Next Article in Journal
Towards the Development of Novel, Point-of-Care Assays for Monitoring Different Forms of Antioxidant Activity: The RoXstaTM System
Previous Article in Journal
Cecal Microbial Diversity and Metabolome Reveal a Reduction in Growth Due to Oxidative Stress Caused by a Low-Energy Diet in Donkeys
Previous Article in Special Issue
Mitochondrial Quality Control in Alzheimer’s Disease: Insights from Caenorhabditis elegans Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer’s Disease: Beneficial or Detrimental?

1
Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan
2
College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
3
Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
4
Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
5
Department of Neurology, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan
6
Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
7
Department of Neurology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
8
Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
9
Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2024, 13(11), 1378; https://doi.org/10.3390/antiox13111378
Submission received: 30 September 2024 / Revised: 4 November 2024 / Accepted: 7 November 2024 / Published: 11 November 2024
(This article belongs to the Special Issue Oxidative Stress and Alzheimer’s Disease)

Abstract

:
The major pathological characteristics of Alzheimer’s disease (AD) include senile plaques and neurofibrillary tangles (NFTs), which are mainly composed of aggregated amyloid-beta (Aβ) peptide and hyperphosphorylated tau protein, respectively. The excessive production of reactive oxygen species (ROS) and neuroinflammation are crucial contributing factors to the pathological mechanisms of AD. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor critical for tissue adaption to low-oxygen tension. Growing evidence has suggested HIF-1 as a potential therapeutic target for AD; conversely, other experimental findings indicate that HIF-1 induction contributes to AD pathogenesis. These previous findings thus point to the complex, even contradictory, roles of HIF-1 in AD. In this review, we first introduce the general pathogenic mechanisms of AD as well as the potential pathophysiological roles of HIF-1 in cancer, immunity, and oxidative stress. Based on current experimental evidence in the literature, we then discuss the possible beneficial as well as detrimental mechanisms of HIF-1 in AD; these sections also include the summaries of multiple chemical reagents and proteins that have been shown to exert beneficial effects in AD via either the induction or inhibition of HIF-1.

1. Pathophysiology of Alzheimer’s Disease

Alzheimer’s disease (AD) is the most common age-related neurodegenerative disease and presents with progressive memory impairment, declined cognition, and behavioral dysfunctions. Currently no effective treatment for the deteriorating clinical course of dementia is available, thus contributing substantial economic burdens to the patients’ families and eventually to the whole society [1,2,3]. According to the World Health Organization (WHO), around 55 million people suffer from dementia worldwide, with approximately 10 million cases added each year. These situations underscore dementia as one of the leading causes of mental and/or physical disability. Among various causes, AD may comprise 60–70% of dementia cases (https://www.who.int/news-room/fact-sheets/detail/dementia, accessed on 15 March 2023). Based on the report “2023 Alzheimer’s disease facts and figures” [4], nearly 6.7 million Americans aged 65 years and above are estimated to suffer from AD. By 2060, the number of AD patients, with a rather steep rise, may increase to 13.8 million. Comparing the death tolls from 2000 to 2019 among various diseases, death caused by human immunodeficiency virus, cardiac diseases, and cerebrovascular disorders declined while those from AD increased by more than 145% [5]. The total expenses for AD healthcare in 2023 were around USD 345 billion and are predicted to escalate to more than USD 1 trillion by 2050 due to the aging population [4,5].
The major pathological features in AD include atrophy of the brain [6], deposition of amyloid-beta (Aβ) plaques, and the presence of neurofibrillary tangles (NFTs) in axonal microtubules [7,8,9] (Figure 1). Aβ is a neurotoxic peptide fragment of 39–43 amino acids derived from the sequential cleavage of amyloid precursor protein (APP) by a group of enzymes called secretases [10,11]. In non-amyloidogenic pathways, most of the APP undergoes cleavage by α-secretase to yield the extracellular soluble APP-α fragment (sAPPα) and C-terminal fragment-α (α-CTF or C83); the latter is then subjected to γ-secretase-mediated proteolysis to produce p3 and an APP intracellular domain (AICD) fragment [12]. Alternatively, in the amyloidogenic pathway, a minor population of APP may first undergo β-secretase cleavage to generate sAPPβ (β-CTF or C99), followed by γ-secretase-mediated cleavage to produce Aβ and AICD [12]. Excessive Aβ aggregation can exert neurotoxicity through diverse mechanisms including excitotoxicity [13], oxidative stress [14], aberrant cell cycle reentry [15,16], mitochondrial dysfunction [17], and impaired DNA function [18], together contributing to the damage or even demise of the neurons.
Tau, a member of the microtubule-associated protein family crucial for microtubule assembly and stabilization, is involved in axonal transport in neurons [19]. The hyperphosphorylation of tau proteins not only impairs the biological functions, with resultant compromised microtubule structures and disturbed axonal transport, but also causes aberrant aggregation leading to neuronal damage [19]. Moreover, emerging evidence indicates that Aβ may also impair axonal transport and further aggravate the AD pathophysiology [20]. Additionally, Aβ can affect gene transcription and alter protein expression that may change the fate of neuronal survival or death under AD-related pathophysiological conditions [17]. Thus, any means to delay or lessen the consequences from AD may mitigate the socioeconomic impacts on the patients’ families and societies.

2. The Pathophysiological Roles of HIF-1 in Cancer, Immunity, and Oxidative Stress

In mammalian tissues, three hypoxia-inducible factors (HIFs) including HIF-1, HIF-2, and HIF-3 have been reported, whose expression levels are strictly regulated by oxygen tension [21]. The activation of HIF-1 is crucial for numerous cellular responses to adapt the tissues to hypoxic environments, such as increasing cell proliferation, accelerating the formation of new blood vessels, regulating glucose and energy metabolisms, affecting the synthesis of glycogen and fatty acids, and adjusting the pH level [22,23,24]. Besides HIF-1, HIF-2 is expressed in a cell-specific manner that plays a pivotal role in pulmonary development, vascularization, and erythropoiesis [25,26]. While HIF-1 and HIF-2 may function as transcription factors to drive the expression of either distinctive or overlapping target genes, the function of HIF-3 is less well understood [27]. In the present article, we only focus on the potential roles of HIF-1 in AD.
HIF-1 consists of an alpha subunit (HIF-1α) that senses the low-oxygen tension for its stabilization and a beta subunit (HIF-1β) that is constitutively expressed [28,29]. The half-life of HIF-1α in living cells under normoxic condition is only minutes [28]. This rapid degradation of HIF-1α occurs through oxygen-dependent hydroxylation catalyzed by the proteins of the prolyl hydroxylase domain (PHD) family; the hydroxylation of HIF-1α promotes binding to an E3 ubiquitin ligase, which contains the von Hippel–Lindau (VHL) protein, to facilitate its polyubiquitination and subsequent proteasomal degradation [30,31]. Under hypoxic conditions, the hydroxylation of HIF-1α is hindered due to insufficient oxygen supply, thus leading to the stabilization and accumulation of the HIF-1α subunit within the cytoplasm of a cell. In addition to hypoxic conditions, HIF-1α can also be induced by various chemical compounds such as pravastatin, deferoxamine (DFO), and cobalt chloride in miscellaneous brain cells including endothelial cells, glioma cells, and neurons under normoxic conditions [32,33,34].
HIF-1α and HIF-1β each contain the structure of basic helix-loop-helix-PAS (bHLH-PAS) domains that are crucial for heterodimerization and DNA binding [28,35]. During hypoxia, translocation of the heterodimeric HIF-1α/β into the nucleus then drives the expression of target genes with the hypoxia-response elements (HREs) in their promoters [36]. Using genome-wide chromatin immunoprecipitation, hundreds of genes have been identified to be altered, either with increased or decreased expression levels, in an HIF-1-dependent manner in response to hypoxia [37]. Several well-known HIF-1 target genes including EPO, VEGF, and PDK1, which encode erythropoietin (EPO), vascular endothelial growth factor (VEGF), and pyruvate dehydrogenase kinase-1 (PDK1), are critical for the production of red blood cells, formation of blood vessels, and oxidative phosphorylation in the mitochondria, respectively. The heightened expression of HIF-1 target genes may assist these cells with inadequate oxygen supply in accommodating the critical hypoxic condition [23] (Figure 2).

2.1. Roles of HIF in Cancer

Mounting evidence supports the pivotal role of HIF-1 in tumor development, invasion, and metastasis [38]. The main components of the tumor microenvironment include blood vessels, lymphatic vessels, extracellular matrix, various immune cells, and fibroblasts [39]. A hypoxic tumor microenvironment is a driving force for cancer invasion [40,41,42]. The unmet demand for oxygen in human cancers can modify cellular metabolisms; these include the shift from oxidative phosphorylation to glycolysis, the enhanced synthesis of glycogen, and changes in the major substrate from glucose to glutamine for fatty acid synthesis. In parallel with these effects of metabolic reprogramming, HIF-1 activation may drive the expression of various genes to accommodate the hypoxic conditions and serve as a network hub to coordinate relevant signaling molecules for the promotion of tumorigenesis [24,43]. In recent years, combining HIF inhibitors with current cancer therapies to enhance anti-tumor activity is one strategy for combating therapeutic resistance [44]. This leads inevitably to an important question as to whether HIF inhibitors used in cancer patients may worsen their cognitive functions, especially in the elderly population, as age is a common risk factor for cancer and neurodegeneration. Similar concerns also exist for the clinical application of HIF inhibitors in the treatment of renal anemia [45], because the US FDA has recently approved Daprodustat, the first HIF prolyl hydroxylase (HIF-PH) inhibitor capable of blocking HIF-1 degradation, to treat anemia caused by chronic kidney disease (https://www.fda.gov/news-events/press-announcements/fda-approves-first-oral-treatment-anemia-caused-chronic-kidney-disease-adults-dialysis, accessed on 1 February 2023). Currently less well understood, these are pivotal issues that also deserve attention besides the roles of HIF-1 in AD.

2.2. Roles of HIF in Immunity

The immune system includes innate and adaptive components that can function efficiently through various immune-related cells such as dendritic cells, macrophages, neutrophils, B cells, and T cells. Infected and inflammatory tissues often appear with hypoxia with HIF-1 activation for adapting to hypoxic conditions; HIF-1 may also stabilize immunity under normoxic condition by regulating the metabolism and stimulating the expression of immune genes [46]. HIF-1 can affect innate immune cells in various aspects in macrophages, dendritic cells, and neutrophils. Depending on the extent of the stimuli, macrophages may polarize toward the proinflammatory status of the M1-type or toward the anti-inflammatory status of the M2-type. Notably, HIF-1α is induced in M1 macrophage polarization, whereas HIF-2α is induced during an M2 response. Macrophage polarization controls functionally divergent processes including the production of nitric oxide (NO), which is in part controlled by HIFs. Overall, NO availability is differentially regulated by HIF-1α versus HIF-2α to increase or suppress NO synthesis, respectively [47]. HIF-1α induced in lipopolysaccharide (LPS)-activated macrophages is involved in glycolysis and the induction of proinflammatory genes [48]. LPS stabilizes HIF-1α by the succinate-dependent inhibition of PHDs; further, pyruvate kinase M2 (PKM2) is also induced to promote the function of HIF-1α, which serves as a key metabolic reprogrammer for the expression of inflammatory genes [48]. Another study also reported that the glycolysis metabolism induced by HIF-1 is crucial for M1 macrophage polarization in mice [49]. Dendrite cells pivotal for antigen presenting play a significant role in innate immunity. HIF-1 is involved in the differentiation, migration, metabolism, and survival of dendrite cells under hypoxia and inflammation [50,51]. In immature dendritic cells, higher levels of HIF-1α are associated with the increased expression of pro-apoptotic molecule, such as the BCL2/adenovirus E1B 19-kDa protein-interacting protein (BNIP3) and BAX, along with the decreased expression of anti-apoptotic proteins like Bcl-2, heightened caspase-3 activity, enhanced cleavage of poly (ADP-ribose) polymerase (PARP), and cell death. On the contrary, the LPS-triggered maturation of dendritic cells attenuates hypoxia-induced cell death, wherein the PI3K/Akt pathway plays a vital part in this protective effect [51]. Pathogens may be captured and eradicated in neutrophil extracellular traps (NETs), which are mainly composed of DNA and relevant proteins generated by neutrophils. However, dysregulated NET formation may lead to excessive inflammation. A previous study showed the critical role of the mTOR/HIF-1α pathway for NET formation in neutrophils in response to LPS stimulation [52]. HIF-1 has also been shown to affect the differentiation and function of different subsets of T cells under both hypoxic and normoxic conditions. For example, HIF-1α is critical for the differentiation of various T cells including Th1, Th17, and CD8-positive effector cells; HIF-1 may also inhibit the development of regulatory T cells [53,54]. The T cell-specific knockout of HIF-1α in mice displays more severe colonic inflammation induced by dextran sodium sulfate; notably, hypoxia-induced HIF-1 is required for Treg activation but suppresses Th17 activation [55]. HIF-1α is also critical for B cell development and enhances the expression of interleukin (IL)-10 and CD11b, which may exert anti-inflammatory action [56,57,58]. B cells derived from HIF-1α-deficient mice manifested decreased IL10 and CD11b expression and became more susceptible to collagen-induced arthritis, experimental autoimmune encephalomyelitis, and inflammatory bowel diseases [58,59].

2.3. Roles of HIF in Oxidative Stress and Cerebral Hypoperfusion

Reactive oxygen species (ROS) are highly unstable, oxygen-containing molecules often with an unpaired electron. Fundamental for an aerobic life, the main sources of ROS in living cells include diverse origins such as the mitochondria, NADPH oxidase, cytochrome P450 enzymes, and 5-lipoxygenase [60,61,62]. To maintain the balance of redox status, cells possess the capability to counteract excessive ROS through endogenous enzymatic antioxidant systems (such as catalase, superoxide dismutase, glutathione peroxidase, and glutathione reductase) and non-enzymatic antioxidants (such as glutathione, thioredoxin, ubiquinone, and uric acid) [63,64]. However, excessive ROS may be generated under various stressful conditions such as hypoxia, inflammation, and serum deprivation [60,65]. These conditions may advance to overoxidation and result in lipid peroxidation, protein carbonylation, and DNA damage that can together proceed to cell death [66,67]. Oxidative stress is involved in numerous aging-related pathological conditions, including cancer, diabetes, chronic neurodegenerative disorders, and acute brain injuries like cerebral ischemia [68,69,70].
The role of HIF-1 in hypoxia-related oxidative stress is well-known in various ischemic conditions with inadequate oxygen supply [71,72,73,74,75]. ROS and several signal transduction pathways, including IKK/NF-kB, PI3K/AKT/mTOR, and RAS/RAF/MEK/ERK, can induce HIF-1 expression [46,76]. Downstream of HIF-1, a number of target genes involved in antioxidation with HREs in their promoters have been reported. Chromatin immunoprecipitation revealed that HIF-1α overexpression in hypoxic cardiomyocytes increases its binding to the HRE in the promoter of antioxidant gene heme oxygenase-1 (HO-1) that is known to attenuate the accumulation of ROS [77]. HO-1, but not HO-2, is also transcriptionally induced by HIF-1α in the renal medullary interstitial cells [78]. The suppression of glutathione peroxidase-8 (GPx8) enhances ER stress and cell death induced by oxidative stress in hepatocellular carcinoma cells [79]; notably, GPx8 is transcriptionally regulated by HIF-1α [80]. Peroxiredoxins (PRDXs) are a group of antioxidant enzymes that reduce hydrogen peroxide and alkyl hydroperoxides [81]; peroxiredoxin-2 (PRDX2) is a direct HIF-1 target gene whose expression is induced by prolonged hypoxia that may reciprocally interact with HIF-1α to suppress expression of other selected HIF-1 target genes under prolonged hypoxic conditions [82]. Hypoxia also affects the antioxidant activity of glutaredoxin-3 (Grx3) through HREs [83]. Depending on the experimental model systems, these previous findings together strongly support the potential antioxidative effects of HIF-1 via transcriptional induction of a multitude of antioxidation genes.
With aging, the competency for oxygen delivery in the body gradually declines and the ability to adapt hypoxia is presumed to be compromised [84,85,86]. Hypoxia may thus exacerbate the progression of neurodegenerative diseases including AD [87]. Emerging evidence has revealed that long-lasting or intermittent hypoxia, as seen in chronic obstructive pulmonary disease (COPD) or sleep apnea, is closely associated with AD [88,89]. In a prospective clinical study, older women with a sleep-related breathing disorder, which was defined as an apnea–hypopnea index of 15 or more events per hour of sleep, have an increased risk for declined cognition as compared with those without this disorder [90]. Apnea or hypopnea during sleep may therefore contribute to cognitive impairment. Nevertheless, discernible acute hypoxia is not often encountered clinically in AD patients and robust evidence for hypoxic microenvironments within the AD brains, as those seen in solid tumors, is still lacking [88,91]. With advanced neuroimaging tools, however, cerebral hypoperfusion, and accordingly hypoxia, is recognized as a constant feature along the AD continuum [92]. Evidence from neuroimaging studies showed that hypoperfusion in the brain areas, as a result of dysfunctional neurovascular units or the immunosuppressive network [93,94,95,96,97], is a potential inducer of AD pathology [98]. Cerebrovascular disease increased tau pathology in AD patients; further, transient cerebral artery occlusion as an animal model for brain hypoperfusion also promoted tau hyperphosphorylation [99]. Magnetic resonance imaging (MRI) studies showed cortical atrophy and hypoperfusion in a transgenic mouse model of AD [100]. In another human study, insufficient cerebral perfusion is correlated with cognitive deficits in AD. Regions with a lower perfusion index showed spatial similarities with atrophy in the posterior cingulate cortex, temporal lobes, and angular gyrus, while regions with lower relative cerebral blood flow were specified to the territories of distal branches in posterior cerebral artery territories [101]. It was also revealed with resting-state functional MRI that hypoperfusion may contribute to cognitive impairment via abnormal brain functional connectivity [102]. A prospective observational study in an aging population with an 11-year follow-up period has shown that anemia, wherein the compromised oxygen-carrying capability of blood may lead to brain hypoxia, was associated with an increased chance of developing dementia [103]; in another study, anemic subjects with a good baseline cognitive performance also had a two-fold higher risk of developing dementia three years later as compared to those without anemia [104]. Although the underlying mechanism linking anemia to incidental dementia is incompletely understood, one hypothesis suggested that chronic brain hypo-oxygenation is associated with an anemia-dependent risk of dementia [104]. Further studies linking cerebrovascular hypoperfusion or chronic hypoxia to dementia development are needed to further clarify these issues.
It was realized that hypoxia, oxidative stress, and neuroinflammation are crucial contributors in neurodegenerative diseases including AD [88,89]. Intriguingly, these factors may also trigger the activation of the HIF-related pathway. As compared to cancers, however, the role of HIF-1 is not well-defined in AD in terms of chronic hypoxia, oxidative stress, and inflammation, all of which are worthy of further exploration. Highly conserved during evolution, HIF-1α is stabilized under hypoxic stress so the organisms may cope with such stressful conditions. Therefore, manipulating the components of the HIF pathway may potentially exert neuroprotective effects in a variety of human neurological disorders including AD [105,106,107].

3. Expression Level of HIF-1α and Its Impacts on AD

It is well understood that the HIF system activates a wide variety of physiological responses to hypoxia, ranging from the enhancement of survival to cell cycle arrest or even cell demise [6]. Whether the Janus-faced HIF-1α acts as a “damaging factor” or a “pro-survival factor” in AD may depend on the cell types and local cellular conditions [108,109]. The altered expression of HIF-1α has been noted in AD patients as well as in transgenic AD mouse models. The downregulation of HIF-1α, which regulates the two major brain glucose transporters GLUT1 and GLUT3 for glucose uptake into neurons, was observed in AD brains; this decrease in HIF-1/GLUT1/3 correlated to the hyperphosphorylated tau and higher density of NFTs [110]. In contrast, HIF-1α is overexpressed in brain microvessels derived from AD patients [111]. Consistently in the brains of adult (50–60 weeks of age) Tg2576 AD mice, immunofluorescence staining of brain microvessels revealed a significantly higher level of HIF-1α along with the heightened expression of pro-angiogenic factors and reduced anti-apoptotic Bcl-xL [112]. Triple (3×-Tg AD; PS1M146V, APPswe, tauP301L) transgenic AD mouse brains also showed hypoxic vessels expressing HIF-1α, which resulted in the formation of NLR family pyrin domain-containing 1 (NLRP1) inflammasome to further stimulate HIF-1α expression and ultimately formed an HIF-1α-NLRP1 vicious circuit [113]. In the microglia of human AD hippocampus, the upregulation of HIF-1α and its target genes correlates with reduced coverage of Aβ plaques by microglia and an increased extent of plaque-associated neuropathology [114]. In APP knock-in mice, complement C3a receptor (C3aR)-positive microglia showed an upregulated expression of HIF-1 signaling with abnormal lipid droplet accumulation; the knockout of C3aR accompanied by reduced HIF-1α signaling reversed AD pathology with improved learning memory [115]. In astrocytes derived from 5×FAD mouse brains, compensatory mechanisms including increased HIF-1 expression was observed, which is expected to protect cells against Aβ toxicity [116]. Depending on the types of cells that express HIF-1, coupled with its multifaceted effects on handling various cellular stress, it is conceivable that HIF-1 may play very complicated roles in AD [117,118]. The potential beneficial and detrimental roles of HIF-1 pathways in AD are discussed in detail below (Figure 3).

3.1. The Beneficial Roles of HIF-1 in AD

3.1.1. Potential Protective Mechanisms of HIF-1 in AD

Energy metabolism—Previous studies revealed a global reduction in glucose metabolism that is associated with AD patients [119]. Several genes involved in glucose transport and glycolysis, such as GLUT1/GLUT3 and phosphoglycerate kinase-1 (PGK1), are transcriptionally regulated by HIF-1 [120,121]. The expression levels of glucose transporters were downregulated in AD brains, which were correlated to abnormal tau phosphorylation and the expression of HIF-1 [110]. HIF-1 critically involved in glucose metabolism mediates neuroprotective effects against Aβ toxicity; further, both HIF-1 and those enzymes in the glycolytic pathways are crucial for neuronal survival over the frontal cortex in AD patients [122]. These studies may denote a possible mechanism underlying AD-related neurodegeneration that involves impaired brain glucose uptake/metabolism due to decreased HIF-1, resulting in deficient expression of GLUT1 and GLUT3. Aerobic glycolysis, the nonoxidative metabolism of glucose despite the presence of abundant oxygen, is a key regulator of synaptic plasticity in neurons, which requires the synthesis of macromolecules like mRNA and proteins [123]. Reduced aerobic glycolysis during aging may ruin cell survival mechanisms and fail to counteract neurodegenerative changes; notably, aerobic glycolysis is regulated at the transcriptional level by HIF-1α and peptidyl-prolyl cis/trans isomerase-1 (Pin1) [124]. Extended from these findings is that, under a critical clinical condition, those drugs capable of decreasing glucose uptake in the brains should be avoided for AD patients [125]. Besides neurodegeneration, glial activation is one of the early alterations during AD progression, possibly related to the deposition of Aβ resulting in gliosis [126,127]. In the early stage of the Alzheimer’s continuum, reactive astrogliosis is linked to higher consumption of cerebral glucose [128]. Preventing the proteolysis of HIF-1α may reverse Aβ-induced glial activation and glycolytic changes [129]. These findings again imply the crucial role of HIF-1α, which may be mediated by its transcriptional activity to maintain metabolic integrity and restore compromised energy metabolism in affected AD brains, in both neurons and glia.
Neuroprotection and neurorestoration—Hypoxia or brain hypoperfusion may contribute to AD pathogenesis [130,131]. HIF-1α, induced under such hypoxic circumstances, may play a compensatory role in coping with the diseased conditions [87]. Emerging evidence has revealed HIF-1α as a potential therapeutic target for various neurological disorders [87,132,133,134]. First, Aβ has been shown to directly induce HIF-1α expression in vitro; interestingly, low levels of Aβ protect neurons from a more severe insult by triggering HIF-1α, whereas the overexpression of HIF-1α alone is sufficient to protect neurons against Aβ toxicity [122]. Second, several downstream genes of HIF-1 are well-known to be vasoactive molecules, such as EPO, endothelial nitric oxide synthase (eNOS), and VEGF [105,117]. Notably, these proteins may also improve neuronal cell growth or survival besides their pro-angiogenic functions. For example, EPO and VEGF carry essential trophic effects during brain development and, in response to neuronal damage, exert neuroprotective or neurorestorative effects [135]. In hippocampal neurons in vitro, EPO was both necessary and sufficient to prevent Aβ-induced apoptosis in both the early and later stages of neurodegeneration, which involves expression and translocation of the p65 subunit of nuclear factor-kappaB (NF-kB) [136]. We have shown previously that EPO may exert neuroprotective effects against metabolic insults induced by mitochondrial inhibitor 3-nitropropionic acid (3-NP) in primary cortical neurons [137].
Neurogenesis—Sleep apnea is known to affect mental performance and may also contribute to neurodegeneration in AD [138,139,140]. Rodents under intermittent hypoxia, an animal model of sleep apnea, presented impaired spatial memory, hippocampal function, and adult neurogenesis; HIF-1α signaling activated by intermittent hypoxia in early neuroprogenitors increases the production of mature neurons upon the termination of intermittent hypoxia [141]. Chronic hypoxia also induces neurogenesis in the subgranular zone (SGZ) in the hippocampus of adult double transgenic APPswe/PS1ΔE9 mice via activation of the Wnt/β-catenin signaling pathway [142], which is HIF-1α-dependent [143]. These findings suggest that adult neurogenesis regulated by HIF-1 may represent one of its beneficial mechanisms in AD.
Counteracting oxidative stress—HIF-1 can generate the reductive equivalents of NADH/NADPH to counter oxidative stress [122]. Under hypoxic conditions, increased cytosolic anaerobic glycolysis over mitochondrial oxidative phosphorylation as a result of HIF-1 induction may also generate more pyruvate, which is an antioxidant capable of scavenging free radicals like H2O2 [144]. In another study of redox proteomics, several hippocampal proteins critical for energy metabolism, neuroplasticity, and mitogenesis/proliferation were found to be oxidatively modified in patients with mild cognitive impairment (MCI), including alpha-enolase, glutamine synthetase, pyruvate kinase M2, and Pin1; notably, the interacteome of these proteins revealed that they functionally interact with several factors including HIF-1 [145]. This study highlights the potentially antioxidative roles of HIF-1, which are expected to be beneficial in AD.
Enhancing brain circulation or angiogenesis?—Earlier it was shown that individuals suffering from severe hypoxia or ischemia are more susceptible to developing AD [146]. HIF-1α is found to be elevated in the microcirculation of AD patients [111]. In animal studies, brain sections from AD transgenic mice also showed heightened expression of HIF-1α as well as the pro-angiogenic proteins including angiopoietin-2 (Ang-2) and matrix metalloproteinase-2 (MMP2) in the brain vasculature [112]. Conceivably, it seems reasonable to predict that HIF-1 activation may exert beneficial effects against AD by enhancing angiogenesis or augmenting cerebral blood flow. Despite these lines of correlative evidence in clinics and animal models in vivo, however, direct causative evidence supporting this beneficial mechanism of HIF-1α in AD is still lacking in the literature. On the contrary, the inhibition of HIF-1α expression was shown to mediate the beneficial effects of schisandrin B, an active component derived from the Chinese herb Wuweizi, in the rat bilateral common carotid artery occlusion (BCCAO) as a vascular dementia model [147]. Therefore, whether enhancing HIF-1 activity may have positive impacts on AD by augmenting brain circulation or enhancing angiogenesis requires further investigation.

3.1.2. Multiple Chemical Reagents and Proteins Exert Beneficial Effects on AD via Induction of HIF-1

Earlier it was demonstrated that iron is required for Aβ toxicity and iron chelators have neuroprotective effects [148]. This may in part due to the fact that aberrant iron metabolism, especially the ferrous iron (Fe2+) in the cytoplasmic labile iron pool, produces superoxide anions (O2·) and a strong ROS hydroxyl radical (OH·) through a Fenton reaction with H2O2.; the resultant ROS cause oxidative damage to lipids, proteins, and DNA, thus leading to cell death and affecting Aβ misfolding along with plaque aggregation [149]. Furthermore, in the presence of Fe2+ and/or lipoxygenases, the polyunsaturated fatty acids in the cell membrane catalyze lipid peroxidation to trigger ferroptosis, an iron-specific programmed cell death, which is closely related to the occurrence, development, and prognosis of AD [150]. Transferrin receptor 1 (TfR1) is a key player in the regulation of the brain distribution of iron. One recent study suggested that, in AD under hypoxia and other non-hypoxic stimuli, such as oxidative stress and inflammation, TfR1 upregulation may be associated with HIF-1 activation causing iron dyshomeostasis in the brain, which ultimately contributes to AD pathology [151]. In this regard, iron chelation may be considered a therapeutic option for AD [152]. Consistently, an iron chelator M30 capable of upregulating HIF-1α with heightened expression of its downstream target genes, including enolase-1, EPO, p21, tyrosine hydroxylase, VEGF, and insulin signaling pathway, in cortical neurons and APP/PS1 AD mice, may have anti-Alzheimer characters [153,154] (Table 1). In APP/PS1 transgenic mice, intranasal delivery of the iron chelator DFO can upregulate the p38/HIF-1α pathway and lessen synaptic loss in the brain [155]. Intranasal DFO treatment also improves memory in healthy mice that is accompanied by reduced GSK-3β activity and intensified HIF-1α activity [156]. In primary cultures of rat and mouse astrocytes, DFO stabilizes HIF-1α to inhibit both PHDs and the proteasome, thereby mitigating Aβ-induced glial activation with enhanced pentose shunt to generate more NADPH for restricting ROS accumulation [129]. We have also shown before that HIF-1 induction with cobalt chloride or DFO can protect rat C6 glioma cells against metabolic insults induced by the irreversible mitochondrial inhibitor 3-NP [157]. These results together denote potential beneficial effects of iron chelators, such as M30 and DFO, through HIF-1α stabilization in AD.
In addition to iron chelators, several chemical compounds or proteins exert beneficial effects against AD via the induction or stabilization of HIF-1α. Recently, it was shown that gut dysbiosis, or disturbance in the gut microbiota, can lead to inflammation and is associated with the pathogenesis of various diseases including AD [158,159]. The term “brain–gut axis” denotes crosstalk between the brain and the gut that involves multiple overlapping pathways, which comprise the autonomic functions, immune systems, neuroendocrine, neuro-modulatory molecules, and bacterial metabolites [160]. It is imperative to understand the mechanisms inherent in the microbiota–gut–brain axis so that microbe-based intervention and therapeutic strategies may be developed for neurodegenerative diseases [161]. One previous study employing 3×Tg-AD mice revealed that chronic supplementation with SLAB51, a multi-strain probiotic formulation, can augment the cerebral expression of HIF-1α, possibly by reducing the level of PHD2 critical for HIF-1α degradation; SLAB51 also decreases the expression of inducible nitric oxide synthase (iNOS) in the brain, with a reduction in NO levels in the plasma of AD mice [162]. This study adds an additional mechanism for probiotics to reduce oxidative stress and inflammation in AD models through the regulation of HIF-1α expression. The lactoferrin present in milk is an iron-binding glycoprotein with pleiotropic functions. Recently, lactoferrin has been suggested as a neuroprotective agent because the intranasal delivery of lactoferrin into AD mice can augment α-secretase-dependent APP processing through the ERK1/2-CREB and HIF-1α pathways [163]. The beneficial effects of lactoferrin in reducing Aβ aggregation with improved spatial learning ability were also revealed. As a highly accessible nutrient supplement, further clinical studies on the advantageous effects of lactoferrin for AD are warranted. Neuroglobin is a hypoxia-inducible protein with protective effects in animal models of AD, stroke, and related nervous system disorders [164]. It was demonstrated that the knockdown and overexpression of HIF-1α, respectively, reduced and increased neuroglobin levels, consistent with a causal relationship between HIF-1 and neuroglobin induction [165]. This finding adds one more mechanism for the beneficial effect of HIF-1α expression and neuroglobin with potential therapeutic values for AD. α-Lipoic acid can maintain brain glucose metabolism through the BDNF/TrkB/HIF-1α signaling pathway in P301S mice, a transgenic mouse model for tauopathy; chronic α-lipoic acid administration into the P301S mouse brains elevated the expression of GLUT3, GLUT4, VEGF, and HO-1 at mRNA or protein levels that together increased glucose availability [166]. Finally, recent epidemiological studies and experimental evidence suggested that coffee consumption lowered the risk of cognitive disorders including AD [167,168]. Coffee was found to induce VEGF expression in human neuroblastoma SH-SY5Y cells through the activation of HIF-1α, which was related to the inhibition of prolyl hydroxylation independent of caffeine or caffeic acid [169]. A previous study revealed that a single nucleotide polymorphism (SNP) rs1868402 in the PPP3R1 gene encoding protein phosphatase 3 regulatory subunit B (PPP3R1) is significantly associated with the rapid progression of AD [170]. A low level of PPP3R1 proteins showed a strong correlation with AD and may serve as a potential biomarker for predicting and preventing AD for future development of personalized medicine; notably, PPP3R1 was found to be involved in the HIF-1 pathway [171]. Not surprisingly, the direct enhancement of HIF-1 activity or its specific downstream target genes is expected to also provide protective effects against AD. For example, viral vectors expressing HIF-1α may inhibit hippocampal neuronal apoptosis induced by Aβs both in vivo and in vitro [172]. Apolipoprotein E4 (ApoE4), the most common genetic risk factor for sporadic AD, is associated with more evident neurodegeneration and vascular impairments [173,174]. ApoE4-driven brain pathology revealed a specific decrease in both VEGF receptor-2 and HIF-1α. Using VEGF-expressing adeno-associated virus (AAV-VEGF) driven by the GFAP promoter that allows VEGF expression exclusively in astrocytes, it was demonstrated that the AAV-VEGF may reverse the ApoE4-related Aβ42 aggregation and accumulation of hyperphosphorylated tau proteins [174]. This evidence indicates the critical role of HIF-1 in AD patients with ApoE4. Further study is warranted to manipulate the HIF-1 expression in these subgroups of AD to examine its potential clinical outcomes. These findings together support the potential use of gene therapy for the treatment of neurodegenerative diseases including AD.
Table 1. List of examples with beneficial roles of HIF-1 in AD-related studies.
Table 1. List of examples with beneficial roles of HIF-1 in AD-related studies.
Chemical Compound, Drug, Nutrient, ProteinMechanismsStudy ModelReference
M30, an iron chelatorupregulated HIF-1α and its target genes: enolase-1, erythropoietin, p21, tyrosine hydroxylase, VEGF, Glut-1in vivo: APP/PS1 double Tg mice in vitro: rat primary cortical neurons[153,154]
DFO, an iron chelator* upregulated the p38/HIF-1α pathway and lessened synaptic loss in the brain
** reduced GSK-3β activity and intensified HIF-1α activity
*** increased HIF-1α, inhibited PHD2 and the proteasome, reduced glial activation, produced more NADPH to limit ROS accumulation
in vivo: APP/PS1 Tg mice
in vivo: healthy mice
in vitro: mouse and rat astrocytes
[155]
[156]
[129]
SLAB51, a multi-strain probiotic
formulation
increased HIF-1α, decreased PHD2, iNOS in brain, and NO levels in plasmain vivo: 3 × Tg-AD and wild-type mice[162]
Lactoferrin, a nutrient derived from milk augmented α-secretase-dependent APP processing through the ERK1/2-CREB and HIF-1α pathways, reduced Aβ aggregationin vivo: APP/PS1 double Tg mice[163]
Neuroglobin, a hypoxia-inducible protein with cytoprotective effectsshRNA-mediated knockdown and lentiviral vector-mediated overexpression of HIF-1α in vitro: HN33 neural cell line[165]
α-Lipoic acidincreased GLUT3, GLUT4, VEGF, and HO-1 expression with enhanced glucose availability through BDNF/TrkB/HIF-1α signalingin vivo: tauopathy model, P301S mice[166]
Coffee inhibited prolyl hydroxylation to activate HIF-1α and induced VEGF expression in vitro: SH-SY5Y human neuroblastoma cell line [169]
Viral vector expressing HIF-1αinhibited hippocampal neuronal apoptosis induced by Aβ proteinin vitro: primary culture of hippocampal neurons
in vivo: Sprague-Dawley rats
[172]
APP/PS1: amyloid precursor protein/presenilin-1, DFO: desferoxamine, GLUT-1: glucose transporter-1, HO-1: heme oxygenase-1, iNOS: inducible nitric oxide synthase, NADPH: nicotinamide adenine dinucleotide phosphate, PHD2: prolyl hydroxylase 2. * denotes Ref [155]; ** denotes Ref [156]; *** denotes Ref [129].

3.2. The Detrimental Roles of HIF-1 in AD

3.2.1. Potential Detrimental Mechanisms of HIF-1 in AD

Although some experimental evidence supports the beneficial effects of the HIF-1 pathway in AD, unfavorable impacts of HIF-1 activation in AD have also been reported. For example, HIF-1α can affect APP processing by regulating the activities of α-, β-, and γ-secretase, thereby increasing the generation of Aβ or decreasing the secretion of sAPPα. Hypoxia, and possibly HIF-1α, may also modulate the expression or activities of Aβ-degrading enzymes like neprilysin (NEP), endothelin-converting enzyme (ECE)-1, and insulin-degrading enzyme (IDE). HIF-1α may also modulate microglial activation to aggravate the severity of neuroinflammation, which is expected to expedite AD pathogenesis [175]. Other possible detrimental effects of HIF-1 in AD include impairing the integrity of the blood–brain barrier (BBB) and compromising the microvasculature. For example, in rat brain capillary endothelial cells (RBE4), oxygen–glucose deprivation (OGD), as an in vitro model of hypoxia/ischemia, elicits Aβ42 production through HIF-1-mediated BACE1 upregulation [176]. Thus, ischemic events may directly contribute to the enhancement of the amyloidogenic metabolism in brain capillary endothelial cells, leading to intracellular deposition of Aβ42, impaired Aβ clearance, and AD-related BBB dysfunctions.
APP processing by BACE1/β-secretase—Previous studies suggested that hypoxia may contribute to AD pathogenesis by overexpressing APP and increasing Aβ formation [177,178]. Hypoperfusion caused by focal ischemia induces APP expression at mRNA levels [179]. Prolonged hypoxia increased production of Aβ that selectively increased the expression of L-type Ca2+ channels, which is considered detrimental in AD; hypoxia also promoted physical association of Aβ with the α1C subunit of the L-type Ca2+ channel that likely contribute to the Ca2+ dyshomeostasis of AD [180,181]. These earlier findings collectively revealed the link between hypoxia/ischemia and APP expression/processing that may contribute to AD pathogenesis. The direct involvements of HIF-1 in APP processing were later confirmed. For example, hypoxia increases BACE1 gene transcription at mRNA levels through the induction of HIF-1 with resultant increased β-secretase activity and Aβ production; notably, gel shift assays reveal HIF-1 binding to the HREs in the BACE1 promoter; indeed, the overexpression of HIF-1α is sufficient to increase BACE1 at both the mRNA and protein levels, whereas the downregulation of HIF-1α reduces the level of BACE1 [182]. A further study revealed that hypoxia triggers BACE1 expression through a biphasic mechanism; the early stage is mediated by the mitochondrial electron transport chain and the later stage is caused by the activation of HIF-1 and oxidative stress [183].
APP processing by α-secretase?—Besides enhancing BACE1 expression along with increased β-secretase activity, chronic hypoxia decreases the protein expression of a disintegrin and metalloproteinase 10 (ADAM10), which is considered an α-secretase mediating the non-amyloidogenic processing of APP, in the neuronal SH-SY5Y cells [184,185], along with the suppressed secretion of sAPPα [184]. However, the direct involvements of HIF-1α were not reported in these two studies.
PS1/2 and γ-secretase?—The γ-secretase is responsible for further digesting the α-CTF (C83) and β-CTF (C99) following the nonamyloidogenic and amyloidogenic cleavage of APP by α- and β-secretase, respectively. Being a high-molecular-weight complex consisting of four components, namely presenilin-1/2 (PS1/2), nicastrin (NCT), anterior pharynx-defective-1 (APH-1), and presenilin enhancer-2 (PEN-2), PS1/2 is the catalytic subunit in the γ-secretase complex [186]. Brain hypoxia induced by cerebral hypoperfusion or breathing 10% O2 triggered HIF-1α binding to γ-secretase to enhance its biological activity without affecting the expression level of individual subunits; further, the expression of full-length HIF-1α in primary neurons increased BACE1 expression and γ-secretase activity in a transcriptional and non-transcriptional manner, respectively, for Aβ production under hypoxic conditions [187]. In another earlier study, the activation of HIF-1 by short-term NiCl2 treatments, a condition known as chemical hypoxia, substantially increases APH-1A expression at both mRNA and protein levels without affecting APP or the other three components of the γ-secretase complex; importantly, NiCl2 treatments also lead to an increased secretion of Aβ and decreased formation of APP CTFs, indicative of elevated γ-secretase activity [188]. Importantly, loss-of-function mutations in PS1/2 or mutations in PS1 associated with familial AD (FAD) may also affect HIF-1 induction. PS1/2 γ-secretase-mediated cleavage of the APP generates AICD, which functions as a transcriptional activator to induce Hif1a gene expression; on the other hand, PS1/2 itself, in a γ-secretase-independent manner, increases PHD2 activity to promote HIF-1α degradation in vitro [189]. Using fibroblasts lacking PS1, the induction of HIF-1α was impaired in response to cobalt chloride, which is known to stabilize HIF-1α under normoxia, or by insulin [190]. The lentivirus-mediated expression of human PS1 in part rescued the responsiveness of PS1-/- fibroblasts to cobalt chloride induction; however, HIF-1α induction did not require γ-secretase activity. Interestingly, PS1 and HIF-1α appeared to physically interact with each other, suggesting that PS1 may protect HIF-1α from degradation [190]. In fibroblasts harboring the M146V PS1 FAD mutation on a mouse PS1-null background, the metabolic induction of HIF-1α by insulin was impaired, but not by cobalt chloride [190]. These findings indicate complex mechanisms whereby PS1/γ-secretase, both wild-type and those containing FAD mutations, modulate the induction of HIF-1α.
Aβ degradation?—In addition to regulating APP expression and processing by secretase activities, HIF-1 is also involved in Aβ degradation. Neprilysin (neutral endopeptidase, NEP) is a zinc-dependent metalloprotease responsible for Aβ degradation [191]. In prostate cancer cell lines and human umbilical vascular endothelial (HUVEC) cells, hypoxia inhibits the expression of NEP at mRNA levels; out of the three putative HREs upstream of the NEP promoter, two demonstrate a specific reduction, rather than an increase, in cobalt-induced HIF-1 binding in gel shift assays [192], suggesting a negative impact on NEP expression on the binding of HIF-1 to these HREs. In contrast to these findings, one study conducted in neuroblastoma cells reported that HIF-1α mediates the upregulation of NEP through HIF-1α binding to histone deacetylase (HDAC)-1, thereby reducing the association of HDAC-1 with the NEP promoter, with a resultant activation of its transcription in a de-repression manner [193]. The causes underlying these contradictory results regarding the HIF-1-mediated regulation of NEP require further investigation, but several contributing factors may be considered, including the different cell types (HUVEC versus N2a neuroblastoma cells) as well as how HIF-1α was induced (cobalt chloride versus hypoxia under low-oxygen tension). Another Aβ-degrading enzyme, ECE-1, has also been proposed to be an HIF-1 target gene in endothelial cells [194]. Hypoxia downregulated the expression of IDE, another peptidase known to degrade Aβ, in U87 glioma cells [195], but the potential roles of HIF-1α remain unclear. Taken together, understanding whether HIF-1 directly regulates the expression or activities of these Aβ-degrading enzymes and their potential impacts on AD pathogenesis requires more investigation.
Tau hyperphosphorylation?—Depending on the experimental model systems, how HIF-1α may affect tau hyperphosphorylation remains controversial. In a clinical study, two major brain glucose transporters, GLUT1 and GLUT3, responsible for glucose uptake into neurons, were decreased in AD brains; notably, this decrease correlated to the hyperphosphorylation of tau and downregulation of HIF-1α, but not HIF-1β [110]. Since HIF-1 is a major regulator of GLUT1 and GLUT3, these studies provide a piece of correlative evidence linking tau hyperphosphorylation to HIF-1α downregulation in AD. Accordingly, HIF-1α has also been shown to suppress tau phosphorylation. For example, one recent study reported that T-2 toxin, a mycotoxin that may potentially lead to the progression of AD, stimulates HIF-1α expression, along with induction of APP and hyperphosphorylation of tau proteins in microglial BV2 cells; intriguingly, further studies revealed that the T2-induced HIF-1α actually functions as a “brake” to negatively regulate APP induction and tau phosphorylation, suggesting a protective effect of HIF-1α in microglial cells challenged with T-2 toxin, in part by suppressing tau hyperphosphorylation [196]. In contrast, however, HIF-1α has also been shown to indirectly contribute to tau phosphorylation. As a risk factor for AD, chronic hypoxia upregulates HIF-1α, which decreases the activity of protein phosphatase-2A (PP2A), thereby mediating tau hyperphosphorylation with resultant cognitive dysfunction [197]. Cobalt, as an environmental toxicant that is also known to stabilize HIF-1α, severely induces Aβ deposition, tau hyperphosphorylation, and dysregulated autophagy in the hippocampus and cortex of mice; importantly, HIF-1α knockdown by siRNA effectively attenuated the increased tau phosphorylation at Thr181 induced by cobalt [198], suggesting that HIF-1α is responsible for cobalt-dependent tau phosphorylation. Finally, HIF-1α may also mediate the beneficial effects of small-molecule compounds without notably affecting tau phosphorylation. The iron chelator DFO, known to stabilize HIF-1α, improves memory in healthy C57 mice [156] and also exerts protective effects in P301L mice [199]. These transgenic mice overexpress the human 4R/2N tau isoform bearing the P301L mutation under the control of the neuron-specific murine Thy1 promoter with an accumulation of hyperphosphorylated tau, which serves as a transgenic model for AD and frontotemporal dementia (FTD). It was found that P301L mice have significantly less HIF-1α with more severe total protein oxidation than wild-type controls, whereas the intranasal delivery of DFO significantly reversed these differences; however, no significant decrease in phosphorylated tau was observed in the brains of these DFO-treated mice, at least for the brain regions examined [199]. Depending on the experimental model systems, therefore, HIF-1α may enhance, suppress, or have no direct effects on the phosphorylation status of tau proteins. Obviously more investigation is required to establish the definite roles of HIF-1 in tau hyperphosphorylation.
Microglia and neuroinflammation?—HIF-1 may affect microglia in different aspects. Using methoxy-X04 (X04), a brain-penetrant fluorescent Aβ probe, to isolate plaque-containing (X04+) and non-containing (X04-) microglia from 5×FAD mouse brains for transcriptome analysis, it was found that HIF-1α and its downstream target genes are involved in the enhanced phagocytosis of synaptic components around plaques and, through a feed-forward loop, ultimately augment Aβ phagocytosis in microglia [200]. The rapid pruning of damaged synapses near dystrophic neurites around plaques and the enhancement of Aβ phagocytosis may be considered protective and, in this regard, HIF-1 likely plays a beneficial role in microglia-dependent phagocytosis in AD. In contrast, however, excessive HIF-1 activity may have detrimental effects on microglia in AD. For example, the prolonged activation of HIF-1α results in cell cycle arrest along with the impaired proliferation of microglia; under in vivo condition, the overstabilization of HIF-1α reduces the proliferation and clustering of plaque-associated microglia along with increased Aβ neuropathology [114]. It has been demonstrated that hypoxia may also induce autophagic cell death and inflammatory responses through HIF-1α in microglia [201], although its potential impact on AD requires more investigation. In addition to affecting phagocytosis, cell proliferation, clustering around Aβ plaques, and possibly autophagic cell death, the HIF-1α pathway plays a pivotal role in the metabolic program and immune responses in microglia. The exposure of microglia to Aβ triggers acute inflammation along with metabolic reprogramming from oxidative phosphorylation to glycolysis, which depends on the mTOR-HIF-1α pathway; however, chronic exposure to Aβ leads to an overall defective energy metabolism with diminished immune responses, including cytokine secretion and phagocytosis [202]. Overall, conditions with prolonged or overwhelming stress like severe or chronic hypoxia may alter the beneficial roles of HIF-1 such that it is converted into a potential activator of cell death in AD [133].
Brain hypoperfusion and impaired vascular functions—As mentioned above, microvessels isolated from the brains of AD patients [111] as well as AD transgenic mice [112] express a number of angiogenic proteins along with elevated HIF-1α expression. However, direct evidence supporting the beneficial mechanism of enhancing cerebral blood flow or microcirculation by HIF-1α in AD models is still lacking. On the contrary, it has been reported that chronic cerebral hypoperfusion commonly observed in AD patients may induce HIF-1α to cause BBB damage and ultimately impair Aβ clearance [203]. Another recent study revealed that chronic hypoxia capable of activating HIF-1 is detected in AD brains and retinas, especially in microvascular endothelial cells, which leads consequently to the formation of NLRP1 inflammasome and upregulates the signaling cascades of “adaptor molecule apoptosis-associated speck-like protein containing a CARD (ASC)-caspase-1- IL-1β”. In turn, NLRP1 can reciprocally stimulate HIF-1α expression to reinforce the HIF-1α-NLRP1 circuit, which may further destroy the vascular system and thus suggest a detrimental role of HIF-1α in microvascular structures in AD [113]. These negative impacts of HIF-1α in the microvasculature of AD, along with the aforementioned HIF-1 effects on APP processing and Aβ production/degradation, may further impair vascular function in AD and compromise BBB function, leading to impaired Aβ clearance [204].
Neuronal cell cycle reentry—Emerging evidence indicates that aberrant cell cycle reentry and subsequent apoptosis in fully differentiated neurons occur during the advancement of AD or other neurodegenerative diseases [205,206,207]. We have shown before that the signal transduction pathways involving the inhibitor of DNA-binding/differentiation protein-1 (Id1), HIF-1α, cyclin-dependent kinases-5 (CDK5), sonic hedgehog, and protein kinase C-delta (PKCδ) may contribute to Aβ-induced cell cycle reentry and neurotoxicity in fully differentiated postmitotic neurons [15,16,206,208,209,210], adding another piece of evidence supporting the detrimental role of HIF-1α in AD under selected circumstances.

3.2.2. Proteins and Chemical Reagents Exert Beneficial Effects in AD via Inhibition of HIF-1

Recently, the relationship between dementia and diabetes mellitus has inspired researchers to explore new therapeutic potential from glucose regulators for devastating neurodegenerative diseases like AD or PD [211,212,213]. Fibroblast growth factor 21 (FGF21), a member of the FGF family, is a liver-secreted peptide hormone encoded by the fgf21 gene [214]. Positively implicated in the regulation of energy, glucose, and lipid metabolism, FGF21 is an appealing and promising therapeutic target for DM [215]. However, FGF21 may also present miscellaneous influences on the central nervous system [216,217]. For example, FGF21 lessened tau hyperphosphorylation and oxidative stress in cellular and rat models of AD; its beneficial effects involved the inhibition of the PP2A/MAPKs/HIF-1α pathway [216] (Table 2). Heparin-binding EGF-like growth factor (HB-EGF), an EGF family member widely distributed in neurons and glia, can be induced by hypoxia and/or ischemia, which contributes to chronic cerebral hypoperfusion (CCH)-mediated Aβ accumulation [218]. An HB-EGF-dependent increase in HIF-1α expression can activate matrix metalloprotease-9 (MMP9) to cause BBB disintegration [203]. Neuregulin 1 (NRG1), which belongs to the epidermal growth factor family, is thought to play a role in synaptic plasticity [219,220]. Cobalt chloride, which is known to stabilize HIF-1α, induced cytotoxicity along with the increased expression of HIF-1α and p53 in SH-SY5Y cells; notably, pretreatment with NRG1 inhibited the accumulation of HIF-1α, decreased p53 stability, and significantly mitigated cell death in the SH-SY5Y cells exposed to cobalt chloride [221]. These studies revealed the detrimental effects of HIF-1 by various protein mediators in Aβ-induced neurotoxicity or AD animal models, both in vivo and in vitro.
In addition to protein mediators, various cellular environments or growth conditions may affect HIF-1α induction and AD pathology. Under high-glucose condition, BACE1 is upregulated with increased Aβ production through HIF-1α activation and decreased LXRα expression via the JNK pathway in a ROS-dependent manner in SK-N-MC, a neuroblastoma cell line [222]. Thiamine (Vitamin B1) insufficiencies (TI) have been associated with AD-like neuropathology. Using the HT22 neuronal hippocampal cell line, it was demonstrated that TI-induced HIF-1α triggers amyloidogenesis through the transcriptional expression of BACE1 along with increased β-secretase activity (BACE1); furthermore, TI also induces the expression of pro-apoptotic protein BNIP3 via HIF-1α and, correspondingly, neurotoxicity caused by TI conditions can be significantly reduced with the knockdown of HIF-1α and BNIP3 [223]. This study suggests that, under selected circumstances, the induction of HIF-1α may also trigger the expression of pro-apoptotic genes that contribute to neuronal death.
Several compounds may also exert beneficial effects through the inhibition of the HIF-1 pathway. Ginsenoside Rg1, one of the active ingredients in Panax ginseng, can ameliorate declined cognitive function and reduce cerebral Aβ levels [224,225]. It was demonstrated that Rg1 reduced Aβ-induced mitochondria-dependent apoptosis in human endothelial cells, which involved lessened HIF-1α expression accompanied by diminished ROS production and protein nitrotyrosination [226]. Salidroside, a glucoside of tyrosol found in the plant Rhodiola rosea, has been reported to mitigate hypoxia-induced abnormal APP processing in SH-SY5Y neuronal cells [227]. Salidroside pretreatment significantly decreased the expression of BACE1 at both mRNA and protein levels along with inhibited β-secretase activity, thus attenuating Aβ generation induced by hypoxia without affecting γ-secretase activity. Salidroside also promoted the secretion of sAPPα in hypoxic condition without affecting APP, ADAM10, and ADAM17; the latter two are considered α-secretases, suggesting that heightened sAPPα secretion may be the consequences of decreased β-secretase activity. Notably, under hypoxic conditions, salidroside pretreatment reduced the protein level of HIF-1α, suggesting that salidroside may attenuate a hypoxia-induced increase in BACE1, possibly through downregulating the HIF-1α protein level [227]. A high-fat diet stimulates amyloidogenic pathways, which is critical for the pathogenesis of AD [228]. Palmitic acid-BSA (PA-BSA) treatment stimulates BACE1 expression in astrocytes with elevated oxidative stress [229,230]. It was found that PA-BSA-induced Aβ production involved the activation of Akt/mTOR/HIF-1α and Akt/NF-κB pathways to stimulate the expression of APP and BACE1; silencing both genes correspondingly attenuated the production of Aβ [231]. Neurotropin® (NTP), a well-known drug for chronic pain, was found to stimulate BDNF expression in SH-SY5Y cells and restore the declined expression of BDNF in the hippocampus [232]. It was reported that Neurotropin® possesses a neuroprotective character by lessening Aβ-induced oxidative damage and alleviating Aβ deposition in the hippocampus through the downregulation of the HIF-1α/MAPK signaling cascade [233]. By transferring mitochondria from a living sporadic AD [205] patient into mitochondrial DNA (mtDNA)-free SH-SY5Y cells, mitochondrial transgenic neuronal cells, or cybrids, can be produced [234,235]. Using the cybrids of SAD and age-matched controls, it was demonstrated that, under hypoxic conditions with different dosages of simvastatin, the extents of HIF-1α and BACE induction may vary in these cybrids. In the low-dose simvastatin group, the reduced expression of HIF-1α and BACE1 was observed in the SAD cybrids as compared to the controls. In the high-dose simvastatin group, however, the heightened expression of HIF-1α and BACE1 was observed in both the SAD and the control cybrids. These results denote the potential usefulness of low-dose simvastatin treatment in attenuating the expression of HIF-1α and BACE to lower Aβ production [236]. In human neuroblastoma SH-SY5Y cells under the in vitro ischemic condition of OGD and OGD with reoxygenation (OGD/R), HIF-1α was highly upregulated at both the mRNA and protein levels, which were downregulated by melatonin; consistently, the heightened BACE1 mRNA and proteins under such ischemic condition were suppressed by melatonin [237]. However, a direct causal relationship between melatonin-induced HIF-1α and BACE1 under ischemic conditions was not established in this study.
Table 2. List of examples with detrimental roles of HIF-1 in AD-related studies.
Table 2. List of examples with detrimental roles of HIF-1 in AD-related studies.
Chemical Compound, Drug, Nutrient, ProteinMechanismsStudy ModelReference
Fibroblast growth factor 21 (FGF21)inhibition of PP2A/MAPKs/HIF-1α pathway triggered by Aβ25-35in vitro: SH-SY5Y human neuroblastoma cell line
in vivo: adult male Wistar rats
[216]
HB-EGFincreased HIF-1α expression can activate MMP9 to cause BBB disintegrationin vivo: BCCAO mouse model,
CCH manifests AD neuropathology
[203]
Neuregulin 1inhibited CoCl2-induced accumulation of HIF-1α and p53 stability to attenuate cell deathin vitro: SH-SY5Y human neuroblastoma cell line[221]
High glucose condition upregulated BACE1-mediated Aβ production through HIF-1α activation, decreased LXRα expression via the JNK pathway in a ROS-dependent mannerin vitro: SK-N-MC neuroblastoma cell line
in vivo: homozygous ZDF and ZLC rats
[222]
Thiamine insufficiency (TI)triggered HIF-1-mediated amyloidogenesis through transcriptional expression of BACE1 and increased activity of β-secretase-
induced HIF-1-dependent expression of pro-apoptotic protein BNIP3
in vitro: HT22 hippocampal neuronal cell line[223]
Ginsenoside Rg1, active ingredients of Panax ginsengreduced the Aβ-induced mitochondrial apoptosis, lessened HIF-1α expression, decreased RNS and protein nitrotyrosinationin vitro: human endothelial cells[226]
Salidroside, a glucoside of tyrosol found in the plant Rhodiola roseareduced protein level of HIF-1α under hypoxia; decreased expression of BACE1 and inhibited β-secretase activity, thus enhancing sAPPα secretion and attenuating Aβ generation induced by hypoxiain vitro: SH-SY5Y human neuroblastoma cell line[227]
PA-BSAinduced expression of APP and BACE1, accelerated Aβ production via Akt/mTOR/HIF-1α and Akt/NF-κB pathwaysin vitro: SK-N-MC neuroblastoma cell line[231]
NTP, a non-protein extract of inflamed rabbit skin inoculated with vaccinia virus clinically used for the treatment of neuropathic painlessened Aβ-induced oxidative damage, improved Aβ deposition in hippocampus through regulating HIF-1α/MAPK signaling pathwayin vitro: HT22 hippocampal cells
in vivo: APP/PS1 Tg mice
[233]
Simvastatin, in low dosage reduced HIF-1α and BACE1 expressionin vitro: sporadic AD and age-matched control neuronal mitochondrial cybrid [236]
Melatoninsuppressed HIF-1α and BACE1 expression under the in vitro ischemic condition induced by OGD and OGD/Rin vitro: SH-SY5Y human neuroblastoma cell line [237]
BCCAO, bilateral common carotid artery occlusion; BNIP3, BCL2/adenovirus E1B 19-kDa protein-interacting protein; CCH, chronic cerebral hypoperfusion; HB-EGF, heparin-binding EGF-like growth factor; NTP, Neurotropin®; OGD and OGD/R, oxygen–glucose deprivation and OGD with reoxygenation; PA-BSA, palmitic acid-bovine serum albumin; RNS, reactive nitrogen species; TI, thiamine insufficiency; ZDF and ZLC rats, Zucker diabetic fatty and Zucker lean control fatty rat.

4. Conclusions and Future Perspectives

AD is the most prevalent neurodegenerative disease in the aging population with progressive memory impairment, deteriorated cognition, and behavioral dysfunctions. At present, there is no effective treatment for this devastating disease. Any means to delay or lessen the consequences of AD may therefore represent a significant improvement for patients, their families, and societies. HIF-1α, a master regulator for gene expression under hypoxic conditions, can increase cell proliferation, accelerate new blood vessel formation, and regulate glucose and energy metabolisms to cope with a stressful cellular milieu such as hypoxia. HIF-1α is also known to be activated under oxidative stress, neuroinflammation, and cerebral hypoperfusion, the conditions often accompanied by AD. Increasing the transcriptional activity of HIF-1α can enhance angiogenesis and erythropoiesis, trigger anti-apoptotic cascades, and regulate autophagy, and thus represent a potential therapeutic scheme in the treatment of neurodegenerative disorders including AD. In contrast, however, ample evidence also indicates that HIF-1α inhibition might exert a beneficial effect on AD. Additionally, the effect of the HIF signaling greatly depends on the extent of hypoxic magnitude, severity of the stress, and the cell types that express HIF-1α. Since HIF-1 signaling could be either beneficial or detrimental, there is a pressing need to design highly selective modulators, either activators or inhibitors, for manipulating the HIF-1 transduction pathway as well as effective means of the targeted delivery of these HIF-1 modulators into proper brain regions or even cell types for AD therapy.

Author Contributions

Conceptualization, T.-K.L. and C.-R.H.; writing—original draft preparation, S.-D.C.; writing—review and editing, T.-K.L., C.-R.H., Y.-C.L., Y.-H.H., and D.-I.Y.; visualization, K.-J.L., and Y.-H.H.; supervision, A.-C.C. and D.-I.Y.; funding acquisition, Y.-C.L., A.-C.C., and D.-I.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by by the National Science and Technology Council in Taiwan (NSTC 111-2314-B-A49-068-MY3 and NSTC 112-2320-B-A49-029-MY3 to Ding-I Yang; NSTC 112-2314-B-037-061-MY3 to A-Ching Chao) and the Department of Health, Taipei City Government in Taiwan (11101-62-006 and 11201-62-043 to Ding-I Yang and Yi-Chun Lin). Yi-Heng Hsieh is a post-doctoral research fellow supported by the National Science and Technology Council (NSTC 112-2811-B-A49A-033 and NSTC 113-2811-B-A49A-021). This study was also financially supported by Kaohsiung Medical University Hospital (KMUH112-2R65 to A-Ching Chao) and by the Brain Research Center, National Yang Ming Chiao Tung University from The Featured Areas Research Center Program within the framework of the Higher Education Sprout Project by the Ministry of Education (MOE) in Taiwan (113W032101).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Dugger, B.N.; Dickson, D.W. Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef] [PubMed]
  2. Hung, C.W.; Chen, Y.C.; Hsieh, W.L.; Chiou, S.H.; Kao, C.L. Ageing and neurodegenerative diseases. Ageing Res. Rev. 2010, 9 (Suppl. 1), S36–S46. [Google Scholar] [CrossRef]
  3. Takizawa, C.; Thompson, P.L.; van Walsem, A.; Faure, C.; Maier, W.C. Epidemiological and economic burden of Alzheimer’s disease: A systematic literature review of data across Europe and the United States of America. J. Alzheimers Dis. 2015, 43, 1271–1284. [Google Scholar] [CrossRef] [PubMed]
  4. Skaria, A.P. The economic and societal burden of Alzheimer disease: Managed care considerations. Am. J. Manag. Care 2022, 28, S188–S196. [Google Scholar]
  5. 2023 Alzheimer’s disease facts and figures. Alzheimers Dement. 2023, 19, 1598–1695. [CrossRef]
  6. Acker, T.; Acker, H. Cellular oxygen sensing need in CNS function: Physiological and pathological implications. J. Exp. Biol. 2004, 207, 3171–3188. [Google Scholar] [CrossRef]
  7. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [PubMed]
  8. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef]
  9. Murphy, M.P.; LeVine, H., 3rd. Alzheimer’s disease and the amyloid-beta peptide. J. Alzheimers Dis. 2010, 19, 311–323. [Google Scholar] [CrossRef]
  10. O’Brien, R.J.; Wong, P.C. Amyloid precursor protein processing and Alzheimer’s disease. Annu. Rev. Neurosci. 2011, 34, 185–204. [Google Scholar] [CrossRef]
  11. Vardy, E.R.; Catto, A.J.; Hooper, N.M. Proteolytic mechanisms in amyloid-beta metabolism: Therapeutic implications for Alzheimer’s disease. Trends Mol. Med. 2005, 11, 464–472. [Google Scholar] [CrossRef] [PubMed]
  12. Flammang, B.; Pardossi-Piquard, R.; Sevalle, J.; Debayle, D.; Dabert-Gay, A.S.; Thevenet, A.; Lauritzen, I.; Checler, F. Evidence that the amyloid-beta protein precursor intracellular domain, AICD, derives from beta-secretase-generated C-terminal fragment. J. Alzheimers Dis. 2012, 30, 145–153. [Google Scholar] [CrossRef] [PubMed]
  13. Esposito, Z.; Belli, L.; Toniolo, S.; Sancesario, G.; Bianconi, C.; Martorana, A. Amyloid β, glutamate, excitotoxicity in Alzheimer’s disease: Are we on the right track? CNS Neurosci. Ther. 2013, 19, 549–555. [Google Scholar] [CrossRef]
  14. Ju, T.C.; Chen, S.D.; Liu, C.C.; Yang, D.I. Protective effects of S-nitrosoglutathione against amyloid beta-peptide neurotoxicity. Free Radic. Biol. Med. 2005, 38, 938–949. [Google Scholar] [CrossRef]
  15. Chao, A.C.; Chen, C.H.; Chang, S.H.; Huang, C.T.; Hwang, W.C.; Yang, D.I. Id1 and sonic hedgehog mediate cell cycle reentry and apoptosis induced by amyloid beta-peptide in post-mitotic cortical neurons. Mol. Neurobiol. 2019, 56, 465–489. [Google Scholar] [CrossRef]
  16. Chao, A.C.; Chen, C.H.; Wu, M.H.; Hou, B.Y.; Yang, D.I. Roles of Id1/HIF-1 and CDK5/HIF-1 in cell cycle reentry induced by amyloid-beta peptide in post-mitotic cortical neuron. Biochim. Biophys. Acta—Mol. Cell Res. 2020, 1867, 118628. [Google Scholar] [CrossRef]
  17. Caldeira, G.L.; Ferreira, I.L.; Rego, A.C. Impaired transcription in Alzheimer’s disease: Key role in mitochondrial dysfunction and oxidative stress. J. Alzheimers Dis. 2013, 34, 115–131. [Google Scholar] [CrossRef] [PubMed]
  18. Wu, M.F.; Yin, J.H.; Hwang, C.S.; Tang, C.M.; Yang, D.I. NAD attenuates oxidative DNA damages induced by amyloid beta-peptide in primary rat cortical neurons. Free Radic. Res. 2014, 48, 794–805. [Google Scholar] [CrossRef]
  19. Lauretti, E.; Praticò, D. Alzheimer’s disease: Phenotypic approaches using disease models and the targeting of tau protein. Expert Opin. Ther. Targets 2020, 24, 319–330. [Google Scholar] [CrossRef]
  20. Vicario-Orri, E.; Opazo, C.M.; Muñoz, F.J. The pathophysiology of axonal transport in Alzheimer’s disease. J. Alzheimers Dis. 2015, 43, 1097–1113. [Google Scholar] [CrossRef]
  21. Majmundar, A.J.; Wong, W.J.; Simon, M.C. Hypoxia-inducible factors and the response to hypoxic stress. Mol. Cell 2010, 40, 294–309. [Google Scholar] [CrossRef] [PubMed]
  22. Goda, N.; Kanai, M. Hypoxia-inducible factors and their roles in energy metabolism. Int. J. Hematol. 2012, 95, 457–463. [Google Scholar] [CrossRef]
  23. Semenza, G.L. Oxygen sensing, homeostasis, and disease. N. Engl. J. Med. 2011, 365, 537–547. [Google Scholar] [CrossRef] [PubMed]
  24. Semenza, G.L. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J. Clin. Investig. 2013, 123, 3664–3671. [Google Scholar] [CrossRef]
  25. Choudhry, H.; Harris, A.L. Advances in hypoxia-inducible factor biology. Cell Metab. 2018, 27, 281–298. [Google Scholar] [CrossRef] [PubMed]
  26. Semenza, G.L. Hypoxia-inducible factors in physiology and medicine. Cell 2012, 148, 399–408. [Google Scholar] [CrossRef]
  27. Serocki, M.; Bartoszewska, S.; Janaszak-Jasiecka, A.; Ochocka, R.J.; Collawn, J.F.; Bartoszewski, R. miRNAs regulate the HIF switch during hypoxia: A novel therapeutic target. Angiogenesis 2018, 21, 183–202. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, G.L.; Jiang, B.H.; Rue, E.A.; Semenza, G.L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc. Natl. Acad. Sci. USA 1995, 92, 5510–5514. [Google Scholar] [CrossRef] [PubMed]
  29. Wang, G.L.; Semenza, G.L. Purification and characterization of hypoxia-inducible factor 1. J. Biol. Chem. 1995, 270, 1230–1237. [Google Scholar] [CrossRef]
  30. Ivan, M.; Kondo, K.; Yang, H.; Kim, W.; Valiando, J.; Ohh, M.; Salic, A.; Asara, J.M.; Lane, W.S.; Kaelin, W.G., Jr. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: Implications for O2 sensing. Science 2001, 292, 464–468. [Google Scholar] [CrossRef]
  31. Jaakkola, P.; Mole, D.R.; Tian, Y.M.; Wilson, M.I.; Gielbert, J.; Gaskell, S.J.; von Kriegsheim, A.; Hebestreit, H.F.; Mukherji, M.; Schofield, C.J.; et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001, 292, 468–472. [Google Scholar] [CrossRef] [PubMed]
  32. Chen, S.D.; Hu, C.J.; Yang, D.I.; Nassief, A.; Chen, H.; Yin, K.; Xu, J.; Hsu, C.Y. Pravastatin attenuates ceramide-induced cytotoxicity in mouse cerebral endothelial cells with HIF-1 activation and VEGF upregulation. Ann. N. Y Acad. Sci. 2005, 1042, 357–364. [Google Scholar] [CrossRef]
  33. Yang, D.I.; Chen, S.D.; Yang, Y.T.; Ju, T.C.; Xu, J.M.; Hsu, C.Y. Carbamoylating chemoresistance induced by cobalt pretreatment in C6 glioma cells: Putative roles of hypoxia-inducible factor-1. Br. J. Pharmacol. 2004, 141, 988–996. [Google Scholar] [CrossRef]
  34. Zaman, K.; Ryu, H.; Hall, D.; O’Donovan, K.; Lin, K.I.; Miller, M.P.; Marquis, J.C.; Baraban, J.M.; Semenza, G.L.; Ratan, R.R. Protection from oxidative stress-induced apoptosis in cortical neuronal cultures by iron chelators is associated with enhanced DNA binding of hypoxia-inducible factor-1 and ATF-1/CREB and increased expression of glycolytic enzymes, p21(waf1/cip1), and erythropoietin. J. Neurosci. 1999, 19, 9821–9830. [Google Scholar] [PubMed]
  35. Jiang, B.H.; Rue, E.; Wang, G.L.; Roe, R.; Semenza, G.L. Dimerization, DNA binding, and transactivation properties of hypoxia-inducible factor 1. J. Biol. Chem. 1996, 271, 17771–17778. [Google Scholar] [CrossRef]
  36. Semenza, G.L.; Agani, F.; Booth, G.; Forsythe, J.; Iyer, N.; Jiang, B.H.; Leung, S.; Roe, R.; Wiener, C.; Yu, A. Structural and functional analysis of hypoxia-inducible factor 1. Kidney Int. 1997, 51, 553–555. [Google Scholar] [CrossRef] [PubMed]
  37. Mole, D.R.; Blancher, C.; Copley, R.R.; Pollard, P.J.; Gleadle, J.M.; Ragoussis, J.; Ratcliffe, P.J. Genome-wide association of hypoxia-inducible factor (HIF)-1alpha and HIF-2alpha DNA binding with expression profiling of hypoxia-inducible transcripts. J. Biol. Chem. 2009, 284, 16767–16775. [Google Scholar] [CrossRef] [PubMed]
  38. Semenza, G.L. HIF-1 and tumor progression: Pathophysiology and therapeutics. Trends Mol. Med. 2002, 8, S62–S67. [Google Scholar] [CrossRef]
  39. Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef]
  40. Balamurugan, K. HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int. J. Cancer 2016, 138, 1058–1066. [Google Scholar] [CrossRef]
  41. Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 2019, 18, 157. [Google Scholar] [CrossRef] [PubMed]
  42. You, L.; Wu, W.; Wang, X.; Fang, L.; Adam, V.; Nepovimova, E.; Wu, Q.; Kuca, K. The role of hypoxia-inducible factor 1 in tumor immune evasion. Med. Res. Rev. 2021, 41, 1622–1643. [Google Scholar] [CrossRef]
  43. Soni, S.; Padwad, Y.S. HIF-1 in cancer therapy: Two decade long story of a transcription factor. Acta Oncol. 2017, 56, 503–515. [Google Scholar] [CrossRef]
  44. Kao, T.W.; Bai, G.H.; Wang, T.L.; Shih, I.M.; Chuang, C.M.; Lo, C.L.; Tsai, M.C.; Chiu, L.Y.; Lin, C.C.; Shen, Y.A. Novel cancer treatment paradigm targeting hypoxia-induced factor in conjunction with current therapies to overcome resistance. J. Exp. Clin. Cancer Res. 2023, 42, 171. [Google Scholar] [CrossRef]
  45. Nakanishi, T.; Kuragano, T. Growing concerns about using hypoxia-inducible factor prolyl hydroxylase inhibitors for the treatment of renal anemia. Clin. Kidney J. 2024, 17, sfae051. [Google Scholar] [CrossRef] [PubMed]
  46. McGettrick, A.F.; O’Neill, L.A.J. The role of HIF in immunity and inflammation. Cell Metab. 2020, 32, 524–536. [Google Scholar] [CrossRef]
  47. Takeda, N.; O’Dea, E.L.; Doedens, A.; Kim, J.W.; Weidemann, A.; Stockmann, C.; Asagiri, M.; Simon, M.C.; Hoffmann, A.; Johnson, R.S. Differential activation and antagonistic function of HIF-{alpha} isoforms in macrophages are essential for NO homeostasis. Genes Dev. 2010, 24, 491–501. [Google Scholar] [CrossRef] [PubMed]
  48. Corcoran, S.E.; O’Neill, L.A. HIF1α and metabolic reprogramming in inflammation. J. Clin. Investig. 2016, 126, 3699–3707. [Google Scholar] [CrossRef]
  49. Wang, T.; Liu, H.; Lian, G.; Zhang, S.Y.; Wang, X.; Jiang, C. HIF1α-induced glycolysis metabolism is essential to the activation of inflammatory macrophages. Mediat. Inflamm. 2017, 2017, 9029327. [Google Scholar] [CrossRef]
  50. Jantsch, J.; Chakravortty, D.; Turza, N.; Prechtel, A.T.; Buchholz, B.; Gerlach, R.G.; Volke, M.; Gläsner, J.; Warnecke, C.; Wiesener, M.S.; et al. Hypoxia and hypoxia-inducible factor-1 alpha modulate lipopolysaccharide-induced dendritic cell activation and function. J. Immunol. 2008, 180, 4697–4705. [Google Scholar] [CrossRef]
  51. Naldini, A.; Morena, E.; Pucci, A.; Miglietta, D.; Riboldi, E.; Sozzani, S.; Carraro, F. Hypoxia affects dendritic cell survival: Role of the hypoxia-inducible factor-1α and lipopolysaccharide. J. Cell. Physiol. 2012, 227, 587–595. [Google Scholar] [CrossRef] [PubMed]
  52. McInturff, A.M.; Cody, M.J.; Elliott, E.A.; Glenn, J.W.; Rowley, J.W.; Rondina, M.T.; Yost, C.C. Mammalian target of rapamycin regulates neutrophil extracellular trap formation via induction of hypoxia-inducible factor 1 α. Blood 2012, 120, 3118–3125. [Google Scholar] [CrossRef] [PubMed]
  53. Dang, E.V.; Barbi, J.; Yang, H.Y.; Jinasena, D.; Yu, H.; Zheng, Y.; Bordman, Z.; Fu, J.; Kim, Y.; Yen, H.R.; et al. Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell 2011, 146, 772–784. [Google Scholar] [CrossRef] [PubMed]
  54. Shi, L.Z.; Wang, R.; Huang, G.; Vogel, P.; Neale, G.; Green, D.R.; Chi, H. HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J. Exp. Med. 2011, 208, 1367–1376. [Google Scholar] [CrossRef]
  55. Higashiyama, M.; Hokari, R.; Hozumi, H.; Kurihara, C.; Ueda, T.; Watanabe, C.; Tomita, K.; Nakamura, M.; Komoto, S.; Okada, Y.; et al. HIF-1 in T cells ameliorated dextran sodium sulfate-induced murine colitis. J. Leukoc. Biol. 2012, 91, 901–909. [Google Scholar] [CrossRef]
  56. Kojima, H.; Gu, H.; Nomura, S.; Caldwell, C.C.; Kobata, T.; Carmeliet, P.; Semenza, G.L.; Sitkovsky, M.V. Abnormal B lymphocyte development and autoimmunity in hypoxia-inducible factor 1alpha-deficient chimeric mice. Proc. Natl. Acad. Sci. USA 2002, 99, 2170–2174. [Google Scholar] [CrossRef]
  57. Kojima, H.; Kobayashi, A.; Sakurai, D.; Kanno, Y.; Hase, H.; Takahashi, R.; Totsuka, Y.; Semenza, G.L.; Sitkovsky, M.V.; Kobata, T. Differentiation stage-specific requirement in hypoxia-inducible factor-1alpha-regulated glycolytic pathway during murine B cell development in bone marrow. J. Immunol. 2010, 184, 154–163. [Google Scholar] [CrossRef]
  58. Meng, X.; Grötsch, B.; Luo, Y.; Knaup, K.X.; Wiesener, M.S.; Chen, X.X.; Jantsch, J.; Fillatreau, S.; Schett, G.; Bozec, A. Hypoxia-inducible factor-1α is a critical transcription factor for IL-10-producing B cells in autoimmune disease. Nat. Commun. 2018, 9, 251. [Google Scholar] [CrossRef]
  59. Qian, T.; Hong, J.; Wang, L.; Wang, Z.; Lu, Z.; Li, Y.; Liu, R.; Chu, Y. Regulation of CD11b by HIF-1α and the STAT3 signaling pathway contributes to the immunosuppressive function of B cells in inflammatory bowel disease. Mol. Immunol. 2019, 111, 162–171. [Google Scholar] [CrossRef]
  60. Bae, Y.S.; Oh, H.; Rhee, S.G.; Yoo, Y.D. Regulation of reactive oxygen species generation in cell signaling. Mol. Cells 2011, 32, 491–509. [Google Scholar] [CrossRef]
  61. Novo, E.; Parola, M. The role of redox mechanisms in hepatic chronic wound healing and fibrogenesis. Fibrogenesis Tissue Repair 2012, 5, S4. [Google Scholar] [CrossRef] [PubMed]
  62. Veith, A.; Moorthy, B. Role of cytochrome P450S in the generation and metabolism of reactive oxygen species. Curr. Opin. Toxicol. 2018, 7, 44–51. [Google Scholar] [CrossRef] [PubMed]
  63. Hajam, Y.A.; Rani, R.; Ganie, S.Y.; Sheikh, T.A.; Javaid, D.; Qadri, S.S.; Pramodh, S.; Alsulimani, A.; Alkhanani, M.F.; Harakeh, S.; et al. Oxidative stress in human pathology and aging: Molecular mechanisms and perspectives. Cells 2022, 11, 552. [Google Scholar] [CrossRef] [PubMed]
  64. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  65. Dan Dunn, J.; Alvarez, L.A.; Zhang, X.; Soldati, T. Reactive oxygen species and mitochondria: A nexus of cellular homeostasis. Redox Biol. 2015, 6, 472–485. [Google Scholar] [CrossRef]
  66. Lin, T.K.; Chen, S.D.; Lin, K.J.; Chuang, Y.C. Seizure-induced oxidative stress in status epilepticus: Is antioxidant beneficial? Antioxidants 2020, 9, 1029. [Google Scholar] [CrossRef]
  67. Liu, Z.; Zhou, T.; Ziegler, A.C.; Dimitrion, P.; Zuo, L. Oxidative stress in neurodegenerative diseases: From molecular mechanisms to clinical applications. Oxid. Med. Cell. Longev. 2017, 2017, 2525967. [Google Scholar] [CrossRef]
  68. Balaban, R.S.; Nemoto, S.; Finkel, T. Mitochondria, oxidants, and aging. Cell 2005, 120, 483–495. [Google Scholar] [CrossRef]
  69. Chen, S.D.; Lin, T.K.; Yang, D.I.; Lee, S.Y.; Shaw, F.Z.; Liou, C.W.; Chuang, Y.C. Protective effects of peroxisome proliferator-activated receptors gamma coactivator-1alpha against neuronal cell death in the hippocampal CA1 subfield after transient global ischemia. J. Neurosci. Res. 2010, 88, 605–613. [Google Scholar] [CrossRef]
  70. Leonard, J.V.; Schapira, A.H. Mitochondrial respiratory chain disorders II: Neurodegenerative disorders and nuclear gene defects. Lancet 2000, 355, 389–394. [Google Scholar] [CrossRef]
  71. Haddad, J.J. Science review: Redox and oxygen-sensitive transcription factors in the regulation of oxidant-mediated lung injury: Role for hypoxia-inducible factor-1alpha. Crit. Care 2003, 7, 47–54. [Google Scholar] [CrossRef] [PubMed]
  72. Harten, S.K.; Ashcroft, M.; Maxwell, P.H. Prolyl hydroxylase domain inhibitors: A route to HIF activation and neuroprotection. Antioxid. Redox Signal. 2010, 12, 459–480. [Google Scholar] [CrossRef] [PubMed]
  73. Kapitsinou, P.P.; Haase, V.H. Molecular mechanisms of ischemic preconditioning in the kidney. Am. J. Physiol. Renal Physiol. 2015, 309, F821–F834. [Google Scholar] [CrossRef] [PubMed]
  74. Loor, G.; Schumacker, P.T. Role of hypoxia-inducible factor in cell survival during myocardial ischemia-reperfusion. Cell Death Differ. 2008, 15, 686–690. [Google Scholar] [CrossRef] [PubMed]
  75. Pan, Z.; Ma, G.; Kong, L.; Du, G. Hypoxia-inducible factor-1: Regulatory mechanisms and drug development in stroke. Pharmacol. Res. 2021, 170, 105742. [Google Scholar] [CrossRef]
  76. Hashmi, S.; Al-Salam, S. Hypoxia-inducible factor-1 alpha in the heart: A double agent? Cardiol. Rev. 2012, 20, 268–273. [Google Scholar] [CrossRef]
  77. Datta Chaudhuri, R.; Banik, A.; Mandal, B.; Sarkar, S. Cardiac-specific overexpression of HIF-1alpha during acute myocardial infarction ameliorates cardiomyocyte apoptosis via differential regulation of hypoxia-inducible pro-apoptotic and anti-oxidative genes. Biochem. Biophys. Res. Commun. 2021, 537, 100–108. [Google Scholar] [CrossRef]
  78. Yang, Z.Z.; Zou, A.P. Transcriptional regulation of heme oxygenases by HIF-1alpha in renal medullary interstitial cells. Am. J. Physiol. Renal Physiol. 2001, 281, F900–F908. [Google Scholar] [CrossRef]
  79. Lee, H.A.; Chu, K.B.; Moon, E.K.; Quan, F.S. Glutathione peroxidase 8 suppression by histone deacetylase inhibitors enhances endoplasmic reticulum stress and cell death by oxidative stress in hepatocellular carcinoma cells. Antioxidants 2021, 10, 1503. [Google Scholar] [CrossRef]
  80. Bosello-Travain, V.; Forman, H.J.; Roveri, A.; Toppo, S.; Ursini, F.; Venerando, R.; Warnecke, C.; Zaccarin, M.; Maiorino, M. Glutathione peroxidase 8 is transcriptionally regulated by HIFalpha and modulates growth factor signaling in HeLa cells. Free Radic. Biol. Med. 2015, 81, 58–68. [Google Scholar] [CrossRef]
  81. Qausain, S.; Basheeruddin, M. Unraveling the peroxidase activity in peroxiredoxins: A comprehensive review of mechanisms, functions, and biological significance. Cureus 2024, 16, e66117. [Google Scholar] [CrossRef] [PubMed]
  82. Luo, W.; Chen, I.; Chen, Y.; Alkam, D.; Wang, Y.; Semenza, G.L. PRDX2 and PRDX4 are negative regulators of hypoxia-inducible factors under conditions of prolonged hypoxia. Oncotarget 2016, 7, 6379–6397. [Google Scholar] [CrossRef] [PubMed]
  83. Jie, Y.K.; Cheng, C.H.; Ma, H.L.; Liu, G.X.; Fan, S.G.; Jiang, J.J.; Guo, Z.X. Hypoxia affects the antioxidant activity of glutaredoxin 3 in Scylla paramamosain through hypoxia response elements. Antioxidants 2022, 12, 76. [Google Scholar] [CrossRef]
  84. Catchlove, S.J.; Macpherson, H.; Hughes, M.E.; Chen, Y.; Parrish, T.B.; Pipingas, A. An investigation of cerebral oxygen utilization, blood flow and cognition in healthy aging. PLoS ONE 2018, 13, e0197055. [Google Scholar] [CrossRef]
  85. Salvatore, S.S.; Zelenski, K.N.; Perkins, R.K. Age-related changes in skeletal muscle oxygen utilization. J. Funct. Morphol. Kinesiol. 2022, 7, 87. [Google Scholar] [CrossRef]
  86. Bennett, H.C.; Zhang, Q.; Wu, Y.T.; Manjila, S.B.; Chon, U.; Shin, D.; Vanselow, D.J.; Pi, H.J.; Drew, P.J.; Kim, Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat. Commun. 2024, 15, 6398. [Google Scholar] [CrossRef]
  87. Ogunshola, O.O.; Antoniou, X. Contribution of hypoxia to Alzheimer’s disease: Is HIF-1alpha a mediator of neurodegeneration? Cell. Mol. Life Sci. 2009, 66, 3555–3563. [Google Scholar] [CrossRef] [PubMed]
  88. Hambali, A.; Kumar, J.; Hashim, N.F.M.; Maniam, S.; Mehat, M.Z.; Cheema, M.S.; Mustapha, M.; Adenan, M.I.; Stanslas, J.; Hamid, H.A. Hypoxia-induced neuroinflammation in Alzheimer’s disease: Potential neuroprotective effects of Centella asiatica. Front. Physiol. 2021, 12, 712317. [Google Scholar] [CrossRef]
  89. Zhang, F.; Niu, L.; Li, S.; Le, W. Pathological impacts of chronic hypoxia on Alzheimer’s disease. ACS Chem. Neurosci. 2019, 10, 902–909. [Google Scholar] [CrossRef]
  90. Yaffe, K.; Laffan, A.M.; Harrison, S.L.; Redline, S.; Spira, A.P.; Ensrud, K.E.; Ancoli-Israel, S.; Stone, K.L. Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA 2011, 306, 613–619. [Google Scholar] [CrossRef]
  91. Ferda, J.; Ferdová, E.; Vítovec, M.; Glanc, D.; Mírka, H. The imaging of the hypoxic microenvironment in tumorous tissue using PET/CT and PET/MRI. Eur. J. Radiol. 2022, 154, 110458. [Google Scholar] [CrossRef] [PubMed]
  92. Correia, S.C.; Moreira, P.I. Oxygen sensing and signaling in Alzheimer’s disease: A breathtaking story! Cell. Mol. Neurobiol. 2022, 42, 3–21. [Google Scholar] [CrossRef] [PubMed]
  93. Barat, M. Disorders of gestural expression in aphasics. Rev. Laryngol. Otol. Rhinol. 1987, 108, 315–321. [Google Scholar]
  94. Bomboi, G.; Castello, L.; Cosentino, F.; Giubilei, F.; Orzi, F.; Volpe, M. Alzheimer’s disease and endothelial dysfunction. Neurol. Sci. 2010, 31, 1–8. [Google Scholar] [CrossRef]
  95. Iadecola, C. The neurovascular unit coming of age: A journey through neurovascular coupling in health and disease. Neuron 2017, 96, 17–42. [Google Scholar] [CrossRef] [PubMed]
  96. Shang, J.; Yamashita, T.; Tian, F.; Li, X.; Liu, X.; Shi, X.; Nakano, Y.; Tsunoda, K.; Nomura, E.; Sasaki, R.; et al. Chronic cerebral hypoperfusion alters amyloid-β transport related proteins in the cortical blood vessels of Alzheimer’s disease model mouse. Brain Res. 2019, 1723, 146379. [Google Scholar] [CrossRef]
  97. Zlokovic, B.V. Neurovascular mechanisms of Alzheimer’s neurodegeneration. Trends Neurosci. 2005, 28, 202–208. [Google Scholar] [CrossRef]
  98. Salminen, A. Hypoperfusion is a potential inducer of immunosuppressive network in Alzheimer’s disease. Neurochem. Int. 2021, 142, 104919. [Google Scholar] [CrossRef] [PubMed]
  99. Laing, K.K.; Simoes, S.; Baena-Caldas, G.P.; Lao, P.J.; Kothiya, M.; Igwe, K.C.; Chesebro, A.G.; Houck, A.L.; Pedraza, L.; Hernández, A.I.; et al. Cerebrovascular disease promotes tau pathology in Alzheimer’s disease. Brain Commun. 2020, 2, fcaa132. [Google Scholar] [CrossRef]
  100. Hébert, F.; Grand’maison, M.; Ho, M.K.; Lerch, J.P.; Hamel, E.; Bedell, B.J. Cortical atrophy and hypoperfusion in a transgenic mouse model of Alzheimer’s disease. Neurobiol. Aging 2013, 34, 1644–1652. [Google Scholar] [CrossRef]
  101. Huang, C.W.; Hsu, S.W.; Chang, Y.T.; Huang, S.H.; Huang, Y.C.; Lee, C.C.; Chang, W.N.; Lui, C.C.; Chen, N.C.; Chang, C.C. Cerebral perfusion insufficiency and relationships with cognitive deficits in Alzheimer’s disease: A multiparametric neuroimaging study. Sci. Rep. 2018, 8, 1541. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, B.; Hua, R.; Qing, Z.; Ni, L.; Zhang, X.; Zhao, H.; Liu, R.; Lu, J.; Wu, S.; Xu, Y.; et al. Abnormal brain functional connectivity coupled with hypoperfusion measured by Resting-State fMRI: An additional contributing factor for cognitive impairment in patients with Alzheimer’s disease. Psychiatry Res. Neuroimaging 2019, 289, 18–25. [Google Scholar] [CrossRef] [PubMed]
  103. Hong, C.H.; Falvey, C.; Harris, T.B.; Simonsick, E.M.; Satterfield, S.; Ferrucci, L.; Metti, A.L.; Patel, K.V.; Yaffe, K. Anemia and risk of dementia in older adults: Findings from the Health ABC study. Neurology 2013, 81, 528–533. [Google Scholar] [CrossRef]
  104. Atti, A.R.; Palmer, K.; Volpato, S.; Zuliani, G.; Winblad, B.; Fratiglioni, L. Anaemia increases the risk of dementia in cognitively intact elderly. Neurobiol. Aging 2006, 27, 278–284. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, Z.; Yan, J.; Chang, Y.; ShiDu Yan, S.; Shi, H. Hypoxia inducible factor-1 as a target for neurodegenerative diseases. Curr. Med. Chem. 2011, 18, 4335–4343. [Google Scholar] [CrossRef]
  106. Mitroshina, E.V.; Vedunova, M.V. The role of oxygen homeostasis and the HIF-1 factor in the development of neurodegeneration. Int. J. Mol. Sci. 2024, 25, 4581. [Google Scholar] [CrossRef]
  107. Mitroshina, E.V.; Savyuk, M.O.; Ponimaskin, E.; Vedunova, M.V. Hypoxia-inducible factor (HIF) in ischemic stroke and neurodegenerative disease. Front. Cell Dev. Biol. 2021, 9, 703084. [Google Scholar] [CrossRef]
  108. Adeyemi, O.S.; Awakan, O.J.; Afolabi, L.B.; Rotimi, D.E.; Oluwayemi, E.; Otuechere, C.A.; Ibraheem, O.; Elebiyo, T.C.; Alejolowo, O.; Arowolo, A.T. Hypoxia and the kynurenine pathway: Implications and therapeutic prospects in Alzheimer’s disease. Oxid. Med. Cell. Longev. 2021, 2021, 5522981. [Google Scholar] [CrossRef]
  109. Merelli, A.; Rodríguez, J.C.G.; Folch, J.; Regueiro, M.R.; Camins, A.; Lazarowski, A. Understanding the role of hypoxia inducible factor during neurodegeneration for new therapeutics opportunities. Curr. Neuropharmacol. 2018, 16, 1484–1498. [Google Scholar] [CrossRef]
  110. Liu, Y.; Liu, F.; Iqbal, K.; Grundke-Iqbal, I.; Gong, C.X. Decreased glucose transporters correlate to abnormal hyperphosphorylation of tau in Alzheimer disease. FEBS Lett. 2008, 582, 359–364. [Google Scholar] [CrossRef]
  111. Grammas, P.; Samany, P.G.; Thirumangalakudi, L. Thrombin and inflammatory proteins are elevated in Alzheimer’s disease microvessels: Implications for disease pathogenesis. J. Alzheimers Dis. 2006, 9, 51–58. [Google Scholar] [CrossRef] [PubMed]
  112. Grammas, P.; Tripathy, D.; Sanchez, A.; Yin, X.; Luo, J. Brain microvasculature and hypoxia-related proteins in Alzheimer’s disease. Int. J. Clin. Exp. Pathol. 2011, 4, 616–627. [Google Scholar] [PubMed]
  113. Jung, E.; Kim, Y.E.; Jeon, H.S.; Yoo, M.; Kim, M.; Kim, Y.M.; Koh, S.H.; Choi, Y.K. Chronic hypoxia of endothelial cells boosts HIF-1alpha-NLRP1 circuit in Alzheimer’s disease. Free Radic. Biol. Med. 2023, 204, 385–393. [Google Scholar] [CrossRef] [PubMed]
  114. March-Diaz, R.; Lara-Urena, N.; Romero-Molina, C.; Heras-Garvin, A.; Ortega-de San Luis, C.; Alvarez-Vergara, M.I.; Sanchez-Garcia, M.A.; Sanchez-Mejias, E.; Davila, J.C.; Rosales-Nieves, A.E.; et al. Hypoxia compromises the mitochondrial metabolism of Alzheimer’s disease microglia via HIF1. Nat. Aging 2021, 1, 385–399. [Google Scholar] [CrossRef] [PubMed]
  115. Gedam, M.; Comerota, M.M.; Propson, N.E.; Chen, T.; Jin, F.; Wang, M.C.; Zheng, H. Complement C3aR depletion reverses HIF-1alpha-induced metabolic impairment and enhances microglial response to Abeta pathology. J. Clin. Investig. 2023, 133, e167501. [Google Scholar] [CrossRef]
  116. van Gijsel-Bonnello, M.; Baranger, K.; Benech, P.; Rivera, S.; Khrestchatisky, M.; de Reggi, M.; Gharib, B. Metabolic changes and inflammation in cultured astrocytes from the 5xFAD mouse model of Alzheimer’s disease: Alleviation by pantethine. PLoS ONE 2017, 12, e0175369. [Google Scholar] [CrossRef]
  117. Ashok, B.S.; Ajith, T.A.; Sivanesan, S. Hypoxia-inducible factors as neuroprotective agent in Alzheimer’s disease. Clin. Exp. Pharmacol. Physiol. 2017, 44, 327–334. [Google Scholar] [CrossRef]
  118. Pinky; Neha; Salman, M.; Kumar, P.; Khan, M.A.; Jamal, A.; Parvez, S. Age-related pathophysiological alterations in molecular stress markers and key modulators of hypoxia. Ageing Res. Rev. 2023, 90, 102022. [Google Scholar] [CrossRef]
  119. Mielke, R.; Herholz, K.; Grond, M.; Kessler, J.; Heiss, W.D. Differences of regional cerebral glucose metabolism between presenile and senile dementia of Alzheimer type. Neurobiol. Aging 1992, 13, 93–98. [Google Scholar] [CrossRef]
  120. Wan, W.; Peng, K.; Li, M.; Qin, L.; Tong, Z.; Yan, J.; Shen, B.; Yu, C. Histone demethylase JMJD1A promotes urinary bladder cancer progression by enhancing glycolysis through coactivation of hypoxia inducible factor 1alpha. Oncogene 2017, 36, 3868–3877. [Google Scholar] [CrossRef]
  121. Semenza, G.L. Regulation of mammalian O2 homeostasis by hypoxia-inducible factor 1. Annu. Rev. Cell Dev. Biol. 1999, 15, 551–578. [Google Scholar] [CrossRef] [PubMed]
  122. Soucek, T.; Cumming, R.; Dargusch, R.; Maher, P.; Schubert, D. The regulation of glucose metabolism by HIF-1 mediates a neuroprotective response to amyloid beta peptide. Neuron 2003, 39, 43–56. [Google Scholar] [CrossRef] [PubMed]
  123. Goyal, M.S.; Hawrylycz, M.; Miller, J.A.; Snyder, A.Z.; Raichle, M.E. Aerobic glycolysis in the human brain is associated with development and neotenous gene expression. Cell Metab. 2014, 19, 49–57. [Google Scholar] [CrossRef] [PubMed]
  124. Harris, R.A.; Tindale, L.; Cumming, R.C. Age-dependent metabolic dysregulation in cancer and Alzheimer’s disease. Biogerontology 2014, 15, 559–577. [Google Scholar] [CrossRef]
  125. Oransky, I. Oxygen sensor is neuroprotective in Alzheimer’s disease. Lancet Neurol. 2003, 2, 456. [Google Scholar] [CrossRef]
  126. Bos, I.; Vos, S.; Verhey, F.; Scheltens, P.; Teunissen, C.; Engelborghs, S.; Sleegers, K.; Frisoni, G.; Blin, O.; Richardson, J.C.; et al. Cerebrospinal fluid biomarkers of neurodegeneration, synaptic integrity, and astroglial activation across the clinical Alzheimer’s disease spectrum. Alzheimers Dement. 2019, 15, 644–654. [Google Scholar] [CrossRef]
  127. Salvadó, G.; Milà-Alomà, M.; Shekari, M.; Minguillon, C.; Fauria, K.; Niñerola-Baizán, A.; Perissinotti, A.; Kollmorgen, G.; Buckley, C.; Farrar, G.; et al. Cerebral amyloid-β load is associated with neurodegeneration and gliosis: Mediation by p-tau and interactions with risk factors early in the Alzheimer’s continuum. Alzheimers Dement. 2021, 17, 788–800. [Google Scholar] [CrossRef]
  128. Salvadó, G.; Milà-Alomà, M.; Shekari, M.; Ashton, N.J.; Operto, G.; Falcon, C.; Cacciaglia, R.; Minguillon, C.; Fauria, K.; Niñerola-Baizán, A.; et al. Reactive astrogliosis is associated with higher cerebral glucose consumption in the early Alzheimer’s continuum. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 4567–4579. [Google Scholar] [CrossRef]
  129. Schubert, D.; Soucek, T.; Blouw, B. The induction of HIF-1 reduces astrocyte activation by amyloid beta peptide. Eur. J. Neurosci. 2009, 29, 1323–1334. [Google Scholar] [CrossRef]
  130. Love, S.; Miners, J.S. Cerebral hypoperfusion and the energy deficit in Alzheimer’s disease. Brain Pathol. 2016, 26, 607–617. [Google Scholar] [CrossRef]
  131. Daulatzai, M.A. Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer’s disease. J. Neurosci. Res. 2017, 95, 943–972. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, H.; Ma, D.; Yue, F.; Qi, Y.; Dou, M.; Cui, L.; Xing, Y. The potential role of hypoxia-inducible factor-1 in the progression and therapy of central nervous system diseases. Curr. Neuropharmacol. 2022, 20, 1651–1666. [Google Scholar] [CrossRef] [PubMed]
  133. Iyalomhe, O.; Swierczek, S.; Enwerem, N.; Chen, Y.; Adedeji, M.O.; Allard, J.; Ntekim, O.; Johnson, S.; Hughes, K.; Kurian, P.; et al. The role of hypoxia-inducible factor 1 in mild cognitive impairment. Cell. Mol. Neurobiol. 2017, 37, 969–977. [Google Scholar] [CrossRef] [PubMed]
  134. Weinreb, O.; Amit, T.; Mandel, S.; Kupershmidt, L.; Youdim, M.B. Neuroprotective multifunctional iron chelators: From redox-sensitive process to novel therapeutic opportunities. Antioxid. Redox Signal. 2010, 13, 919–949. [Google Scholar] [CrossRef] [PubMed]
  135. Urena-Guerrero, M.E.; Castaneda-Cabral, J.L.; Rivera-Cervantes, M.C.; Macias-Velez, R.J.; Jarero-Basulto, J.J.; Gudino-Cabrera, G.; Beas-Zarate, C. Neuroprotective and neurorestorative effects of Epo and VEGF: Perspectives for new therapeutic approaches to neurological diseases. Curr. Pharm. Des. 2020, 26, 1263–1276. [Google Scholar] [CrossRef]
  136. Chong, Z.Z.; Li, F.; Maiese, K. Erythropoietin requires NF-kappaB and its nuclear translocation to prevent early and late apoptotic neuronal injury during beta-amyloid toxicity. Curr. Neurovasc. Res. 2005, 2, 387–399. [Google Scholar] [CrossRef]
  137. Wu, C.L.; Chen, S.D.; Yin, J.H.; Hwang, C.S.; Yang, D.I. Erythropoietin and sonic hedgehog mediate the neuroprotective effects of brain-derived neurotrophic factor against mitochondrial inhibition. Neurobiol. Dis. 2010, 40, 146–154. [Google Scholar] [CrossRef]
  138. Andrade, A.G.; Bubu, O.M.; Varga, A.W.; Osorio, R.S. The relationship between obstructive sleep apnea and Alzheimer’s disease. J. Alzheimers Dis. 2018, 64, S255–S270. [Google Scholar] [CrossRef]
  139. Bhuniya, S.; Goyal, M.; Chowdhury, N.; Mishra, P. Intermittent hypoxia and sleep disruption in obstructive sleep apnea increase serum tau and amyloid-beta levels. J. Sleep Res. 2022, 31, e13566. [Google Scholar] [CrossRef]
  140. Liguori, C.; Maestri, M.; Spanetta, M.; Placidi, F.; Bonanni, E.; Mercuri, N.B.; Guarnieri, B. Sleep-disordered breathing and the risk of Alzheimer’s disease. Sleep Med. Rev. 2021, 55, 101375. [Google Scholar] [CrossRef]
  141. Khuu, M.A.; Nallamothu, T.; Castro-Rivera, C.I.; Arias-Cavieres, A.; Szujewski, C.C.; Garcia Iii, A.J. Stage-dependent effects of intermittent hypoxia influence the outcome of hippocampal adult neurogenesis. Sci. Rep. 2021, 11, 6005. [Google Scholar] [CrossRef] [PubMed]
  142. Varela-Nallar, L.; Rojas-Abalos, M.; Abbott, A.C.; Moya, E.A.; Iturriaga, R.; Inestrosa, N.C. Chronic hypoxia induces the activation of the Wnt/beta-catenin signaling pathway and stimulates hippocampal neurogenesis in wild-type and APPswe-PS1DeltaE9 transgenic mice in vivo. Front. Cell. Neurosci. 2014, 8, 17. [Google Scholar] [CrossRef]
  143. Mazumdar, J.; O’Brien, W.T.; Johnson, R.S.; LaManna, J.C.; Chavez, J.C.; Klein, P.S.; Simon, M.C. O2 regulates stem cells through Wnt/beta-catenin signalling. Nat. Cell Biol. 2010, 12, 1007–1013. [Google Scholar] [CrossRef] [PubMed]
  144. Brand, K.A.; Hermfisse, U. Aerobic glycolysis by proliferating cells: A protective strategy against reactive oxygen species. FASEB J. 1997, 11, 388–395. [Google Scholar] [CrossRef]
  145. Butterfield, D.A.; Poon, H.F.; St Clair, D.; Keller, J.N.; Pierce, W.M.; Klein, J.B.; Markesbery, W.R. Redox proteomics identification of oxidatively modified hippocampal proteins in mild cognitive impairment: Insights into the development of Alzheimer’s disease. Neurobiol. Dis. 2006, 22, 223–232. [Google Scholar] [CrossRef]
  146. Desmond, D.W.; Moroney, J.T.; Sano, M.; Stern, Y. Incidence of dementia after ischemic stroke: Results of a longitudinal study. Stroke 2002, 33, 2254–2260. [Google Scholar] [CrossRef] [PubMed]
  147. He, J.B.; Zhang, H.; Zheng, H.X.; Jia, J.X.; Zhang, Y.C.; Yan, X.S.; Li, X.X.; Wei, K.W.; Mao, J.; Chen, H.; et al. Effects of schisandrin B on hypoxia-related cognitive function and protein expression in vascular dementia rats. J. Toxicol. Environ. Health A 2024, 87, 421–427. [Google Scholar] [CrossRef] [PubMed]
  148. Schubert, D.; Chevion, M. The role of iron in beta amyloid toxicity. Biochem. Biophys. Res. Commun. 1995, 216, 702–707. [Google Scholar] [CrossRef]
  149. Wang, T.; Xu, S.F.; Fan, Y.G.; Li, L.B.; Guo, C. Iron pathophysiology in Alzheimer’s diseases. Adv. Exp. Med. Biol. 2019, 1173, 67–104. [Google Scholar]
  150. Feng, L.; Sun, J.; Xia, L.; Shi, Q.; Hou, Y.; Zhang, L.; Li, M.; Fan, C.; Sun, B. Ferroptosis mechanism and Alzheimer’s disease. Neural Regen. Res. 2024, 19, 1741–1750. [Google Scholar] [CrossRef]
  151. Petralla, S.; Saveleva, L.; Kanninen, K.M.; Oster, J.S.; Panayotova, M.; Fricker, G.; Puris, E. Increased expression of transferrin receptor 1 in the brain cortex of 5xFAD mouse model of Alzheimer’s disease is associated with activation of HIF-1 signaling pathway. Mol. Neurobiol. 2024, 61, 6383–6394. [Google Scholar] [CrossRef] [PubMed]
  152. Schreiner, O.D.; Schreiner, T.G. Iron chelators as a therapeutic option for Alzheimer’s disease-A mini-review. Front. Aging 2023, 4, 1234958. [Google Scholar] [CrossRef] [PubMed]
  153. Avramovich-Tirosh, Y.; Bar-Am, O.; Amit, T.; Youdim, M.B.; Weinreb, O. Up-regulation of hypoxia-inducible factor (HIF)-1α and HIF-target genes in cortical neurons by the novel multifunctional iron chelator anti-Alzheimer drug, M30. Curr. Alzheimer Res. 2010, 7, 300–306. [Google Scholar] [CrossRef] [PubMed]
  154. Mechlovich, D.; Amit, T.; Bar-Am, O.; Mandel, S.; Youdim, M.B.; Weinreb, O. The novel multi-target iron chelator, M30 modulates HIF-1α-related glycolytic genes and insulin signaling pathway in the frontal cortex of APP/PS1 Alzheimer’s disease mice. Curr. Alzheimer Res. 2014, 11, 119–127. [Google Scholar] [CrossRef] [PubMed]
  155. Guo, C.; Zhang, Y.X.; Wang, T.; Zhong, M.L.; Yang, Z.H.; Hao, L.J.; Chai, R.; Zhang, S. Intranasal deferoxamine attenuates synapse loss via up-regulating the P38/HIF-1α pathway on the brain of APP/PS1 transgenic mice. Front. Aging Neurosci. 2015, 7, 104. [Google Scholar] [CrossRef]
  156. Fine, J.M.; Kosyakovsky, J.; Baillargeon, A.M.; Tokarev, J.V.; Cooner, J.M.; Svitak, A.L.; Faltesek, K.A.; Frey, W.H., 2nd; Hanson, L.R. Intranasal deferoxamine can improve memory in healthy C57 mice, suggesting a partially non-disease-specific pathway of functional neurologic improvement. Brain Behav. 2020, 10, e01536. [Google Scholar] [CrossRef]
  157. Yang, Y.T.; Ju, T.C.; Yang, D.I. Induction of hypoxia inducible factor-1 attenuates metabolic insults induced by 3-nitropropionic acid in rat C6 glioma cells. J. Neurochem. 2005, 93, 513–525. [Google Scholar] [CrossRef]
  158. Jiang, C.; Li, G.; Huang, P.; Liu, Z.; Zhao, B. The gut microbiota and Alzheimer’s disease. J. Alzheimers Dis. 2017, 58, 1–15. [Google Scholar] [CrossRef]
  159. Liu, S.; Gao, J.; Zhu, M.; Liu, K.; Zhang, H.L. Gut microbiota and dysbiosis in Alzheimer’s disease: Implications for pathogenesis and treatment. Mol. Neurobiol. 2020, 57, 5026–5043. [Google Scholar] [CrossRef]
  160. Janeiro, M.H.; Ramírez, M.J.; Solas, M. Dysbiosis and Alzheimer’s disease: Cause or treatment opportunity? Cell. Mol. Neurobiol. 2022, 42, 377–387. [Google Scholar] [CrossRef]
  161. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut-rain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
  162. Bonfili, L.; Gong, C.; Lombardi, F.; Cifone, M.G.; Eleuteri, A.M. Strategic modification of gut microbiota through oral bacteriotherapy influences hypoxia inducible factor-1α: Therapeutic implication in Alzheimer’s disease. Int. J. Mol. Sci. 2021, 23, 357. [Google Scholar] [CrossRef]
  163. Guo, C.; Yang, Z.H.; Zhang, S.; Chai, R.; Xue, H.; Zhang, Y.H.; Li, J.Y.; Wang, Z.Y. Intranasal lactoferrin enhances α-secretase-dependent amyloid precursor protein processing via the ERK1/2-CREB and HIF-1α pathways in an Alzheimer’s disease mouse model. Neuropsychopharmacology 2017, 42, 2504–2515. [Google Scholar] [CrossRef]
  164. Yu, Z.; Liu, N.; Liu, J.; Yang, K.; Wang, X. Neuroglobin, a novel target for endogenous neuroprotection against stroke and neurodegenerative disorders. Int. J. Mol. Sci. 2012, 13, 6995–7014. [Google Scholar] [CrossRef]
  165. Haines, B.; Demaria, M.; Mao, X.; Xie, L.; Campisi, J.; Jin, K.; Greenberg, D.A. Hypoxia-inducible factor-1 and neuroglobin expression. Neurosci. Lett. 2012, 514, 137–140. [Google Scholar] [CrossRef] [PubMed]
  166. Zhang, Y.H.; Yan, X.Z.; Xu, S.F.; Pang, Z.Q.; Li, L.B.; Yang, Y.; Fan, Y.G.; Wang, Z.; Yu, X.; Guo, C.; et al. α-Lipoic acid maintains brain glucose metabolism via BDNF/TrkB/HIF-1α signaling pathway in P301S mice. Front. Aging Neurosci. 2020, 12, 262. [Google Scholar] [CrossRef] [PubMed]
  167. Londzin, P.; Zamora, M.; Kąkol, B.; Taborek, A.; Folwarczna, J. Potential of caffeine in Alzheimer’s disease-a review of experimental studies. Nutrients 2021, 13, 537. [Google Scholar] [CrossRef]
  168. Zhu, Y.; Hu, C.X.; Liu, X.; Zhu, R.X.; Wang, B.Q. Moderate coffee or tea consumption decreased the risk of cognitive disorders: An updated dose-response meta-analysis. Nutr. Rev. 2023, 82, 738–748. [Google Scholar] [CrossRef]
  169. Kakio, S.; Funakoshi-Tago, M.; Kobata, K.; Tamura, H. Coffee induces vascular endothelial growth factor (VEGF) expression in human neuroblastama SH-SY5Y cells. Nutr. Neurosci. 2017, 20, 336–342. [Google Scholar] [CrossRef]
  170. Cruchaga, C.; Kauwe, J.S.; Mayo, K.; Spiegel, N.; Bertelsen, S.; Nowotny, P.; Shah, A.R.; Abraham, R.; Hollingworth, P.; Harold, D.; et al. SNPs associated with cerebrospinal fluid phospho-tau levels influence rate of decline in Alzheimer’s disease. PLoS Genet. 2010, 6, e1001101. [Google Scholar] [CrossRef]
  171. Zhou, Z.; Bai, J.; Zhong, S.; Zhang, R.; Kang, K.; Zhang, X.; Xu, Y.; Zhao, C.; Zhao, M. Integrative genomic analysis of PPP3R1 in Alzheimer’s disease: A potential biomarker for predictive, preventive, and personalized medical approach. EPMA J. 2021, 12, 647–658. [Google Scholar] [CrossRef]
  172. Chai, X.; Kong, W.; Liu, L.; Yu, W.; Zhang, Z.; Sun, Y. A viral vector expressing hypoxia-inducible factor 1 alpha inhibits hippocampal neuronal apoptosis. Neural Regen. Res. 2014, 9, 1145–1153. [Google Scholar] [PubMed]
  173. Brandon, J.A.; Farmer, B.C.; Williams, H.C.; Johnson, L.A. APOE and Alzheimer’s disease: Neuroimaging of metabolic and cerebrovascular dysfunction. Front. Aging Neurosci. 2018, 10, 180. [Google Scholar] [CrossRef] [PubMed]
  174. Salomon-Zimri, S.; Glat, M.J.; Barhum, Y.; Luz, I.; Boehm-Cagan, A.; Liraz, O.; Ben-Zur, T.; Offen, D.; Michaelson, D.M. Reversal of ApoE4-driven brain pathology by vascular endothelial growth factor treatment. J. Alzheimers Dis. 2016, 53, 1443–1458. [Google Scholar] [CrossRef] [PubMed]
  175. Wang, Y.Y.; Huang, Z.T.; Yuan, M.H.; Jing, F.; Cai, R.L.; Zou, Q.; Pu, Y.S.; Wang, S.Y.; Chen, F.; Yi, W.M.; et al. Role of hypoxia inducible factor-1α in Alzheimer’s disease. J. Alzheimers Dis. 2021, 80, 949–961. [Google Scholar] [CrossRef] [PubMed]
  176. Bulbarelli, A.; Lonati, E.; Brambilla, A.; Orlando, A.; Cazzaniga, E.; Piazza, F.; Ferrarese, C.; Masserini, M.; Sancini, G. Abeta42 production in brain capillary endothelial cells after oxygen and glucose deprivation. Mol. Cell. Neurosci. 2012, 49, 415–422. [Google Scholar] [CrossRef]
  177. Chen, G.J.; Xu, J.; Lahousse, S.A.; Caggiano, N.L.; de la Monte, S.M. Transient hypoxia causes Alzheimer-type molecular and biochemical abnormalities in cortical neurons: Potential strategies for neuroprotection. J. Alzheimers Dis. 2003, 5, 209–228. [Google Scholar] [CrossRef]
  178. Mattson, M.P. Cellular actions of beta-amyloid precursor protein and its soluble and fibrillogenic derivatives. Physiol. Rev. 1997, 77, 1081–1132. [Google Scholar] [CrossRef]
  179. Shi, J.; Yang, S.H.; Stubley, L.; Day, A.L.; Simpkins, J.W. Hypoperfusion induces overexpression of beta-amyloid precursor protein mRNA in a focal ischemic rodent model. Brain Res. 2000, 853, 1–4. [Google Scholar] [CrossRef]
  180. Scragg, J.L.; Fearon, I.M.; Boyle, J.P.; Ball, S.G.; Varadi, G.; Peers, C. Alzheimer’s amyloid peptides mediate hypoxic up-regulation of L-type Ca2+ channels. FASEB J. 2005, 19, 150–152. [Google Scholar] [CrossRef]
  181. Webster, N.J.; Ramsden, M.; Boyle, J.P.; Pearson, H.A.; Peers, C. Amyloid peptides mediate hypoxic increase of L-type Ca2+ channels in central neurones. Neurobiol. Aging 2006, 27, 439–445. [Google Scholar] [CrossRef] [PubMed]
  182. Zhang, X.; Zhou, K.; Wang, R.; Cui, J.; Lipton, S.A.; Liao, F.F.; Xu, H.; Zhang, Y.W. Hypoxia-inducible factor 1alpha (HIF-1alpha)-mediated hypoxia increases BACE1 expression and beta-amyloid generation. J. Biol. Chem. 2007, 282, 10873–10880. [Google Scholar] [CrossRef] [PubMed]
  183. Guglielmotto, M.; Aragno, M.; Autelli, R.; Giliberto, L.; Novo, E.; Colombatto, S.; Danni, O.; Parola, M.; Smith, M.A.; Perry, G.; et al. The up-regulation of BACE1 mediated by hypoxia and ischemic injury: Role of oxidative stress and HIF1alpha. J. Neurochem. 2009, 108, 1045–1056. [Google Scholar] [CrossRef] [PubMed]
  184. Webster, N.J.; Green, K.N.; Peers, C.; Vaughan, P.F. Altered processing of amyloid precursor protein in the human neuroblastoma SH-SY5Y by chronic hypoxia. J. Neurochem. 2002, 83, 1262–1271. [Google Scholar] [CrossRef] [PubMed]
  185. Marshall, A.J.; Rattray, M.; Vaughan, P.F. Chronic hypoxia in the human neuroblastoma SH-SY5Y causes reduced expression of the putative alpha-secretases, ADAM10 and TACE, without altering their mRNA levels. Brain Res. 2006, 1099, 18–24. [Google Scholar] [CrossRef]
  186. Sun, Y.; Islam, S.; Michikawa, M.; Zou, K. Presenilin: A multi-functional molecule in the pathogenesis of Alzheimer’s disease and other neurodegenerative diseases. Int. J. Mol. Sci. 2024, 25, 1757. [Google Scholar] [CrossRef]
  187. Alexander, C.; Li, T.; Hattori, Y.; Chiu, D.; Frost, G.R.; Jonas, L.; Liu, C.; Anderson, C.J.; Wong, E.; Park, L.; et al. Hypoxia Inducible Factor-1alpha binds and activates gamma-secretase for Abeta production under hypoxia and cerebral hypoperfusion. Mol. Psychiatry 2022, 27, 4264–4273. [Google Scholar] [CrossRef]
  188. Wang, R.; Zhang, Y.W.; Zhang, X.; Liu, R.; Zhang, X.; Hong, S.; Xia, K.; Xia, J.; Zhang, Z.; Xu, H. Transcriptional regulation of APH-1A and increased gamma-secretase cleavage of APP and Notch by HIF-1 and hypoxia. FASEB J. 2006, 20, 1275–1277. [Google Scholar] [CrossRef]
  189. Kaufmann, M.R.; Barth, S.; Konietzko, U.; Wu, B.; Egger, S.; Kunze, R.; Marti, H.H.; Hick, M.; Muller, U.; Camenisch, G.; et al. Dysregulation of hypoxia-inducible factor by presenilin/gamma-secretase loss-of-function mutations. J. Neurosci. 2013, 33, 1915–1926. [Google Scholar] [CrossRef]
  190. De Gasperi, R.; Sosa, M.A.; Dracheva, S.; Elder, G.A. Presenilin-1 regulates induction of hypoxia inducible factor-1alpha: Altered activation by a mutation associated with familial Alzheimer’s disease. Mol. Neurodegener. 2010, 5, 38. [Google Scholar] [CrossRef]
  191. Iwata, N.; Tsubuki, S.; Takaki, Y.; Watanabe, K.; Sekiguchi, M.; Hosoki, E.; Kawashima-Morishima, M.; Lee, H.J.; Hama, E.; Sekine-Aizawa, Y.; et al. Identification of the major Abeta1-42-degrading catabolic pathway in brain parenchyma: Suppression leads to biochemical and pathological deposition. Nat. Med. 2000, 6, 143–150. [Google Scholar] [CrossRef] [PubMed]
  192. Mitra, R.; Chao, O.S.; Nanus, D.M.; Goodman, O.B., Jr. Negative regulation of NEP expression by hypoxia. Prostate 2013, 73, 706–714. [Google Scholar] [CrossRef] [PubMed]
  193. Wang, H.; Sun, M.; Yang, H.; Tian, X.; Tong, Y.; Zhou, T.; Zhang, T.; Fu, Y.; Guo, X.; Fan, D.; et al. Hypoxia-inducible factor-1alpha mediates up-regulation of neprilysin by histone deacetylase-1 under hypoxia condition in neuroblastoma cells. J. Neurochem. 2014, 131, 4–11. [Google Scholar] [CrossRef] [PubMed]
  194. Khamaisi, M.; Toukan, H.; Axelrod, J.H.; Rosenberger, C.; Skarzinski, G.; Shina, A.; Meidan, R.; Koesters, R.; Rosen, S.; Walkinshaw, G.; et al. Endothelin-converting enzyme is a plausible target gene for hypoxia-inducible factor. Kidney Int. 2015, 87, 761–770. [Google Scholar] [CrossRef]
  195. Minchenko, D.O.; Khita, O.O.; Tsymbal, D.O.; Danilovskyi, S.V.; Rudnytska, O.V.; Halkin, O.V.; Kryvdiuk, I.V.; Smeshkova, M.V.; Yakymchuk, M.M.; Bezrodnyi, B.H.; et al. Expression of IDE and PITRM1 genes in ERN1 knockdown U87 glioma cells: Effect of hypoxia and glucose deprivation. Endocr. Regul. 2020, 54, 183–195. [Google Scholar] [CrossRef]
  196. Zhao, Y.; Valis, M.; Wang, X.; Nepovimova, E.; Wu, Q.; Kuca, K. HIF-1alpha is a “brake” in JNK-mediated activation of amyloid protein precursor and hyperphosphorylation of tau induced by T-2 toxin in BV2 cells. Mycotoxin Res. 2024, 40, 223–234. [Google Scholar] [CrossRef]
  197. Lei, L.; Feng, J.; Wu, G.; Wei, Z.; Wang, J.Z.; Zhang, B.; Liu, R.; Liu, F.; Wang, X.; Li, H.L. HIF-1alpha causes LCMT1/PP2A deficiency and mediates tau hyperphosphorylation and cognitive dysfunction during chronic hypoxia. Int. J. Mol. Sci. 2022, 23, 16140. [Google Scholar] [CrossRef]
  198. Tang, J.; Li, Y.; Liu, X.; Yu, G.; Zheng, F.; Guo, Z.; Zhang, Y.; Shao, W.; Wu, S.; Li, H. Cobalt induces neurodegenerative damages through impairing autophagic flux by activating hypoxia-inducible factor-1alpha triggered ROS overproduction. Sci. Total Environ. 2023, 857, 159432. [Google Scholar] [CrossRef]
  199. Fine, J.M.; Baillargeon, A.M.; Renner, D.B.; Hoerster, N.S.; Tokarev, J.; Colton, S.; Pelleg, A.; Andrews, A.; Sparley, K.A.; Krogh, K.M.; et al. Intranasal deferoxamine improves performance in radial arm water maze, stabilizes HIF-1alpha, and phosphorylates GSK3beta in P301L tau transgenic mice. Exp. Brain Res. 2012, 219, 381–390. [Google Scholar] [CrossRef]
  200. Grubman, A.; Choo, X.Y.; Chew, G.; Ouyang, J.F.; Sun, G.; Croft, N.P.; Rossello, F.J.; Simmons, R.; Buckberry, S.; Landin, D.V.; et al. Transcriptional signature in microglia associated with Aβ plaque phagocytosis. Nat. Commun. 2021, 12, 3015. [Google Scholar] [CrossRef]
  201. Yang, Z.; Zhao, T.Z.; Zou, Y.J.; Zhang, J.H.; Feng, H. Hypoxia induces autophagic cell death through hypoxia-inducible factor 1alpha in microglia. PLoS ONE 2014, 9, e96509. [Google Scholar]
  202. Baik, S.H.; Kang, S.; Lee, W.; Choi, H.; Chung, S.; Kim, J.I.; Mook-Jung, I. A breakdown in metabolic reprogramming causes microglia dysfunction in Alzheimer’s disease. Cell Metab. 2019, 30, 493–507. [Google Scholar] [CrossRef] [PubMed]
  203. Ashok, A.; Rai, N.K.; Raza, W.; Pandey, R.; Bandyopadhyay, S. Chronic cerebral hypoperfusion-induced impairment of Aβ clearance requires HB-EGF-dependent sequential activation of HIF1α and MMP9. Neurobiol. Dis. 2016, 95, 179–193. [Google Scholar] [CrossRef] [PubMed]
  204. Salminen, A.; Kauppinen, A.; Kaarniranta, K. Hypoxia/ischemia activate processing of Amyloid Precursor Protein: Impact of vascular dysfunction in the pathogenesis of Alzheimer’s disease. J. Neurochem. 2017, 140, 536–549. [Google Scholar] [CrossRef] [PubMed]
  205. Bonda, D.J.; Bajić, V.P.; Spremo-Potparevic, B.; Casadesus, G.; Zhu, X.; Smith, M.A.; Lee, H.G. Review: Cell cycle aberrations and neurodegeneration. Neuropathol. Appl. Neurobiol. 2010, 36, 157–163. [Google Scholar] [CrossRef]
  206. Chen, S.D.; Yang, J.L.; Lin, Y.C.; Chao, A.C.; Yang, D.I. Emerging roles of inhibitor of differentiation-1 in Alzheimer’s disease: Cell cycle reentry and beyond. Cells 2020, 9, 1746. [Google Scholar] [CrossRef]
  207. Requejo-Aguilar, R. Cdk5 and aberrant cell cycle activation at the core of neurodegeneration. Neural Regen. Res. 2023, 18, 1186–1190. [Google Scholar] [CrossRef]
  208. Hung, Y.H.; Chang, S.H.; Huang, C.T.; Yin, J.H.; Hwang, C.S.; Yang, L.Y.; Yang, D.I. Inhibitor of differentiation-1 and hypoxia-inducible factor-1 mediate sonic hedgehog induction by amyloid beta-peptide in rat cortical neurons. Mol. Neurobiol. 2016, 53, 793–809. [Google Scholar] [CrossRef]
  209. Wu, M.H.; Chao, A.C.; Hsieh, Y.H.; Lien, Y.; Lin, Y.C.; Yang, D.I. Protein kinase C-delta mediates cell cycle reentry and apoptosis induced by amyloid-beta peptide in post-mitotic cortical neurons. Int. J. Mol. Sci. 2024, 25, 9626. [Google Scholar] [CrossRef]
  210. Hou, B.Y.; Wu, M.H.; Hsu, H.Y.; Lin, Y.C.; Yang, D.I. Polysaccharides from Basella alba protect post-mitotic neurons against cell cycle re-entry and apoptosis induced by the amyloid-beta peptide by blocking sonic hedgehog expression. Int. J. Mol. Sci. 2024, 25, 7316. [Google Scholar] [CrossRef]
  211. Chen, S.; An, F.M.; Yin, L.; Liu, A.R.; Yin, D.K.; Yao, W.B.; Gao, X.D. Glucagon-like peptide-1 protects hippocampal neurons against advanced glycation end product-induced tau hyperphosphorylation. Neuroscience 2014, 256, 137–146. [Google Scholar] [CrossRef] [PubMed]
  212. Chen, S.D.; Chuang, Y.C.; Lin, T.K.; Yang, J.L. Alternative role of glucagon-like Peptide-1 receptor agonists in neurodegenerative diseases. Eur. J. Pharmacol. 2023, 938, 175439. [Google Scholar] [CrossRef] [PubMed]
  213. Lin, K.J.; Wang, T.J.; Chen, S.D.; Lin, K.L.; Liou, C.W.; Lan, M.Y.; Chuang, Y.C.; Chuang, J.H.; Wang, P.W.; Lee, J.J.; et al. Two birds one stone: The neuroprotective effect of antidiabetic agents on Parkinson disease-focus on sodium-glucose cotransporter 2 (SGLT2) inhibitors. Antioxidants 2021, 10, 1935. [Google Scholar] [CrossRef] [PubMed]
  214. Itoh, N. Hormone-like (endocrine) Fgfs: Their evolutionary history and roles in development, metabolism, and disease. Cell Tissue Res. 2010, 342, 1–11. [Google Scholar] [CrossRef]
  215. Spann, R.A.; Morrison, C.D.; den Hartigh, L.J. The nuanced metabolic functions of endogenous FGF21 depend on the nature of the stimulus, tissue source, and experimental model. Front. Endocrinol. 2021, 12, 802541. [Google Scholar] [CrossRef]
  216. Chen, S.; Chen, S.T.; Sun, Y.; Xu, Z.; Wang, Y.; Yao, S.Y.; Yao, W.B.; Gao, X.D. Fibroblast growth factor 21 ameliorates neurodegeneration in rat and cellular models of Alzheimer’s disease. Redox Biol. 2019, 22, 101133. [Google Scholar] [CrossRef]
  217. Sa-Nguanmoo, P.; Chattipakorn, N.; Chattipakorn, S.C. Potential roles of fibroblast growth factor 21 in the brain. Metab. Brain Dis. 2016, 31, 239–248. [Google Scholar] [CrossRef]
  218. Jin, K.; Mao, X.O.; Sun, Y.; Xie, L.; Jin, L.; Nishi, E.; Klagsbrun, M.; Greenberg, D.A. Heparin-binding epidermal growth factor-like growth factor: Hypoxia-inducible expression in vitro and stimulation of neurogenesis in vitro and in vivo. J. Neurosci. 2002, 22, 5365–5373. [Google Scholar] [CrossRef] [PubMed]
  219. Agarwal, A.; Zhang, M.; Trembak-Duff, I.; Unterbarnscheidt, T.; Radyushkin, K.; Dibaj, P.; Martins de Souza, D.; Boretius, S.; Brzózka, M.M.; Steffens, H.; et al. Dysregulated expression of neuregulin-1 by cortical pyramidal neurons disrupts synaptic plasticity. Cell Rep. 2014, 8, 1130–1145. [Google Scholar] [CrossRef]
  220. Orr-Urtreger, A.; Trakhtenbrot, L.; Ben-Levy, R.; Wen, D.; Rechavi, G.; Lonai, P.; Yarden, Y. Neural expression and chromosomal mapping of Neu differentiation factor to 8p12-p21. Proc. Natl. Acad. Sci. USA 1993, 90, 1867–1871. [Google Scholar] [CrossRef]
  221. Yoo, S.Y.; Yoo, J.Y.; Kim, H.B.; Baik, T.K.; Lee, J.H.; Woo, R.S. Neuregulin-1 Protects Neuronal Cells Against Damage due to CoCl2-Induced Hypoxia by Suppressing Hypoxia-Inducible Factor-1α and P53 in SH-SY5Y Cells. Int. Neurourol. J. 2019, 23, S111–S118. [Google Scholar] [CrossRef] [PubMed]
  222. Lee, H.J.; Ryu, J.M.; Jung, Y.H.; Lee, S.J.; Kim, J.Y.; Lee, S.H.; Hwang, I.K.; Seong, J.K.; Han, H.J. High glucose upregulates BACE1-mediated Aβ production through ROS-dependent HIF-1α and LXRα/ABCA1-regulated lipid raft reorganization in SK-N-MC cells. Sci. Rep. 2016, 6, 36746. [Google Scholar] [CrossRef] [PubMed]
  223. Valle, M.L.; Anderson, Y.T.; Grimsey, N.; Zastre, J. Thiamine insufficiency induces Hypoxia Inducible Factor-1alpha as an upstream mediator for neurotoxicity and AD-like pathology. Mol. Cell. Neurosci. 2022, 123, 103785. [Google Scholar] [CrossRef] [PubMed]
  224. Li, X.; Huang, L.; Kong, L.; Su, Y.; Zhou, H.; Ji, P.; Sun, R.; Wang, C.; Li, W.; Li, W. Ginsenoside Rg1 alleviates learning and memory impairments and Aβ disposition through inhibiting NLRP1 inflammasome and autophagy dysfunction in APP/PS1 mice. Mol. Med. Rep. 2023, 27, 6. [Google Scholar] [CrossRef]
  225. Wang, N.; Yang, J.; Chen, R.; Liu, Y.; Liu, S.; Pan, Y.; Lei, Q.; Wang, Y.; He, L.; Song, Y.; et al. Ginsenoside Rg1 ameliorates Alzheimer’s disease pathology via restoring mitophagy. J. Ginseng Res. 2023, 47, 448–457. [Google Scholar] [CrossRef]
  226. Yan, J.; Liu, Q.; Dou, Y.; Hsieh, Y.; Liu, Y.; Tao, R.; Zhu, D.; Lou, Y. Activating glucocorticoid receptor-ERK signaling pathway contributes to ginsenoside Rg1 protection against β-amyloid peptide-induced human endothelial cells apoptosis. J. Ethnopharmacol. 2013, 147, 456–466. [Google Scholar] [CrossRef]
  227. Li, Q.Y.; Wang, H.M.; Wang, Z.Q.; Ma, J.F.; Ding, J.Q.; Chen, S.D. Salidroside attenuates hypoxia-induced abnormal processing of amyloid precursor protein by decreasing BACE1 expression in SH-SY5Y cells. Neurosci. Lett. 2010, 481, 154–158. [Google Scholar] [CrossRef]
  228. Granholm, A.C.; Bimonte-Nelson, H.A.; Moore, A.B.; Nelson, M.E.; Freeman, L.R.; Sambamurti, K. Effects of a saturated fat and high cholesterol diet on memory and hippocampal morphology in the middle-aged rat. J. Alzheimers Dis. 2008, 14, 133–145. [Google Scholar] [CrossRef]
  229. Liu, L.; Martin, R.; Chan, C. Palmitate-activated astrocytes via serine palmitoyltransferase increase BACE1 in primary neurons by sphingomyelinases. Neurobiol. Aging 2013, 34, 540–550. [Google Scholar] [CrossRef]
  230. Patil, S.; Sheng, L.; Masserang, A.; Chan, C. Palmitic acid-treated astrocytes induce BACE1 upregulation and accumulation of C-terminal fragment of APP in primary cortical neurons. Neurosci. Lett. 2006, 406, 55–59. [Google Scholar] [CrossRef]
  231. Kim, J.Y.; Lee, H.J.; Lee, S.J.; Jung, Y.H.; Yoo, D.Y.; Hwang, I.K.; Seong, J.K.; Ryu, J.M.; Han, H.J. Palmitic Acid-BSA enhances Amyloid-β production through GPR40-mediated dual pathways in neuronal cells: Involvement of the Akt/mTOR/HIF-1α and Akt/NF-κB pathways. Sci. Rep. 2017, 7, 4335. [Google Scholar] [CrossRef] [PubMed]
  232. Fukuda, Y.; Berry, T.L.; Nelson, M.; Hunter, C.L.; Fukuhara, K.; Imai, H.; Ito, S.; Granholm-Bentley, A.C.; Kaplan, A.P.; Mutoh, T. Stimulated neuronal expression of brain-derived neurotrophic factor by Neurotropin. Mol. Cell. Neurosci. 2010, 45, 226–233. [Google Scholar] [CrossRef] [PubMed]
  233. Fang, W.L.; Zhao, D.Q.; Wang, F.; Li, M.; Fan, S.N.; Liao, W.; Zheng, Y.Q.; Liao, S.W.; Xiao, S.H.; Luan, P.; et al. Neurotropin® alleviates hippocampal neuron damage through a HIF-1α/MAPK pathway. CNS Neurosci. Ther. 2017, 23, 428–437. [Google Scholar] [CrossRef] [PubMed]
  234. Onyango, I.G.; Bennett, J.P., Jr.; Tuttle, J.B. Endogenous oxidative stress in sporadic Alzheimer’s disease neuronal cybrids reduces viability by increasing apoptosis through pro-death signaling pathways and is mimicked by oxidant exposure of control cybrids. Neurobiol. Dis. 2005, 19, 312–322. [Google Scholar] [CrossRef] [PubMed]
  235. Onyango, I.G.; Tuttle, J.B.; Bennett, J.P., Jr. Altered intracellular signaling and reduced viability of Alzheimer’s disease neuronal cybrids is reproduced by beta-amyloid peptide acting through receptor for advanced glycation end products (RAGE). Mol. Cell. Neurosci. 2005, 29, 333–343. [Google Scholar] [CrossRef]
  236. Jeong, J.H.; Yum, K.S.; Chang, J.Y.; Kim, M.; Ahn, J.Y.; Kim, S.; Lapchak, P.A.; Han, M.K. Dose-specific effect of simvastatin on hypoxia-induced HIF-1α and BACE expression in Alzheimer’s disease cybrid cells. BMC Neurol. 2015, 15, 127. [Google Scholar] [CrossRef]
  237. Singrang, N.; Nopparat, C.; Panmanee, J.; Govitrapong, P. Melatonin inhibits hypoxia-induced Alzheimer’s disease pathogenesis by regulating the amyloidogenic pathway in human neuroblastoma cells. Int. J. Mol. Sci. 2024, 25, 5225. [Google Scholar] [CrossRef]
Figure 1. The major pathologies in AD brains include deposition of extracellular Aβ plaques and intraneuronal neurofibrillary tangles (NFTs) mainly composed of hyperphosphorylated tau proteins. Excessive Aβ aggregation can trigger diverse mechanisms including excitotoxicity, oxidative stress with heightened ROS levels, mitochondrial dysfunction with compromised ATP production, aberrant cell cycle reentry with subsequent apoptosis, and activation of neurotoxic glial cells like microglia to trigger neuroinflammation; these effects together lead to the damage or even demise of the neurons. Tau belongs to the microtubule-associated protein (MAP) family that is vital for microtubule assembly and stabilization in neuronal axons. Hyperphosphorylated tau proteins not only compromise microtubule structures to disturb axonal transport but also aberrantly aggregate into NFTs, which also contribute to neuroinflammation and neuronal apoptosis. Excessive neuronal death ultimately results in brain atrophy in AD patients.
Figure 1. The major pathologies in AD brains include deposition of extracellular Aβ plaques and intraneuronal neurofibrillary tangles (NFTs) mainly composed of hyperphosphorylated tau proteins. Excessive Aβ aggregation can trigger diverse mechanisms including excitotoxicity, oxidative stress with heightened ROS levels, mitochondrial dysfunction with compromised ATP production, aberrant cell cycle reentry with subsequent apoptosis, and activation of neurotoxic glial cells like microglia to trigger neuroinflammation; these effects together lead to the damage or even demise of the neurons. Tau belongs to the microtubule-associated protein (MAP) family that is vital for microtubule assembly and stabilization in neuronal axons. Hyperphosphorylated tau proteins not only compromise microtubule structures to disturb axonal transport but also aberrantly aggregate into NFTs, which also contribute to neuroinflammation and neuronal apoptosis. Excessive neuronal death ultimately results in brain atrophy in AD patients.
Antioxidants 13 01378 g001
Figure 2. Activation of HIF-1 and its biological functions. HIF-1 is a heterodimeric transcription factor consisting of an oxygen-sensitive alpha subunit (HIF-1α) and a constitutively expressed beta subunit (HIF-1β). Under normoxia, HIF-1α undergoes hydroxylation of the proline residues catalyzed by the prolyl hydroxylase (PHD), which requires molecular oxygen (O2). The hydroxylated HIF-1α is then recognized by the von Hippel–Lindau (VHL) protein and E3 ubiquitin ligase for ubiquitination and subsequent proteasomal degradation. Under hypoxia, low-oxygen tension interferes with PHD hydroxylation and disrupts the interaction between HIF-1α and VHL, thereby stabilizing HIF-1α for its accumulation to form the heterodimeric HIF-1α/β. Translocation of the HIF-1α/β complex into the nucleus, along with coactivators p300/CBP, then drive the expression of target genes containing the hypoxia-response element (HRE) sequences in their promoters. HIF-1-dependent gene expression is crucial for numerous cellular responses to adapt the tissues to hypoxic environments, such as promoting angiogenesis and regulating vascular tone, enhancing antioxidation, regulating glucose transport and reprogramming energy metabolisms, affecting apoptosis, and regulating immune responses.
Figure 2. Activation of HIF-1 and its biological functions. HIF-1 is a heterodimeric transcription factor consisting of an oxygen-sensitive alpha subunit (HIF-1α) and a constitutively expressed beta subunit (HIF-1β). Under normoxia, HIF-1α undergoes hydroxylation of the proline residues catalyzed by the prolyl hydroxylase (PHD), which requires molecular oxygen (O2). The hydroxylated HIF-1α is then recognized by the von Hippel–Lindau (VHL) protein and E3 ubiquitin ligase for ubiquitination and subsequent proteasomal degradation. Under hypoxia, low-oxygen tension interferes with PHD hydroxylation and disrupts the interaction between HIF-1α and VHL, thereby stabilizing HIF-1α for its accumulation to form the heterodimeric HIF-1α/β. Translocation of the HIF-1α/β complex into the nucleus, along with coactivators p300/CBP, then drive the expression of target genes containing the hypoxia-response element (HRE) sequences in their promoters. HIF-1-dependent gene expression is crucial for numerous cellular responses to adapt the tissues to hypoxic environments, such as promoting angiogenesis and regulating vascular tone, enhancing antioxidation, regulating glucose transport and reprogramming energy metabolisms, affecting apoptosis, and regulating immune responses.
Antioxidants 13 01378 g002
Figure 3. Multiple AD-related mechanisms, including cerebral hypoperfusion, oxidative stress, and neuroinflammation, may trigger activation of HIF-1 to exert either positive or negative impacts on AD progression. The beneficial effects triggered by HIF-1 include affecting energy metabolisms, promoting neuroprotection/neurorestoration, enhancing neurogenesis, and counteracting oxidative stress, together allowing the tissues to adapt to the hypoxic environment. The detrimental effects include enhancing BACE1 expression with heightened β-secretase activity to promote Aβ production, impairing brain microvascular functions, and triggering neuronal cell cycle reentry followed by apoptosis. Notably, several unclear effects of HIF-1 in AD deserve detailed investigation. These include modulating brain circulation/angiogenesis, regulating tau hyperphosphorylation, affecting microglial functions and neuroinflammation, controlling the activities of α-secretase, γ-secretase, PS1/2 functions, and even Aβ degradation.
Figure 3. Multiple AD-related mechanisms, including cerebral hypoperfusion, oxidative stress, and neuroinflammation, may trigger activation of HIF-1 to exert either positive or negative impacts on AD progression. The beneficial effects triggered by HIF-1 include affecting energy metabolisms, promoting neuroprotection/neurorestoration, enhancing neurogenesis, and counteracting oxidative stress, together allowing the tissues to adapt to the hypoxic environment. The detrimental effects include enhancing BACE1 expression with heightened β-secretase activity to promote Aβ production, impairing brain microvascular functions, and triggering neuronal cell cycle reentry followed by apoptosis. Notably, several unclear effects of HIF-1 in AD deserve detailed investigation. These include modulating brain circulation/angiogenesis, regulating tau hyperphosphorylation, affecting microglial functions and neuroinflammation, controlling the activities of α-secretase, γ-secretase, PS1/2 functions, and even Aβ degradation.
Antioxidants 13 01378 g003
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

Lin, T.-K.; Huang, C.-R.; Lin, K.-J.; Hsieh, Y.-H.; Chen, S.-D.; Lin, Y.-C.; Chao, A.-C.; Yang, D.-I. Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer’s Disease: Beneficial or Detrimental? Antioxidants 2024, 13, 1378. https://doi.org/10.3390/antiox13111378

AMA Style

Lin T-K, Huang C-R, Lin K-J, Hsieh Y-H, Chen S-D, Lin Y-C, Chao A-C, Yang D-I. Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer’s Disease: Beneficial or Detrimental? Antioxidants. 2024; 13(11):1378. https://doi.org/10.3390/antiox13111378

Chicago/Turabian Style

Lin, Tsu-Kung, Chi-Ren Huang, Kai-Jung Lin, Yi-Heng Hsieh, Shang-Der Chen, Yi-Chun Lin, A-Ching Chao, and Ding-I Yang. 2024. "Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer’s Disease: Beneficial or Detrimental?" Antioxidants 13, no. 11: 1378. https://doi.org/10.3390/antiox13111378

APA Style

Lin, T. -K., Huang, C. -R., Lin, K. -J., Hsieh, Y. -H., Chen, S. -D., Lin, Y. -C., Chao, A. -C., & Yang, D. -I. (2024). Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer’s Disease: Beneficial or Detrimental? Antioxidants, 13(11), 1378. https://doi.org/10.3390/antiox13111378

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