalities, such as hormonal therapy, radiotherapy, and chemotherapy, for more effective cancer treatments.
In conclusion, Targeting HO-1 represents a novel and promising approach in cancer therapy, including PC. By inhibiting this key enzyme, it may be possible to disrupt crucial pathways involved in tumor growth, metastasis, and immune evasion, thereby improving patient outcomes. As research progresses, the development of HO-1 inhibitors and their integration into clinical practice holds the potential to revolutionize cancer treatment strategies.

Author Contributions

Conceptualization, R.J.S. and M.Y.A.; writing-original draft preparation, R.J.S., I.M.A., D.Z.S., M.J.B. and M.Y.A.; writing-review and editing, R.J.S., M.J.B. and M.Y.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef] [PubMed]
  2. Rawla, P. Epidemiology of Prostate Cancer. World J. Oncol. 2019, 10, 63–89. [Google Scholar] [CrossRef]
  3. Panigrahi, G.K.; Praharaj, P.P.; Kittaka, H.; Mridha, A.R.; Black, O.M.; Singh, R.; Mercer, R.; van Bokhoven, A.; Torkko, K.C.; Agarwal, C.; et al. Exosome proteomic analyses identify inflammatory phenotype and novel biomarkers in African American prostate cancer patients. Cancer Med. 2019, 8, 1110–1123. [Google Scholar] [CrossRef] [PubMed]
  4. Shore, N.D.; Morgans, A.K.; El-Haddad, G.; Srinivas, S.; Abramowitz, M. Addressing Challenges and Controversies in the Management of Prostate Cancer with Multidisciplinary Teams. Target. Oncol. 2022, 17, 709–725. [Google Scholar] [CrossRef] [PubMed]
  5. Wilson, A.H. The Prostate Gland: A Review of its Anatomy, Pathology, and Treatment. JAMA 2014, 312, 562. [Google Scholar] [CrossRef]
  6. Litwin, M.S.; Tan, H.-J. The Diagnosis and Treatment of Prostate Cancer: A Review. JAMA 2017, 317, 2532–2542. [Google Scholar] [CrossRef]
  7. Berenguer, C.V.; Pereira, F.; Câmara, J.S.; Pereira, J.A.M. Underlying Features of Prostate Cancer-Statistics, Risk Factors, and Emerging Methods for Its Diagnosis. Curr. Oncol. 2023, 30, 2300–2321. [Google Scholar] [CrossRef] [PubMed]
  8. Leitzmann, M.F.; Rohrmann, S. Risk factors for the onset of prostatic cancer: Age, location, and behavioral correlates. Clin. Epidemiol. 2012, 4, 1–11. [Google Scholar] [CrossRef]
  9. Oskar, B.; Kelly, R.P.; Konstantina, M.; Jonathan, F.; Sean, F.M.; Ola, B.; Freddie, B.; Otis, B.; Amy, N.L.; Lorelei, M.; et al. 2022 Update on Prostate Cancer Epidemiology and Risk Factors—A Systematic Review. Eur. Urol. 2023, 84, 191–206. [Google Scholar] [CrossRef]
  10. Giorgio, G.; Riccardo, L.; Freddie, B.; Neil, F.; Stephen, J.F.; Adam, K.; Pär, S.; Hendrick, C. Epidemiology and Prevention of Prostate Cancer. Eur. Urol. Oncol. 2021, 4, 877–892. [Google Scholar] [CrossRef]
  11. Ilic, D.; Djulbegovic, M.; Jung, J.H.; Hwang, E.C.; Zhou, Q.; Cleves, A.; Agoritsas, T.; Dahm, P. Prostate cancer screening with prostate-specific antigen (PSA) test: A systematic review and meta-analysis. BMJ 2018, 362, k3519. [Google Scholar] [CrossRef] [PubMed]
  12. Wu, J.T. Assay for prostate specific antigen (PSA): Problems and possible solutions. J. Clin. Lab. Anal. 1994, 8, 51–62. [Google Scholar] [CrossRef] [PubMed]
  13. Fenton, J.J.; Weyrich, M.S.; Durbin, S.; Liu, Y.; Bang, H.; Melnikow, J.U.S. Preventive Services Task Force Evidence Syntheses, formerly Systematic Evidence Reviews. In Prostate-Specific Antigen-Based Screening for Prostate Cancer: A Systematic Evidence Review for the U.S. Preventive Services Task Force; Agency for Healthcare Research and Quality: Rockville, MD, USA, 2018. [Google Scholar]
  14. Gleason, D.F. Classification of prostatic carcinomas. Cancer Chemother. Rep. 1966, 50, 125–128. [Google Scholar] [PubMed]
  15. Qiu, J.; Cai, D.; Wang, Z.; Zhou, J.; Gong, Y.; Cai, L.; Gong, K. Prognostic Models for Patients With Gleason Score 9 Prostate Cancer: A Population-Based Study. Front. Oncol. 2021, 11, 633312. [Google Scholar] [CrossRef] [PubMed]
  16. Tsao, C.K.; Gray, K.P.; Nakabayashi, M.; Evan, C.; Kantoff, P.W.; Huang, J.; Galsky, M.D.; Pomerantz, M.; Oh, W.K. Patients with Biopsy Gleason 9 and 10 Prostate Cancer Have Significantly Worse Outcomes Compared to Patients with Gleason 8 Disease. J. Urol. 2015, 194, 91–97. [Google Scholar] [CrossRef]
  17. Chen, F.Z.; Zhao, X.K. Prostate cancer: Current treatment and prevention strategies. Iran. Red Crescent Med. J. 2013, 15, 279–284. [Google Scholar] [CrossRef]
  18. Komura, K.; Sweeney, C.J.; Inamoto, T.; Ibuki, N.; Azuma, H.; Kantoff, P.W. Current treatment strategies for advanced prostate cancer. Int. J. Urol. 2018, 25, 220–231. [Google Scholar] [CrossRef]
  19. Coutinho, I.; Day, T.K.; Tilley, W.D.; Selth, L.A. Androgen receptor signaling in castration-resistant prostate cancer: A lesson in persistence. Endocr. Relat. Cancer 2016, 23, T179–T197. [Google Scholar] [CrossRef]
  20. Leung, D.K.; Chiu, P.K.; Ng, C.F.; Teoh, J.Y. Novel Strategies for Treating Castration-Resistant Prostate Cancer. Biomedicines 2021, 9, 339. [Google Scholar] [CrossRef]
  21. Posdzich, P.; Darr, C.; Hilser, T.; Wahl, M.; Herrmann, K.; Hadaschik, B.; Grünwald, V. Metastatic Prostate Cancer-A Review of Current Treatment Options and Promising New Approaches. Cancers 2023, 15, 461. [Google Scholar] [CrossRef]
  22. Taylor, J.M.; Chen, V.E.; Miller, R.C.; Greenberger, B.A. The Impact of Prostate Cancer Treatment on Quality of Life: A Narrative Review with a Focus on Randomized Data. Res. Rep. Urol. 2020, 12, 533–546. [Google Scholar] [CrossRef]
  23. Simon, N.I.; Parker, C.; Hope, T.A.; Paller, C.J. Best Approaches and Updates for Prostate Cancer Biochemical Recurrence. Am. Soc. Clin. Oncol. Educ. Book 2022, 42, 1–8. [Google Scholar] [CrossRef] [PubMed]
  24. Lin, X.; Kapoor, A.; Gu, Y.; Chow, M.J.; Xu, H.; Major, P.; Tang, D. Assessment of biochemical recurrence of prostate cancer (Review). Int. J. Oncol. 2019, 55, 1194–1212. [Google Scholar] [CrossRef]
  25. Garcia-Marques, F.; Liu, S.; Totten, S.M.; Bermudez, A.; Tanimoto, C.; Hsu, E.C.; Nolley, R.; Hembree, A.; Stoyanova, T.; Brooks, J.D.; et al. Protein signatures to distinguish aggressive from indolent prostate cancer. Prostate 2022, 82, 605–616. [Google Scholar] [CrossRef]
  26. Seetharaman, A.; Bhattacharya, I.; Chen, L.C.; Kunder, C.A.; Shao, W.; Soerensen, S.J.C.; Wang, J.B.; Teslovich, N.C.; Fan, R.E.; Ghanouni, P.; et al. Automated detection of aggressive and indolent prostate cancer on magnetic resonance imaging. Med. Phys. 2021, 48, 2960–2972. [Google Scholar] [CrossRef] [PubMed]
  27. Ryter, S.W. Heme Oxgenase-1, a Cardinal Modulator of Regulated Cell Death and Inflammation. Cells 2021, 10, 515. [Google Scholar] [CrossRef]
  28. 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]
  29. Hayashi, S.; Omata, Y.; Sakamoto, H.; Higashimoto, Y.; Hara, T.; Sagara, Y.; Noguchi, M. Characterization of rat heme oxygenase-3 gene. Implication of processed pseudogenes derived from heme oxygenase-2 gene. Gene 2004, 336, 241–250. [Google Scholar] [CrossRef]
  30. Luu Hoang, K.N.; Anstee, J.E.; Arnold, J.N. The Diverse Roles of Heme Oxygenase-1 in Tumor Progression. Front. Immunol. 2021, 12, 658315. [Google Scholar] [CrossRef]
  31. Naito, Y.; Takagi, T.; Higashimura, Y. Heme oxygenase-1 and anti-inflammatory M2 macrophages. Arch. Biochem. Biophys. 2014, 564, 83–88. [Google Scholar] [CrossRef]
  32. Halin Bergström, S.; Nilsson, M.; Adamo, H.; Thysell, E.; Jernberg, E.; Stattin, P.; Widmark, A.; Wikström, P.; Bergh, A. Extratumoral Heme Oxygenase-1 (HO-1) Expressing Macrophages Likely Promote Primary and Metastatic Prostate Tumor Growth. PLoS ONE 2016, 11, e0157280. [Google Scholar] [CrossRef] [PubMed]
  33. Chiang, S.K.; Chen, S.E.; Chang, L.C. The Role of HO-1 and Its Crosstalk with Oxidative Stress in Cancer Cell Survival. Cells 2021, 10, 2401. [Google Scholar] [CrossRef] [PubMed]
  34. Kiening, M.; Lange, N. A Recap of Heme Metabolism towards Understanding Protoporphyrin IX Selectivity in Cancer Cells. Int. J. Mol. Sci. 2022, 23, 7974. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, J.-J.; Zhang, S. Heme-regulated eIF2α kinase in erythropoiesis and hemoglobinopathies. Blood 2019, 134, 1697–1707. [Google Scholar] [CrossRef] [PubMed]
  36. Yuan, Z.; De La Cruz, L.K.; Yang, X.; Wang, B. Carbon Monoxide Signaling: Examining Its Engagement with Various Molecular Targets in the Context of Binding Affinity, Concentration, and Biologic Response. Pharmacol. Rev. 2022, 74, 823–873. [Google Scholar] [CrossRef]
  37. Adin, C.A. Bilirubin as a Therapeutic Molecule: Challenges and Opportunities. Antioxidants 2021, 10, 1536. [Google Scholar] [CrossRef]
  38. Galy, B.; Conrad, M.; Muckenthaler, M. Mechanisms controlling cellular and systemic iron homeostasis. Nat. Rev. Mol. Cell Biol. 2024, 25, 133–155. [Google Scholar] [CrossRef]
  39. Kim, K.M.; Pae, H.-O.; Zheng, M.; Park, R.; Kim, Y.-M.; Chung, H.-T. Carbon Monoxide Induces Heme Oxygenase-1 via Activation of Protein Kinase R–Like Endoplasmic Reticulum Kinase and Inhibits Endothelial Cell Apoptosis Triggered by Endoplasmic Reticulum Stress. Circ. Res. 2007, 101, 919–927. [Google Scholar] [CrossRef]
  40. Migita, C.T.; Matera, K.M.; Ikeda-Saito, M.; Olson, J.S.; Fujii, H.; Yoshimura, T.; Zhou, H.; Yoshida, T. The Oxygen and Carbon Monoxide Reactions of Heme Oxygenase. J. Biol. Chem. 1998, 273, 945–949. [Google Scholar] [CrossRef]
  41. Matera, K.M.; Takahashi, S.; Fujii, H.; Zhou, H.; Ishikawa, K.; Yoshimura, T.; Rousseau, D.L.; Yoshida, T.; Ikeda-Saito, M. Oxygen and one reducing equivalent are both required for the conversion of alpha-hydroxyhemin to verdoheme in heme oxygenase. J. Biol. Chem. 1996, 271, 6618–6624. [Google Scholar] [CrossRef]
  42. Sugishima, M.; Moffat, K.; Noguchi, M. Discrimination between CO and O2 in heme oxygenase: Comparison of static structures and dynamic conformation changes following CO photolysis. Biochemistry 2012, 51, 8554–8562. [Google Scholar] [CrossRef]
  43. Yang, Q.; Wang, W. The Nuclear Translocation of Heme Oxygenase-1 in Human Diseases. Front. Cell Dev. Biol. 2022, 10, 890186. [Google Scholar] [CrossRef] [PubMed]
  44. Chau, L.-Y. Heme oxygenase-1: Emerging target of cancer therapy. J. Biomed. Sci. 2015, 22, 22. [Google Scholar] [CrossRef]
  45. Lin, Q.; Weis, S.; Yang, G.; Weng, Y.H.; Helston, R.; Rish, K.; Smith, A.; Bordner, J.; Polte, T.; Gaunitz, F.; et al. Heme oxygenase-1 protein localizes to the nucleus and activates transcription factors important in oxidative stress. J. Biol. Chem. 2007, 282, 20621–20633. [Google Scholar] [CrossRef] [PubMed]
  46. Vanella, L.; Barbagallo, I.; Tibullo, D.; Forte, S.; Zappalà, A.; Li Volti, G. The non-canonical functions of the heme oxygenases. Oncotarget 2016, 7, 69075–69086. [Google Scholar] [CrossRef]
  47. Hsu, F.F.; Chiang, M.T.; Li, F.A.; Yeh, C.T.; Lee, W.H.; Chau, L.Y. Acetylation is essential for nuclear heme oxygenase-1-enhanced tumor growth and invasiveness. Oncogene 2017, 36, 6805–6814. [Google Scholar] [CrossRef] [PubMed]
  48. Scaffa, A.; Tollefson, G.A.; Yao, H.; Rizal, S.; Wallace, J.; Oulhen, N.; Carr, J.F.; Hegarty, K.; Uzun, A.; Dennery, P.A. Identification of Heme Oxygenase-1 as a Putative DNA-Binding Protein. Antioxidants 2022, 11, 2135. [Google Scholar] [CrossRef]
  49. Mascaró, M.; Alonso, E.N.; Alonso, E.G.; Lacunza, E.; Curino, A.C.; Facchinetti, M.M. Nuclear Localization of Heme Oxygenase-1 in Pathophysiological Conditions: Does It Explain the Dual Role in Cancer? Antioxidants 2021, 10, 87. [Google Scholar] [CrossRef]
  50. Canesin, G.; Muralidharan, A.M.; Swanson, K.D.; Wegiel, B. HO-1 and Heme: G-Quadruplex Interaction Choreograph DNA Damage Responses and Cancer Growth. Cells 2021, 10, 1801. [Google Scholar] [CrossRef]
  51. Miyata, Y.; Kanda, S.; Mitsunari, K.; Asai, A.; Sakai, H. Heme oxygenase-1 expression is associated with tumor aggressiveness and outcomes in patients with bladder cancer: A correlation with smoking intensity. Transl. Res. 2014, 164, 468–476. [Google Scholar] [CrossRef]
  52. Muliaditan, T.; Opzoomer, J.W.; Caron, J.; Okesola, M.; Kosti, P.; Lall, S.; Van Hemelrijck, M.; Dazzi, F.; Tutt, A.; Grigoriadis, A.; et al. Repurposing Tin Mesoporphyrin as an Immune Checkpoint Inhibitor Shows Therapeutic Efficacy in Preclinical Models of Cancer. Clin. Cancer Res. 2018, 24, 1617–1628. [Google Scholar] [CrossRef] [PubMed]
  53. Yin, H.; Fang, J.; Liao, L.; Maeda, H.; Su, Q. Upregulation of heme oxygenase-1 in colorectal cancer patients with increased circulation carbon monoxide levels, potentially affects chemotherapeutic sensitivity. BMC Cancer 2014, 14, 436. [Google Scholar] [CrossRef]
  54. Fest, S.; Soldati, R.; Christiansen, N.M.; Zenclussen, M.L.; Kilz, J.; Berger, E.; Starke, S.; Lode, H.N.; Engel, C.; Zenclussen, A.C.; et al. Targeting of heme oxygenase-1 as a novel immune regulator of neuroblastoma. Int. J. Cancer 2016, 138, 2030–2042. [Google Scholar] [CrossRef] [PubMed]
  55. Wegiel, B.; Gallo, D.; Csizmadia, E.; Harris, C.; Belcher, J.; Vercellotti, G.M.; Penacho, N.; Seth, P.; Sukhatme, V.; Ahmed, A.; et al. Carbon monoxide expedites metabolic exhaustion to inhibit tumor growth. Cancer Res. 2013, 73, 7009–7021. [Google Scholar] [CrossRef] [PubMed]
  56. He, F.; Ru, X.; Wen, T. NRF2, a Transcription Factor for Stress Response and Beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef] [PubMed]
  57. Medina, M.V.; Sapochnik, D.; Garcia Solá, M.; Coso, O. Regulation of the Expression of Heme Oxygenase-1: Signal Transduction, Gene Promoter Activation, and Beyond. Antioxid. Redox Signal. 2020, 32, 1033–1044. [Google Scholar] [CrossRef]
  58. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An Overview of Nrf2 Signaling Pathway and Its Role in Inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef]
  59. Tong, K.I.; Kobayashi, A.; Katsuoka, F.; Yamamoto, M. Two-site substrate recognition model for the Keap1-Nrf2 system: A hinge and latch mechanism. Biol. Chem. 2006, 387, 1311–1320. [Google Scholar] [CrossRef]
  60. Tong, K.I.; Padmanabhan, B.; Kobayashi, A.; Shang, C.; Hirotsu, Y.; Yokoyama, S.; Yamamoto, M. Different electrostatic potentials define ETGE and DLG motifs as hinge and latch in oxidative stress response. Mol. Cell. Biol. 2007, 27, 7511–7521. [Google Scholar] [CrossRef]
  61. Dhakshinamoorthy, S.; Jain, A.K.; Bloom, D.A.; Jaiswal, A.K. Bach1 competes with Nrf2 leading to negative regulation of the antioxidant response element (ARE)-mediated NAD(P)H:quinone oxidoreductase 1 gene expression and induction in response to antioxidants. J. Biol. Chem. 2005, 280, 16891–16900. [Google Scholar] [CrossRef]
  62. Reichard, J.F.; Motz, G.T.; Puga, A. Heme oxygenase-1 induction by NRF2 requires inactivation of the transcriptional repressor BACH1. Nucleic Acids Res. 2007, 35, 7074–7086. [Google Scholar] [CrossRef]
  63. Masoud, G.N.; Li, W. HIF-1α pathway: Role, regulation and intervention for cancer therapy. Acta. Pharm. Sin. B 2015, 5, 378–389. [Google Scholar] [CrossRef] [PubMed]
  64. Ma, L.L.; Sun, L.; Wang, Y.X.; Sun, B.H.; Li, Y.F.; Jin, Y.L. Association between HO-1 gene promoter polymorphisms and diseases (Review). Mol. Med. Rep. 2022, 25, 29. [Google Scholar] [CrossRef] [PubMed]
  65. Global, regional, and national burden of colorectal cancer and its risk factors, 1990-2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Gastroenterol. Hepatol. 2022, 7, 627–647. [CrossRef] [PubMed]
  66. Pezzuto, A.; Carico, E. Role of HIF-1 in Cancer Progression: Novel Insights. A Review. Curr. Mol. Med. 2018, 18, 343–351. [Google Scholar] [CrossRef]
  67. Maruyama, A.; Mimura, J.; Itoh, K. Non-coding RNA derived from the region adjacent to the human HO-1 E2 enhancer selectively regulates HO-1 gene induction by modulating Pol II binding. Nucleic Acids Res. 2014, 42, 13599–13614. [Google Scholar] [CrossRef] [PubMed]
  68. Beckman, J.D.; Chen, C.; Nguyen, J.; Thayanithy, V.; Subramanian, S.; Steer, C.J.; Vercellotti, G.M. Regulation of heme oxygenase-1 protein expression by miR-377 in combination with miR-217. J. Biol. Chem. 2011, 286, 3194–3202. [Google Scholar] [CrossRef]
  69. Skrzypek, K.; Tertil, M.; Golda, S.; Ciesla, M.; Weglarczyk, K.; Collet, G.; Guichard, A.; Kozakowska, M.; Boczkowski, J.; Was, H.; et al. Interplay between heme oxygenase-1 and miR-378 affects non-small cell lung carcinoma growth, vascularization, and metastasis. Antioxid. Redox Signal. 2013, 19, 644–660. [Google Scholar] [CrossRef]
  70. Gao, N.; Li, Y.; Li, J.; Gao, Z.; Yang, Z.; Li, Y.; Liu, H.; Fan, T. Long Non-Coding RNAs: The Regulatory Mechanisms, Research Strategies, and Future Directions in Cancers. Front. Oncol. 2020, 10, 598817. [Google Scholar] [CrossRef] [PubMed]
  71. Bolton, E.M.; Tuzova, A.V.; Walsh, A.L.; Lynch, T.; Perry, A.S. Noncoding RNAs in Prostate Cancer: The Long and the Short of It. Clin. Cancer Res. 2014, 20, 35–43. [Google Scholar] [CrossRef]
  72. Bu, T.; Li, L.; Tian, J. Unlocking the role of non-coding RNAs in prostate cancer progression: Exploring the interplay with the Wnt signaling pathway. Front. Pharmacol. 2023, 14, 1269233. [Google Scholar] [CrossRef]
  73. Zinatizadeh, M.R.; Schock, B.; Chalbatani, G.M.; Zarandi, P.K.; Jalali, S.A.; Miri, S.R. The Nuclear Factor Kappa B (NF-kB) signaling in cancer development and immune diseases. Genes. Dis. 2021, 8, 287–297. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed]
  75. Alam, J.; Cook, J.L. How many transcription factors does it take to turn on the heme oxygenase-1 gene? Am. J. Respir. Cell Mol. Biol. 2007, 36, 166–174. [Google Scholar] [CrossRef]
  76. 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]
  77. Yang, C.-M.; Lin, C.-C.; Yang, C.-C.; Cho, R.-L.; Hsiao, L.-D. Mevastatin-Induced AP-1-Dependent HO-1 Expression Suppresses Vascular Cell Adhesion Molecule-1 Expression and Monocyte Adhesion on Human Pulmonary Alveolar Epithelial Cells Challenged with TNF-α. Biomolecules 2020, 10, 381. [Google Scholar] [CrossRef]
  78. Seo, S.H.; Jeong, G.S. Fisetin inhibits TNF-α-induced inflammatory action and hydrogen peroxide-induced oxidative damage in human keratinocyte HaCaT cells through PI3K/AKT/Nrf-2-mediated heme oxygenase-1 expression. Int. Immunopharmacol. 2015, 29, 246–253. [Google Scholar] [CrossRef] [PubMed]
  79. Cho, R.L.; Lin, W.N.; Wang, C.Y.; Yang, C.C.; Hsiao, L.D.; Lin, C.C.; Yang, C.M. Heme oxygenase-1 induction by rosiglitazone via PKCα/AMPKα/p38 MAPKα/SIRT1/PPARγ pathway suppresses lipopolysaccharide-mediated pulmonary inflammation. Biochem. Pharmacol. 2018, 148, 222–237. [Google Scholar] [CrossRef]
  80. Salloom, R.J.; Ahmad, I.M.; Abdalla, M.Y. Targeting heme degradation pathway augments prostate cancer cell sensitivity to docetaxel-induced apoptosis and attenuates migration. Front. Oncol. 2024, 14, 1431362. [Google Scholar] [CrossRef]
  81. Wardyn, J.D.; Ponsford, A.H.; Sanderson, C.M. Dissecting molecular cross-talk between Nrf2 and NF-κB response pathways. Biochem. Soc. Trans. 2015, 43, 621–626. [Google Scholar] [CrossRef]
  82. Mohamed, O.A.A.; Tesen, H.S.; Hany, M.; Sherif, A.; Abdelwahab, M.M.; Elnaggar, M.H. The role of hypoxia on prostate cancer progression and metastasis. Mol. Biol. Rep. 2023, 50, 3873–3884. [Google Scholar] [CrossRef] [PubMed]
  83. Muz, B.; de la Puente, P.; Azab, F.; Azab, A.K. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia 2015, 3, 83–92. [Google Scholar] [CrossRef] [PubMed]
  84. Fraga, A.; Ribeiro, R.; Príncipe, P.; Lopes, C.; Medeiros, R. Hypoxia and Prostate Cancer Aggressiveness: A Tale With Many Endings. Clin. Genitourin. Cancer 2015, 13, 295–301. [Google Scholar] [CrossRef] [PubMed]
  85. Stewart, G.D.; Ross, J.A.; McLaren, D.B.; Parker, C.C.; Habib, F.K.; Riddick, A.C. The relevance of a hypoxic tumour microenvironment in prostate cancer. BJU Int. 2010, 105, 8–13. [Google Scholar] [CrossRef]
  86. Muñoz-Sánchez, J.; Chánez-Cárdenas, M.E. A review on hemeoxygenase-2: Focus on cellular protection and oxygen response. Oxid. Med. Cell. Longev. 2014, 2014, 604981. [Google Scholar] [CrossRef]
  87. Neubauer, J.A.; Sunderram, J. Heme oxygenase-1 and chronic hypoxia. Respir. Physiol. Neurobiol. 2012, 184, 178–185. [Google Scholar] [CrossRef] [PubMed]
  88. Lee, P.J.; Jiang, B.H.; Chin, B.Y.; Iyer, N.V.; Alam, J.; Semenza, G.L.; Choi, A.M. Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia. J. Biol. Chem. 1997, 272, 5375–5381. [Google Scholar] [CrossRef]
  89. Kitamuro, T.; Takahashi, K.; Ogawa, K.; Udono-Fujimori, R.; Takeda, K.; Furuyama, K.; Nakayama, M.; Sun, J.; Fujita, H.; Hida, W.; et al. Bach1 functions as a hypoxia-inducible repressor for the heme oxygenase-1 gene in human cells. J. Biol. Chem. 2003, 278, 9125–9133. [Google Scholar] [CrossRef]
  90. Linnenbaum, M.; Busker, M.; Kraehling, J.R.; Behrends, S. Heme oxygenase isoforms differ in their subcellular trafficking during hypoxia and are differentially modulated by cytochrome P450 reductase. PLoS ONE 2012, 7, e35483. [Google Scholar] [CrossRef]
  91. Mbenza, N.M.; Nasarudin, N.; Vadakkedath, P.G.; Patel, K.; Ismail, A.Z.; Hanif, M.; Wright, L.J.; Sarojini, V.; Hartinger, C.G.; Leung, I.K.H. Carbon Monoxide is an Inhibitor of HIF Prolyl Hydroxylase Domain 2. ChemBioChem 2021, 22, 2521–2525. [Google Scholar] [CrossRef]
  92. Choi, Y.K.; Kim, C.K.; Lee, H.; Jeoung, D.; Ha, K.S.; Kwon, Y.G.; Kim, K.W.; Kim, Y.M. Carbon monoxide promotes VEGF expression by increasing HIF-1alpha protein level via two distinct mechanisms, translational activation and stabilization of HIF-1alpha protein. J. Biol. Chem. 2010, 285, 32116–32125. [Google Scholar] [CrossRef] [PubMed]
  93. Chin, B.Y.; Jiang, G.; Wegiel, B.; Wang, H.J.; Macdonald, T.; Zhang, X.C.; Gallo, D.; Cszimadia, E.; Bach, F.H.; Lee, P.J.; et al. Hypoxia-inducible factor 1alpha stabilization by carbon monoxide results in cytoprotective preconditioning. Proc. Natl. Acad. Sci. USA 2007, 104, 5109–5114. [Google Scholar] [CrossRef] [PubMed]
  94. Telarovic, I.; Wenger, R.H.; Pruschy, M. Interfering with Tumor Hypoxia for Radiotherapy Optimization. J. Exp. Clin. Cancer Res. 2021, 40, 197. [Google Scholar] [CrossRef] [PubMed]
  95. Emami Nejad, A.; Najafgholian, S.; Rostami, A.; Sistani, A.; Shojaeifar, S.; Esparvarinha, M.; Nedaeinia, R.; Haghjooy Javanmard, S.; Taherian, M.; Ahmadlou, M.; et al. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: A novel approach to developing treatment. Cancer Cell Int. 2021, 21, 62. [Google Scholar] [CrossRef] [PubMed]
  96. Shi, R.; Liao, C.; Zhang, Q. Hypoxia-Driven Effects in Cancer: Characterization, Mechanisms, and Therapeutic Implications. Cells 2021, 10, 678. [Google Scholar] [CrossRef] [PubMed]
  97. Yotnda, P.; Wu, D.; Swanson, A.M. Hypoxic tumors and their effect on immune cells and cancer therapy. Methods Mol. Biol. 2010, 651, 1–29. [Google Scholar] [CrossRef]
  98. Hassan Venkatesh, G.; Abou Khouzam, R.; Shaaban Moustafa Elsayed, W.; Ahmed Zeinelabdin, N.; Terry, S.; Chouaib, S. Tumor hypoxia: An important regulator of tumor progression or a potential modulator of tumor immunogenicity? Oncoimmunology 2021, 10, 1974233. [Google Scholar] [CrossRef]
  99. Schöning, J.P.; Monteiro, M.; Gu, W. Drug resistance and cancer stem cells: The shared but distinct roles of hypoxia-inducible factors HIF1α and HIF2α. Clin. Exp. Pharmacol. Physiol. 2017, 44, 153–161. [Google Scholar] [CrossRef]
  100. Moitra, K.; Lou, H.; Dean, M. Multidrug efflux pumps and cancer stem cells: Insights into multidrug resistance and therapeutic development. Clin. Pharmacol. Ther. 2011, 89, 491–502. [Google Scholar] [CrossRef]
  101. Gupte, A.; Mumper, R.J. Elevated copper and oxidative stress in cancer cells as a target for cancer treatment. Cancer Treat. Rev. 2009, 35, 32–46. [Google Scholar] [CrossRef]
  102. Riley, P.A. Free radicals in biology: Oxidative stress and the effects of ionizing radiation. Int. J. Radiat. Biol. 1994, 65, 27–33. [Google Scholar] [CrossRef] [PubMed]
  103. Rizvi, A.; Farhan, M.; Nabi, F.; Khan, R.H.; Adil, M.; Ahmad, A. Transcriptional Control of the Oxidative Stress Response and Implications of Using Plant Derived Molecules for Therapeutic Interventions in Cancer. Curr. Med. Chem. 2021, 28, 8480–8495. [Google Scholar] [CrossRef]
  104. Loreto Palacio, P.; Godoy, J.R.; Aktas, O.; Hanschmann, E.M. Changing Perspectives from Oxidative Stress to Redox Signaling-Extracellular Redox Control in Translational Medicine. Antioxidants 2022, 11, 1181. [Google Scholar] [CrossRef] [PubMed]
  105. Klaunig, J.E. Oxidative Stress and Cancer. Curr. Pharm. Des. 2018, 24, 4771–4778. [Google Scholar] [CrossRef] [PubMed]
  106. Oberley, L.W. Anticancer therapy by overexpression of superoxide dismutase. Antioxid. Redox Signal. 2001, 3, 461–472. [Google Scholar] [CrossRef]
  107. McCord, J.M.; Fridovich, I. Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J. Biol. Chem. 1969, 244, 6049–6055. [Google Scholar] [CrossRef] [PubMed]
  108. Aebi, H. Catalase in vitro. Methods Enzym. 1984, 105, 121–126. [Google Scholar] [CrossRef]
  109. Shimizu, T.; Iwanaga, M.; Yasunaga, A.; Urata, Y.; Goto, S.; Shibata, S.; Kondo, T. Protective role of glutathione synthesis on radiation-induced DNA damage in rabbit brain. Cell. Mol. Neurobiol. 1998, 18, 299–310. [Google Scholar] [CrossRef]
  110. Loboda, A.; Damulewicz, M.; Pyza, E.; Jozkowicz, A.; Dulak, J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: An evolutionarily conserved mechanism. Cell. Mol. Life Sci. 2016, 73, 3221–3247. [Google Scholar] [CrossRef]
  111. Yachie, A.; Niida, Y.; Wada, T.; Igarashi, N.; Kaneda, H.; Toma, T.; Ohta, K.; Kasahara, Y.; Koizumi, S. Oxidative stress causes enhanced endothelial cell injury in human heme oxygenase-1 deficiency. J. Clin. Invest. 1999, 103, 129–135. [Google Scholar] [CrossRef]
  112. Yachie, A. Heme Oxygenase-1 Deficiency and Oxidative Stress: A Review of 9 Independent Human Cases and Animal Models. Int. J. Mol. Sci. 2021, 22, 1514. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, N.; Wu, L.; Yuan, H.; Wang, J. ROS/Autophagy/Nrf2 Pathway Mediated Low-Dose Radiation Induced Radio-Resistance in Human Lung Adenocarcinoma A549 Cell. Int. J. Biol. Sci. 2015, 11, 833–844. [Google Scholar] [CrossRef] [PubMed]
  114. Sun, X.; Wang, S.; Gai, J.; Guan, J.; Li, J.; Li, Y.; Zhao, J.; Zhao, C.; Fu, L.; Li, Q. SIRT5 Promotes Cisplatin Resistance in Ovarian Cancer by Suppressing DNA Damage in a ROS-Dependent Manner via Regulation of the Nrf2/HO-1 Pathway. Front. Oncol. 2019, 9, 754. [Google Scholar] [CrossRef] [PubMed]
  115. Li, S.; Fujino, M.; Takahara, T.; Li, X.K. Protective role of heme oxygenase-1 in fatty liver ischemia-reperfusion injury. Med. Mol. Morphol. 2019, 52, 61–72. [Google Scholar] [CrossRef]
  116. Abraham, N.G.; Lin, J.H.; Dunn, M.W.; Schwartzman, M.L. Presence of heme oxygenase and NADPH cytochrome P-450 (c) reductase in human corneal epithelium. Investig. Ophthalmol. Vis. Sci. 1987, 28, 1464–1472. [Google Scholar]
  117. Lee, P.J.; Alam, J.; Wiegand, G.W.; Choi, A.M. Overexpression of heme oxygenase-1 in human pulmonary epithelial cells results in cell growth arrest and increased resistance to hyperoxia. Proc. Natl. Acad. Sci. USA 1996, 93, 10393–10398. [Google Scholar] [CrossRef]
  118. Stocker, R.; McDonagh, A.F.; Glazer, A.N.; Ames, B.N. Antioxidant activities of bile pigments: Biliverdin and bilirubin. Methods Enzym. 1990, 186, 301–309. [Google Scholar] [CrossRef]
  119. Baranano, D.E.; Rao, M.; Ferris, C.D.; Snyder, S.H. Biliverdin reductase: A major physiologic cytoprotectant. Proc. Natl. Acad. Sci. USA 2002, 99, 16093–16098. [Google Scholar] [CrossRef] [PubMed]
  120. Jansen, T.; Daiber, A. Direct Antioxidant Properties of Bilirubin and Biliverdin. Is there a Role for Biliverdin Reductase? Front. Pharmacol. 2012, 3, 30. [Google Scholar] [CrossRef]
  121. Ryter, S.W.; Alam, J.; Choi, A.M. Heme oxygenase-1/carbon monoxide: From basic science to therapeutic applications. Physiol. Rev. 2006, 86, 583–650. [Google Scholar] [CrossRef]
  122. Brouard, S.; Otterbein, L.E.; Anrather, J.; Tobiasch, E.; Bach, F.H.; Choi, A.M.; Soares, M.P. Carbon monoxide generated by heme oxygenase 1 suppresses endothelial cell apoptosis. J. Exp. Med. 2000, 192, 1015–1026. [Google Scholar] [CrossRef]
  123. Gutteridge, J.M. Iron promoters of the Fenton reaction and lipid peroxidation can be released from haemoglobin by peroxides. FEBS Lett. 1986, 201, 291–295. [Google Scholar] [CrossRef]
  124. Birrane, G.; Li, H.; Yang, S.; Tachado, S.D.; Seng, S. Cigarette smoke induces nuclear translocation of heme oxygenase 1 (HO-1) in prostate cancer cells: Nuclear HO-1 promotes vascular endothelial growth factor secretion. Int. J. Oncol. 2013, 42, 1919–1928. [Google Scholar] [CrossRef] [PubMed]
  125. Abdalla, M.Y.; Ahmad, I.M.; Switzer, B.; Britigan, B.E. Induction of heme oxygenase-1 contributes to survival of Mycobacterium abscessus in human macrophages-like THP-1 cells. Redox Biol. 2015, 4, 328–339. [Google Scholar] [CrossRef] [PubMed]
  126. Ahmad, I.M.; Dafferner, A.J.; O’Connell, K.A.; Mehla, K.; Britigan, B.E.; Hollingsworth, M.A.; Abdalla, M.Y. Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers 2021, 13, 2264. [Google Scholar] [CrossRef] [PubMed]
  127. Maines, M.D.; Abrahamsson, P.A. Expression of heme oxygenase-1 (HSP32) in human prostate: Normal, hyperplastic, and tumor tissue distribution. Urology 1996, 47, 727–733. [Google Scholar] [CrossRef]
  128. Blann, A.D.; Balakrishnan, B.; Ryan, P.; Lip, G.Y. Increased levels of plasma haemoxygenase-1 in prostate cancer. Prostate Cancer Prostatic Dis. 2011, 14, 114–117. [Google Scholar] [CrossRef]
  129. Alaoui-Jamali, M.A.; Bismar, T.A.; Gupta, A.; Szarek, W.A.; Su, J.; Song, W.; Xu, Y.; Xu, B.; Liu, G.; Vlahakis, J.Z.; et al. A Novel Experimental Heme Oxygenase-1–Targeted Therapy for Hormone-Refractory Prostate Cancer. Cancer Res. 2009, 69, 8017–8024. [Google Scholar] [CrossRef]
  130. Li, Y.; Su, J.; DingZhang, X.; Zhang, J.; Yoshimoto, M.; Liu, S.; Bijian, K.; Gupta, A.; Squire, J.A.; Alaoui Jamali, M.A.; et al. PTEN deletion and heme oxygenase-1 overexpression cooperate in prostate cancer progression and are associated with adverse clinical outcome. J. Pathol. 2011, 224, 90–100. [Google Scholar] [CrossRef] [PubMed]
  131. Sacca, P.; Meiss, R.; Casas, G.; Mazza, O.; Calvo, J.C.; Navone, N.; Vazquez, E. Nuclear translocation of haeme oxygenase-1 is associated to prostate cancer. Br. J. Cancer 2007, 97, 1683–1689. [Google Scholar] [CrossRef]
  132. Gueron, G.; De Siervi, A.; Ferrando, M.; Salierno, M.; De Luca, P.; Elguero, B.; Meiss, R.; Navone, N.; Vazquez, E.S. Critical role of endogenous heme oxygenase 1 as a tuner of the invasive potential of prostate cancer cells. Mol. Cancer Res. 2009, 7, 1745–1755. [Google Scholar] [CrossRef] [PubMed]
  133. Miller, D.R.; Ingersoll, M.A.; Chou, Y.W.; Kosmacek, E.A.; Oberley-Deegan, R.E.; Lin, M.F. Dynamics of antioxidant heme oxygenase-1 and pro-oxidant p66Shc in promoting advanced prostate cancer progression. Free Radic. Biol. Med. 2022, 193, 274–291. [Google Scholar] [CrossRef]
  134. Nakayama, T.; Kobayashi, T.; Shimpei, O.; Fukuhara, H.; Namikawa, T.; Inoue, K.; Hanazaki, K.; Takahashi, K.; Nakajima, M.; Tanaka, T.; et al. Photoirradiation after aminolevulinic acid treatment suppresses cancer cell proliferation through the HO-1/p21 pathway. Photodiagnosis Photodyn. Ther. 2019, 28, 10–17. [Google Scholar] [CrossRef] [PubMed]
  135. Podkalicka, P.; Mucha, O.; Józkowicz, A.; Dulak, J.; Łoboda, A. Heme oxygenase inhibition in cancers: Possible tools and targets. Contemp. Oncol. (Pozn.) 2018, 22, 23–32. [Google Scholar] [CrossRef]
  136. Romeo, G.; Ciaffaglione, V.; Amata, E.; Dichiara, M.; Calabrese, L.; Vanella, L.; Sorrenti, V.; Grosso, S.; D’Amico, A.G.; D’Agata, V.; et al. Combination of Heme Oxygenase-1 Inhibition and Sigma Receptor Modulation for Anticancer Activity. Molecules 2021, 26, 3860. [Google Scholar] [CrossRef] [PubMed]
  137. Mucha, O.; Podkalicka, P.; Mikulski, M.; Barwacz, S.; Andrysiak, K.; Biela, A.; Mieczkowski, M.; Kachamakova-Trojanowska, N.; Ryszawy, D.; Białas, A.; et al. Development and characterization of a new inhibitor of heme oxygenase activity for cancer treatment. Arch. Biochem. Biophys. 2019, 671, 130–142. [Google Scholar] [CrossRef] [PubMed]
  138. Baldessari, C.; Pipitone, S.; Molinaro, E.; Cerma, K.; Fanelli, M.; Nasso, C.; Oltrecolli, M.; Pirola, M.; D’Agostino, E.; Pugliese, G.; et al. Bone Metastases and Health in Prostate Cancer: From Pathophysiology to Clinical Implications. Cancers 2023, 15, 1518. [Google Scholar] [CrossRef]
  139. Zhang, A.C.; Rasul, R.; Golden, A.; Feuerstein, M.A. Incidence and mortality trends of metastatic prostate cancer: Surveillance, Epidemiology, and End Results database analysis. Can. Urol. Assoc. J. 2021, 15, E637–E643. [Google Scholar] [CrossRef] [PubMed]
  140. Lange, J.M.; Laviana, A.A.; Penson, D.F.; Lin, D.W.; Bill-Axelson, A.; Carlsson, S.V.; Newcomb, L.F.; Trock, B.J.; Carter, H.B.; Carroll, P.R.; et al. Prostate cancer mortality and metastasis under different biopsy frequencies in North American active surveillance cohorts. Cancer 2020, 126, 583–592. [Google Scholar] [CrossRef] [PubMed]
  141. Zhang, A.; Suzuki, T.; Adachi, S.; Naganuma, E.; Suzuki, N.; Hosoya, T.; Itoh, K.; Sporn, M.B.; Yamamoto, M. Distinct Regulations of HO-1 Gene Expression for Stress Response and Substrate Induction. Mol. Cell. Biol. 2021, 41, e0023621. [Google Scholar] [CrossRef]
  142. Ferrando, M.; Wan, X.; Meiss, R.; Yang, J.; De Siervi, A.; Navone, N.; Vazquez, E. Heme oxygenase-1 (HO-1) expression in prostate cancer cells modulates the oxidative response in bone cells. PLoS ONE 2013, 8, e80315. [Google Scholar] [CrossRef] [PubMed]
  143. Consoli, V.; Sorrenti, V.; Grosso, S.; Vanella, L. Heme Oxygenase-1 Signaling and Redox Homeostasis in Physiopathological Conditions. Biomolecules 2021, 11, 589. [Google Scholar] [CrossRef] [PubMed]
  144. Choi, Y.K.; Kim, Y.M. Regulation of Endothelial and Vascular Functions by Carbon Monoxide via Crosstalk With Nitric Oxide. Front. Cardiovasc. Med. 2021, 8, 649630. [Google Scholar] [CrossRef] [PubMed]
  145. Stefan, W.R.; Augustine, M.K.C. Targeting heme oxygenase-1 and carbon monoxide for therapeutic modulation of inflammation. Transl. Res. 2016, 167, 7–34. [Google Scholar] [CrossRef]
  146. Agnieszka, L.; Alicja, J.; Jozef, D. HO-1/CO system in tumor growth, angiogenesis and metabolism—Targeting HO-1 as an anti-tumor therapy. Vasc. Pharmacol. 2015, 74, 11–22. [Google Scholar] [CrossRef]
  147. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef]
  148. Teleanu, R.I.; Chircov, C.; Grumezescu, A.M.; Teleanu, D.M. Tumor Angiogenesis and Anti-Angiogenic Strategies for Cancer Treatment. J. Clin. Med. 2019, 9, 84. [Google Scholar] [CrossRef]
  149. Tsai, C.F.; Chen, J.H.; Chang, C.N.; Lu, D.Y.; Chang, P.C.; Wang, S.L.; Yeh, W.L. Fisetin inhibits cell migration via inducing HO-1 and reducing MMPs expression in breast cancer cell lines. Food Chem. Toxicol. 2018, 120, 528–535. [Google Scholar] [CrossRef]
  150. Gong, Y.; Chippada-Venkata, U.D.; Oh, W.K. Roles of matrix metalloproteinases and their natural inhibitors in prostate cancer progression. Cancers 2014, 6, 1298–1327. [Google Scholar] [CrossRef]
  151. Goncharov, A.P.; Vashakidze, N.; Kharaishvili, G. Epithelial-Mesenchymal Transition: A Fundamental Cellular and Microenvironmental Process in Benign and Malignant Prostate Pathologies. Biomedicines 2024, 12, 418. [Google Scholar] [CrossRef]
  152. Zhao, Z.; Zhao, J.; Xue, J.; Zhao, X.; Liu, P. Autophagy inhibition promotes epithelial-mesenchymal transition through ROS/HO-1 pathway in ovarian cancer cells. Am. J. Cancer Res. 2016, 6, 2162–2177. [Google Scholar] [PubMed]
  153. Adamo, H.H.; Strömvall, K.; Nilsson, M.; Halin Bergström, S.; Bergh, A. Adaptive (TINT) Changes in the Tumor Bearing Organ Are Related to Prostate Tumor Size and Aggressiveness. PLoS ONE 2015, 10, e0141601. [Google Scholar] [CrossRef]
  154. Trujillo, K.A.; Jones, A.C.; Griffith, J.K.; Bisoffi, M. Markers of field cancerization: Proposed clinical applications in prostate biopsies. Prostate Cancer 2012, 2012, 302894. [Google Scholar] [CrossRef]
  155. Zhou, Y.; Zhang, Y.; Gong, H.; Luo, S.; Cui, Y. The Role of Exosomes and Their Applications in Cancer. Int. J. Mol. Sci. 2021, 22, 12204. [Google Scholar] [CrossRef] [PubMed]
  156. Jiang, Y.; Zhao, H.; Chen, Y.; Li, K.; Li, T.; Chen, J.; Zhang, B.; Guo, C.; Qing, L.; Shen, J.; et al. Exosomal long noncoding RNA HOXD-AS1 promotes prostate cancer metastasis via miR-361-5p/FOXM1 axis. Cell Death Dis. 2021, 12, 1129. [Google Scholar] [CrossRef] [PubMed]
  157. Tai, Y.L.; Chen, K.C.; Hsieh, J.T.; Shen, T.L. Exosomes in cancer development and clinical applications. Cancer Sci. 2018, 109, 2364–2374. [Google Scholar] [CrossRef]
  158. Dai, J.; Su, Y.; Zhong, S.; Cong, L.; Liu, B.; Yang, J.; Tao, Y.; He, Z.; Chen, C.; Jiang, Y. Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct. Target. Ther. 2020, 5, 145. [Google Scholar] [CrossRef]
  159. Ben-Eltriki, M.; Gayle, E.J.; Walker, N.; Deb, S. Pharmacological Significance of Heme Oxygenase 1 in Prostate Cancer. Curr. Issues Mol. Biol. 2023, 45, 4301–4316. [Google Scholar] [CrossRef]
  160. Wang, H.; Cheng, Q.; Bao, L.; Li, M.; Chang, K.; Yi, X. Cytoprotective Role of Heme Oxygenase-1 in Cancer Chemoresistance: Focus on Antioxidant, Antiapoptotic, and Pro-Autophagy Properties. Antioxidants 2023, 12, 1217. [Google Scholar] [CrossRef] [PubMed]
  161. Chung, C.; Abboud, K. Targeting the androgen receptor signaling pathway in advanced prostate cancer. Am. J. Health Syst. Pharm. 2022, 79, 1224–1235. [Google Scholar] [CrossRef]
  162. Huang, J.; Lin, B.; Li, B. Anti-Androgen Receptor Therapies in Prostate Cancer: A Brief Update and Perspective. Front. Oncol. 2022, 12, 865350. [Google Scholar] [CrossRef] [PubMed]
  163. Turco, F.; Gillessen, S.; Cathomas, R.; Buttigliero, C.; Vogl, U.M. Treatment Landscape for Patients with Castration-Resistant Prostate Cancer: Patient Selection and Unmet Clinical Needs. Res. Rep. Urol. 2022, 14, 339–350. [Google Scholar] [CrossRef] [PubMed]
  164. Spratt, D.E.; Shore, N.; Sartor, O.; Rathkopf, D.; Olivier, K. Treating the patient and not just the cancer: Therapeutic burden in prostate cancer. Prostate Cancer Prostatic Dis. 2021, 24, 647–661. [Google Scholar] [CrossRef] [PubMed]
  165. Mondal, D.; Narwani, D.; Notta, S.; Ghaffar, D.; Mardhekar, N.; Quadri, S.S.A. Oxidative stress and redox signaling in CRPC progression: Therapeutic potential of clinically-tested Nrf2-activators. Cancer Drug Resist. 2021, 4, 96–124. [Google Scholar] [CrossRef] [PubMed]
  166. Luo, X.-h.; Liu, J.-z.; Wang, B.; Men, Q.-l.; Ju, Y.-q.; Yin, F.-y.; Zheng, C.; Li, W. KLF14 potentiates oxidative adaptation via modulating HO-1 signaling in castrate-resistant prostate cancer. Endocr.-Relat. Cancer 2019, 26, 181–195. [Google Scholar] [CrossRef]
  167. Bolla, M.; Henry, A.; Mason, M.; Wiegel, T. The role of radiotherapy in localised and locally advanced prostate cancer. Asian J. Urol. 2019, 6, 153–161. [Google Scholar] [CrossRef]
  168. Lyu, F.; Shang, S.-Y.; Gao, X.-S.; Ma, M.-W.; Xie, M.; Ren, X.-Y.; Liu, M.-Z.; Chen, J.-Y.; Li, S.-S.; Huang, L. Uncovering the Secrets of Prostate Cancer’s Radiotherapy Resistance: Advances in Mechanism Research. Biomedicines 2023, 11, 1628. [Google Scholar] [CrossRef]
  169. Ashrafizadeh, M.; Zhang, W.; Tian, Y.; Sethi, G.; Zhang, X.; Qiu, A. Molecular panorama of therapy resistance in prostate cancer: A pre-clinical and bioinformatics analysis for clinical translation. Cancer Metastasis Rev. 2024, 43, 229–260. [Google Scholar] [CrossRef] [PubMed]
  170. Zhang, W.; Qiao, T.; Zha, L. Inhibition of heme oxygenase-1 enhances the radiosensitivity in human nonsmall cell lung cancer a549 cells. Cancer Biother. Radiopharm. 2011, 26, 639–645. [Google Scholar] [CrossRef]
  171. Nitti, M.; Ortolan, J.; Furfaro, A.L. Role of heme oxygenase-1 in tumor immune escape. Redox Exp. Med. 2023, 2023, e230006. [Google Scholar] [CrossRef]
  172. Nemeth, Z.; Li, M.; Csizmadia, E.; Döme, B.; Johansson, M.; Liao Persson, J.; Seth, P.; Otterbein, L.; Wegiel, B. Heme oxygenase-1 in macrophages controls prostate cancer progression. Oncotarget 2015, 6, 33675. [Google Scholar] [CrossRef] [PubMed]
  173. Muliaditan, T.; Caron, J.; Okesola, M.; Opzoomer, J.W.; Kosti, P.; Georgouli, M.; Gordon, P.; Lall, S.; Kuzeva, D.M.; Pedro, L.; et al. Macrophages are exploited from an innate wound healing response to facilitate cancer metastasis. Nat. Commun. 2018, 9, 2951. [Google Scholar] [CrossRef]
  174. Nitti, M.; Piras, S.; Marinari, U.M.; Moretta, L.; Pronzato, M.A.; Furfaro, A.L. HO-1 Induction in Cancer Progression: A Matter of Cell Adaptation. Antioxidants 2017, 6, 29. [Google Scholar] [CrossRef]
  175. Orecchioni, M.; Ghosheh, Y.; Pramod, A.B.; Ley, K. Macrophage Polarization: Different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. Alternatively Activated Macrophages. Front. Immunol. 2019, 10, 1084. [Google Scholar] [CrossRef]
  176. Weis, N.; Weigert, A.; von Knethen, A.; Brüne, B. Heme oxygenase-1 contributes to an alternative macrophage activation profile induced by apoptotic cell supernatants. Mol. Biol. Cell 2009, 20, 1280–1288. [Google Scholar] [CrossRef]
  177. Alaluf, E.; Vokaer, B.; Detavernier, A.; Azouz, A.; Splittgerber, M.; Carrette, A.; Boon, L.; Libert, F.; Soares, M.; Le Moine, A.; et al. Heme oxygenase-1 orchestrates the immunosuppressive program of tumor-associated macrophages. JCI Insight 2020, 5, e133929. [Google Scholar] [CrossRef]
  178. Arnold, J.N.; Magiera, L.; Kraman, M.; Fearon, D.T. Tumoral immune suppression by macrophages expressing fibroblast activation protein-α and heme oxygenase-1. Cancer Immunol. Res. 2014, 2, 121–126. [Google Scholar] [CrossRef] [PubMed]
  179. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef] [PubMed]
  180. Wolchok, J.D.; Kluger, H.; Callahan, M.K.; Postow, M.A.; Rizvi, N.A.; Lesokhin, A.M.; Segal, N.H.; Ariyan, C.E.; Gordon, R.A.; Reed, K.; et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 2013, 369, 122–133. [Google Scholar] [CrossRef]
  181. George, S.D.; Jeffrey, B.; Menachem, G.; David, L.; Nader, G.A. HO-1 overexpression and underexpression: Clinical implications. Arch. Biochem. Biophys. 2019, 673, 108073. [Google Scholar] [CrossRef]
  182. Alaoui-Jamali, M.; Szarek, W.; Nakatsu, K.; Vlahakis, J.; Gupta, A.; Schipper, H. OB-24, a novel selective and potent HO-1 inhibitor, induces a wide spectrum anti-tumor activity in vitro and in vivo and synergizes with chemotherapy drugs. Mol. Cancer Ther. 2007, 6, C82. [Google Scholar]
  183. Abdelrahim, M.; Safe, S.; Baker, C.; Abudayyeh, A. RNAi and cancer: Implications and applications. J. RNAi Gene Silenc. 2006, 2, 136–145. [Google Scholar]
  184. Kim, H.R.; Kim, S.; Kim, E.J.; Park, J.H.; Yang, S.H.; Jeong, E.T.; Park, C.; Youn, M.J.; So, H.S.; Park, R. Suppression of Nrf2-driven heme oxygenase-1 enhances the chemosensitivity of lung cancer A549 cells toward cisplatin. Lung Cancer 2008, 60, 47–56. [Google Scholar] [CrossRef]
  185. Paez, A.V.; Pallavicini, C.; Schuster, F.; Valacco, M.P.; Giudice, J.; Ortiz, E.G.; Anselmino, N.; Labanca, E.; Binaghi, M.; Salierno, M.; et al. Heme oxygenase-1 in the forefront of a multi-molecular network that governs cell-cell contacts and filopodia-induced zippering in prostate cancer. Cell Death Dis. 2016, 7, e2570. [Google Scholar] [CrossRef]
  186. Drummond, G.S.; Kappas, A. Chemoprevention of Neonatal Jaundice: Potency of Tin-Protoporphyrin in an Animal Model. Science 1982, 217, 1250–1252. [Google Scholar] [CrossRef]
  187. Fernández-Fierro, A.; Funes, S.C.; Rios, M.; Covián, C.; González, J.; Kalergis, A.M. Immune Modulation by Inhibitors of the HO System. Int. J. Mol. Sci. 2021, 22, 294. [Google Scholar] [CrossRef]
  188. Bhutani, V.K.; Poland, R.; Meloy, L.D.; Hegyi, T.; Fanaroff, A.A.; Maisels, M.J. Clinical trial of tin mesoporphyrin to prevent neonatal hyperbilirubinemia. J. Perinatol. 2016, 36, 533–539. [Google Scholar] [CrossRef]
  189. Costa, D.L.; Namasivayam, S.; Amaral, E.P.; Arora, K.; Chao, A.; Mittereder, L.R.; Maiga, M.; Boshoff, H.I.; Barry, C.E.; Goulding, C.W.; et al. Pharmacological Inhibition of Host Heme Oxygenase-1 Suppresses Mycobacterium tuberculosis Infection <i>In Vivo</i> by a Mechanism Dependent on T Lymphocytes. mBio 2016, 7, 10–1128. [Google Scholar] [CrossRef]
  190. Andrade, B.B.; Pavan Kumar, N.; Amaral, E.P.; Riteau, N.; Mayer-Barber, K.D.; Tosh, K.W.; Maier, N.; Conceição, E.L.; Kubler, A.; Sridhar, R.; et al. Heme Oxygenase-1 Regulation of Matrix Metalloproteinase-1 Expression Underlies Distinct Disease Profiles in Tuberculosis. J. Immunol. 2015, 195, 2763–2773. [Google Scholar] [CrossRef]
  191. Fernández-Mendívil, C.; Arreola, M.A.; Hohsfield, L.A.; Green, K.N.; Lopez, M.G. Aging and Progression of Beta-Amyloid Pathology in Alzheimer’s Disease Correlates with Microglial Heme-Oxygenase-1 Overexpression. Antioxidants 2020, 9, 644. [Google Scholar] [CrossRef] [PubMed]
  192. Stahnke, T.; Richter-Landsberg, C.; Stadelmann, C.; Netzler, A.; Brück, W. Differential upregulation of heme oxygenase-1 (HSP32) in glial cells after oxidative stress and in demyelinating disorders. J. Mol. Neurosci. 2007, 32, 25–37. [Google Scholar] [CrossRef] [PubMed]
  193. Lilljebjörn, L.V.; Csizmadia, E.; Hedblom, A.; Canesin, G.; Kalbasi, A.; Li, M.; Kramer, F.; Bornfeldt, K.E.; Wegiel, B. A Role of the Heme Degradation Pathway in Shaping Prostate Inflammatory Responses and Lipid Metabolism. Am. J. Pathol. 2020, 190, 830–843. [Google Scholar] [CrossRef]
  194. Wu, B.; Wu, Y.; Tang, W. Heme Catabolic Pathway in Inflammation and Immune Disorders. Front. Pharmacol. 2019, 10, 825. [Google Scholar] [CrossRef]
  195. Tsai, J.R.; Wang, H.M.; Liu, P.L.; Chen, Y.H.; Yang, M.C.; Chou, S.H.; Cheng, Y.J.; Yin, W.H.; Hwang, J.J.; Chong, I.W. High expression of heme oxygenase-1 is associated with tumor invasiveness and poor clinical outcome in non-small cell lung cancer patients. Cell. Oncol. (Dordr.) 2012, 35, 461–471. [Google Scholar] [CrossRef]
Figure 1. Diagnosis of PC. The diagnosis begins with the measurement of PSA levels in the blood, where elevated levels may indicate the presence of PC. If elevated PSA levels are detected, a biopsy is performed on prostate tissues, and a Gleason score is assigned to determine the aggressiveness of the disease. The biopsy results can differentiate between BPH and cancer. Tumor stage, Gleason score, and PSA levels determine the treatment strategy, which may include radical prostatectomy, hormonal therapy, radiotherapy, or chemotherapy. (Created with BioRender.com).
Figure 1. Diagnosis of PC. The diagnosis begins with the measurement of PSA levels in the blood, where elevated levels may indicate the presence of PC. If elevated PSA levels are detected, a biopsy is performed on prostate tissues, and a Gleason score is assigned to determine the aggressiveness of the disease. The biopsy results can differentiate between BPH and cancer. Tumor stage, Gleason score, and PSA levels determine the treatment strategy, which may include radical prostatectomy, hormonal therapy, radiotherapy, or chemotherapy. (Created with BioRender.com).
Ijms 25 09195 g001
Figure 2. Cellular Localization and Function of HO-1. HO-1 is upregulated by various stimuli, including hypoxia, ROS, oxidative stress, and heme. HO-1 is initially synthesized and localized to the endoplasmic reticulum (ER), where it catalyzes the degradation of heme. This process produces biliverdin, which is subsequently converted to bilirubin by biliverdin reductase A (BVRA), carbon monoxide (CO), and ferrous iron (Fe2+). Ferrous iron is quickly sequestered into ferritin for storage. Both biliverdin and bilirubin serve as potent antioxidants, while CO exerts cytoprotective effects by activating anti-inflammatory and anti-apoptotic pathways. Under stress conditions, HO-1 undergoes proteolytic cleavage (t-HO-1) and translocates to the nucleus, where it regulates the expression of genes related to antioxidants, angiogenesis, metastasis, anti-apoptosis, and other crucial cellular processes. (Created with BioRender.com).
Figure 2. Cellular Localization and Function of HO-1. HO-1 is upregulated by various stimuli, including hypoxia, ROS, oxidative stress, and heme. HO-1 is initially synthesized and localized to the endoplasmic reticulum (ER), where it catalyzes the degradation of heme. This process produces biliverdin, which is subsequently converted to bilirubin by biliverdin reductase A (BVRA), carbon monoxide (CO), and ferrous iron (Fe2+). Ferrous iron is quickly sequestered into ferritin for storage. Both biliverdin and bilirubin serve as potent antioxidants, while CO exerts cytoprotective effects by activating anti-inflammatory and anti-apoptotic pathways. Under stress conditions, HO-1 undergoes proteolytic cleavage (t-HO-1) and translocates to the nucleus, where it regulates the expression of genes related to antioxidants, angiogenesis, metastasis, anti-apoptosis, and other crucial cellular processes. (Created with BioRender.com).
Ijms 25 09195 g002
Figure 3. The regulation of HO-1 in PC cells. Multiple pathways regulate HO-1 expression in PC cells. The NRF2 pathway is crucial, where under oxidative stress, NRF2 dissociates from KEAP1, translocates to the nucleus, binds to ARE in the HO-1 promotor region, and activates transcription. The HIF-1α pathway also activates HO-1 transcription, under hypoxic conditions, HIF-1α translocates to the nucleus where it dimerizes with HIF-1β to form the HIF-1 complex, which binds to HREs in the HO-1 promotor. Additionally, inflammation activates NF-ƙB pathway, which translocates to the nucleus and binds the HO-1 promotor, inducing transcription. The MAPK pathway promote HO-1 expression through intermediate signaling pathways, including the activation of AP-1 transcription factor, which binds to the HO-1 promotor, inducing transcription. (Created with BioRender.com).
Figure 3. The regulation of HO-1 in PC cells. Multiple pathways regulate HO-1 expression in PC cells. The NRF2 pathway is crucial, where under oxidative stress, NRF2 dissociates from KEAP1, translocates to the nucleus, binds to ARE in the HO-1 promotor region, and activates transcription. The HIF-1α pathway also activates HO-1 transcription, under hypoxic conditions, HIF-1α translocates to the nucleus where it dimerizes with HIF-1β to form the HIF-1 complex, which binds to HREs in the HO-1 promotor. Additionally, inflammation activates NF-ƙB pathway, which translocates to the nucleus and binds the HO-1 promotor, inducing transcription. The MAPK pathway promote HO-1 expression through intermediate signaling pathways, including the activation of AP-1 transcription factor, which binds to the HO-1 promotor, inducing transcription. (Created with BioRender.com).
Ijms 25 09195 g003
Figure 4. The Role of HO-1 in PC Progression. In hypoxic tumors, HIF-1α is activated, which in turn upregulates HO-1 expression. This upregulation triggers pathways that promote PC invasion, tumorigenesis, metastasis, and therapy resistance. HO-1 plays a key role in mitigating the effects of elevated ROS levels in PC cells by activating survival pathways and inhibiting apoptosis. Inhibition of HO-1 using SnPP or ZnPP results in increased ROS levels, leading to cancer cell apoptosis. Additionally, HO-1 facilitates PC metastasis by promoting angiogenesis, enhancing MMP activity, and driving EMT processes. HO-1 also exerts immunomodulatory effects, where elevated HO-1 levels are associated with the anti-inflammatory and tumor-promoting M2 phenotype. Conversely, inhibiting HO-1 activity shifts macrophage polarization towards the M1 phenotype, which is pro-inflammatory and anti-tumorigenic (arrows indicate enhancement or inhibition). (Created with BioRender.com).
Figure 4. The Role of HO-1 in PC Progression. In hypoxic tumors, HIF-1α is activated, which in turn upregulates HO-1 expression. This upregulation triggers pathways that promote PC invasion, tumorigenesis, metastasis, and therapy resistance. HO-1 plays a key role in mitigating the effects of elevated ROS levels in PC cells by activating survival pathways and inhibiting apoptosis. Inhibition of HO-1 using SnPP or ZnPP results in increased ROS levels, leading to cancer cell apoptosis. Additionally, HO-1 facilitates PC metastasis by promoting angiogenesis, enhancing MMP activity, and driving EMT processes. HO-1 also exerts immunomodulatory effects, where elevated HO-1 levels are associated with the anti-inflammatory and tumor-promoting M2 phenotype. Conversely, inhibiting HO-1 activity shifts macrophage polarization towards the M1 phenotype, which is pro-inflammatory and anti-tumorigenic (arrows indicate enhancement or inhibition). (Created with BioRender.com).
Ijms 25 09195 g004
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

Salloom, R.J.; Ahmad, I.M.; Sahtout, D.Z.; Baine, M.J.; Abdalla, M.Y. Heme Oxygenase-1 and Prostate Cancer: Function, Regulation, and Implication in Cancer Therapy. Int. J. Mol. Sci. 2024, 25, 9195. https://doi.org/10.3390/ijms25179195

AMA Style

Salloom RJ, Ahmad IM, Sahtout DZ, Baine MJ, Abdalla MY. Heme Oxygenase-1 and Prostate Cancer: Function, Regulation, and Implication in Cancer Therapy. International Journal of Molecular Sciences. 2024; 25(17):9195. https://doi.org/10.3390/ijms25179195

Chicago/Turabian Style

Salloom, Ramia J., Iman M. Ahmad, Dania Z. Sahtout, Michael J. Baine, and Maher Y. Abdalla. 2024. "Heme Oxygenase-1 and Prostate Cancer: Function, Regulation, and Implication in Cancer Therapy" International Journal of Molecular Sciences 25, no. 17: 9195. https://doi.org/10.3390/ijms25179195

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