Abstract
The endometrium is a multicellular tissue that is exquisitely responsive to the ovarian hormones. The local mechanisms of endometrial regulation to ensure optimal function are less well characterised. Transient physiological hypoxia has been proposed as a critical regulator of endometrial function. Herein, we review the literature on hypoxia in the non-pregnant endometrium. We discuss the pros and cons of animal models, human laboratory studies and novel in vivo imaging for the study of endometrial hypoxia. These research tools provide mounting evidence of a transient hypoxic episode in the menstrual endometrium and suggest that endometrial hypoxia may be present at the time of implantation. This local hypoxia may modify the inflammatory environment, influence vascular remodelling and modulate endometrial proliferation to optimise endometrial function. Finally, we review current knowledge of the impact of this hypoxia on endometrial pathologies, with a focus on abnormal uterine bleeding. Throughout the manuscript areas for future research are highlighted with the aim of concentrating research efforts to maximise future benefits for women and society.
Introduction
The human endometrium is a heterogeneous and dynamic tissue that undergoes cyclical breakdown and repair/regeneration more than 400 times during the female reproductive lifespan (Short 1976, Critchley et al. 2020). This occurs each month without scarring or loss of function. However, the regulation and local mechanisms of this endometrial breakdown and repair remain elusive. In particular, our knowledge of the contribution of local endometrial hypoxia to this process is in its infancy. The presence of hypoxia, usually defined as a partial oxygen pressure below 10 mmHg, is not an uncommon phenomenon in human physiology, for example, in bone marrow and intestinal mucosa (Suda et al. 2011, Zheng et al. 2015). Its presence in the menstrual endometrium has been proposed following progesterone withdrawal and intense vasoconstriction of the specialised spiral arterioles (Markee 1940). Unravelling the role of hypoxia in the endometrium has the potential to improve our understanding of menstrual and implantation disorders and reveal novel therapeutic strategies for those suffering from these common, devastating conditions.
Endometrial histology and ovarian hormone regulation
Histologically, the endometrium can be divided into the functional and basal layer (Noyes et al. 1950). The functional layer occupies the upper two-thirds of the endometrium and is composed of stroma and glands. This layer undergoes constant remodelling throughout the menstrual cycle and is shed during menstruation. The basal layer, adjacent to the myometrium, comprises the lower third of the endometrium.
Oestradiol is the dominant hormone in the first half of the menstrual cycle, during the proliferative phase. It acts via the oestrogen receptor (ER), which has two structurally related subtypes, ERα and ERβ (Lessey et al. 1988, Critchley et al. 2002). After ovulation, levels of oestradiol decline and the corpus luteum increases its progesterone production, prompting endometrial differentiation and decidualisation. This process, driven by cAMP signalling, reshapes the stromal compartment in order to keep the endometrium receptive for future implantation (Dunn et al. 2003). In contrast with non-menstruating species, where implantation of an embryo is required to trigger decidualisation (Brasted et al. 2003), the human endometrium spontaneously decidualises with endometrial stromal cells in close proximity to spiral arterioles initiating their own transformation (Gellersen & Brosens 2014). They morphologically transition from fibroblast-like cells to rounded epithelioid-like cells (Dunn et al. 2003).
Endometrial breakdown and regeneration
In the absence of implantation, the corpus luteum regresses, causing significant progesterone withdrawal (Corker et al. 1976, Maybin et al. 2011b). This decrease in progesterone levels triggers a cascade of local physiological inflammatory events that initiate menstruation.
Progesterone withdrawal leads to the induction of the transcription factor NFκB, which upregulates the expression of pro-inflammatory cytokines (IL6, TNF) and chemokines (CCL2, CXCL8) (King et al. 2001). In addition, this fall in progesterone levels increases endometrial cyclooxygenase 2 (COX-2), responsible for the synthesis of prostaglandins (PG) (Critchley et al. 1999). Increased levels of these inflammatory mediators drive the recruitment of myeloid leukocytes, activation of matrix metalloproteinases (MMPs) and the shedding of the upper endometrial layers (Critchley et al. 2001, Kelly et al. 2001). Hypoxia has been identified in the endometrium following progesterone withdrawal (Fan et al. 2008, Cousins et al. 2016b, Maybin et al. 2018) and may be due to vasoconstriction of the endometrial vessels. PGF2α and endothelin-1 (EDN1) are two endometrial factors with known vasoconstrictive properties that are present following progesterone withdrawal (Baird et al. 1996, Marsh et al. 1997). Vasoconstriction of specialised endometrial spiral arterioles may limit blood loss during menstruation. The subsequent tissue hypoxia does not appear to be necessary for endometrial breakdown but may have an important role in endometrial repair/regeneration (Maybin et al. 2018, Chen et al. 2020).
Shedding of the functional endometrial layer necessitates repair of the denuded endometrial surface and regeneration of endometrial tissue. This takes place when oestradiol and progesterone levels are low but local glucocorticoid action may be increased (McDonald et al. 2006, Kaitu’u-Lino et al. 2007a, Rae et al. 2009). Evidence from mouse models and human tissue studies suggest that hypoxia is required for physiological endometrial repair (Fan et al. 2008, Maybin et al. 2018). The processes involved are likely to be similar to those of wound healing, involving haemostasis, inflammation, proliferation and remodelling (Velnar et al. 2009, Mutsaers et al. 2015).
Detection of Hypoxia throughout the menstrual cycle
The first suggestion that hypoxia was present at menses derived from findings in a primate model in 1940 (Markee 1940). Transplantation of endometrial explants to the Rhesus macaque eye allowed direct observation of intense vasoconstriction of spiral arterioles and focal bleeding following progesterone withdrawal. Since then, the use and refinement of animal models for the study of menstrual physiology and endometrial hypoxia has become more common.
In vivo animal models
Menstruation is restricted to humans and few other species. These include higher order primates (baboons, Rhesus macaques), the elephant shrew (Van der Horst & Gillman 1940), certain bats (Hamlett 1934, Rasweiler & de Bonilla 1992, Zhang et al. 2007) and the spiny mouse (Bellofiore et al. 2017). The majority of menstrual studies have been carried out in rodents and non-human primates, including the Rhesus macaque (Brenner & Slayden 2012).
Rodent models
Despite physiological differences between mice and humans (e.g. a shorter length of cycle and lack of spontaneous decidualisation) mouse models replicate the events of human menstruation and decidualisation well (Wang et al. 2013, Cousins et al. 2016a, Armstrong et al. 2017). The feasible management of large experimental groups, short breeding times and availability of laboratory antibodies/reagents provide advantages over macaque models. Mouse models also offer the possibility of genetic, environmental and pharmacological manipulation of hypoxia (see ‘Role of hypoxia throughout the menstrual cycle’ section). Technically, culling by carbon dioxide (CO2) inhalation can impact tissue hypoxia and may distort results. Hence, cervical dislocation is the recommended culling method for these studies. Great care must be taken to handle, process and fix tissue rapidly to capture the physiological events of menstruation.
The mouse model of simulated menstruation
The menses-like model was first described in 1984 (Finn & Pope 1984) and further optimised in the 2000s (Brasted et al. 2003). Since then, it has been the most popular model to investigate the dynamics of endometrial repair (Fan et al. 2008, Evans et al. 2011, Cousins et al. 2014, Maybin et al. 2018, Chen et al. 2020) (Fig. 1). Mice are ovariectomised and supplemented with exogenous oestradiol and progesterone to mimic the human hormonal endometrial environment. They require artificial induction of decidualisation, via a transcervical or surgical intrauterine injection of oil. Once decidualisation has taken place, progesterone withdrawal leads to active bleeding in the mouse uterus and subsequent repair (Fig. 1A). Alternatively, the simulation of menses can be achieved by inducing pseudopregnancy (Fig. 1B). In this model, female mice are mated with vasectomised males to mimic fertilisation events. Progesterone withdrawal occurs naturally or is induced by ovariectomy or administration of a progesterone antagonist (Rudolph et al. 2012).
The first work to describe the presence of hypoxia during endometrial breakdown and repair in the mouse utilised the ‘pseudopregnancy’ model variant (Fan et al. 2008). Pimonidazole is a hypoxic marker that, when oxygen partial pressures are below 10 mmHg, forms protein adducts which can be visualised using specific monoclonal antibodies. Due to its chemical stability, pimonidazole is considered one of the most reliable means of tissue oxygen level detection, even when it is temporally and spatially transient. Fan et al. found the endometrial area undergoing regeneration to be hypoxic and that this hypoxia decreased and eventually disappeared with endometrial reepithelialisation (Fan et al. 2008). Subsequent confirmation of the presence of menstrual hypoxia was found in the ‘exogenous hormone’ model of simulated menses (Cousins et al. 2016b, Maybin et al. 2018, Chen et al. 2020). Using pimonidazole, hypoxia was detected during bleeding and later confined to areas undergoing active repair. Hypoxia may also be present in the endometrium at the time of implantation. As the uterine epithelium contains no blood vessels during initial embryo contact, it has been suggested that the onset of implantation occurs in a hypoxic environment (Daikoku et al. 2003). The detection of pimonidazole adducts in the area of implantation in mice reinforces this hypothesis (Pringle et al. 2007).
Another method to determine tissue hypoxia is the detection of the oxygen-sensing transcription factor hypoxia-inducible factor (HIF). HIFs have a key role in the cellular response to oxygen and are heterodimers composed of two subunits: a constitutively expressed beta subunit (HIF1B) and an O2-sensitive alpha subunit (Semenza 2000). There are three known α subunits: HIF1A, HIF2A, and HIF3A. HIF1A and HIF2A are the most common alpha isoforms and present overlapping but distinct target gene specificities (Mole et al. 2009). HIF3A is structurally different from the other isoforms and is the least characterised (Pasanen et al. 2010). Along with promoting genes related to nitrogen metabolism and immune response, HIF3A has the ability to inhibit HIF1A/2A action (Zhang et al. 2014).
The regulation of HIF takes place predominantly at the protein level. In normoxia, prolyl hydroxylase domain enzymes (PHDs) hydroxylate specific residues within the alpha subunit, leading to its ubiquitination and subsequent degradation via the proteasome (Salceda & Caro 1997). In hypoxia these PHDs are inhibited, resulting in HIFA stabilisation. HIFA translocates to the nucleus, dimerises with HIF1A and binds to hypoxia-response elements (HREs) to enhance transcription of a plethora of genes involved in energy metabolism, angiogenesis, tissue remodelling and inflammatory responses (Semenza 2012).
The presence of nuclear HIF1A protein is, therefore, indicative of active HIF1 and consistent with tissue hypoxia. Using this approach, HIF1A has been detected during menstruation in both the exogenous hormone (Maybin et al. 2018, Chen et al. 2020) and pseudopregnancy menstruation models (Chen et al. 2015), decreasing during endometrial regeneration. Examination of HIF1A and HIF2A in the mouse uterus during pre-implantation (day 4) and decidualisation (days 5–8) of pregnancy, revealed HIF1A was present in the luminal epithelium prior to implantation and throughout the epithelium and stroma during decidualisation and implantation (Daikoku et al. 2003). HIF2A was seen in the stroma on day 4 and limited to cells surrounding the blastocyst on day 5. The authors suggested that HIF1 was involved in maintaining oxygen homeostasis and that HIF2 was driving the angiogenesis necessary for successful implantation.
Various concerns have been raised about using HIF as a hypoxic surrogate marker. Transient hypoxic events can be too brief to stabilise HIF for immunohistochemical detection (Wang et al. 1995). Antibody unreliability is an added factor, which is compounded by the fact that tissue collection and fixation can also affect HIF detection (Zhang & Salamonsen 2002). Furthermore, HIF stabilisation can be induced by NF-κB-driven cytokine production in a non-hypoxic dependent manner, and hypoxia can exert downstream effects independently of HIF signalling (Lin & Simon 2016).
Alongside detection of pimonidazole and HIF, hypoxia-inducible factor downstream targets may indicate a hypoxic response in the mouse menstrual endometrium. HIF1A-mediated induction of the angiogenic factors vascular endothelial growth factor (VEGF) and the chemokine receptor CXCR4 was increased during menstruation and endometrial repair (Fan et al. 2008, Chen et al. 2015, Cousins et al. 2016b, Maybin et al. 2018).
Xenograft mouse model
The xenograft mouse model provides an alternative model for the study of menstrual physiology and pathology (extensively reviewed in Kuokkanen et al. 2017). Human functional endometrium is transplanted into immunodeficient mice (Fig. 1C). This is usually collected during the proliferative phase and can be transplanted as (1) small fragments (1–2 mm3) of endometrial tissue (Guo et al. 2011, Coudyzer et al. 2013) or (2) dissociated endometrial cells from epithelial and stromal fractions that are mixed before implantation (Masuda et al. 2007, Polotsky et al. 2009).
The recipient mice are selected to limit xenograft tissue rejection, but the immunodeficient strain used can vary. The most commonly used in xenograft menstruation models is the severe combined immunodeficiency (SCID) mouse, which has T and B cell deficiencies (Gaide Chevronnay et al. 2009, Guo et al. 2011, Coudyzer et al. 2013). The best engraftment results are achieved with the non-obese diabetic (NOD)/SCID/γcnull mice (NOG), which also have defective NK cell activity (Matsuura-Sawada et al. 2005, Masuda et al. 2007).
Generally, the patches of endometrial tissue are placed subcutaneously in mice (Guo et al. 2011, Coudyzer et al. 2013) with a survival time of 4 weeks, whereas the dissociated endometrial cells are implanted below the kidney capsule and survive up to 10 weeks (Masuda et al. 2007). This latter mode of implantation allows extension of the duration of experiments, making this the method of choice for studies of the proliferation kinetics of the endometrium after pharmacological treatments (Polotsky et al. 2009).
Xenograft menstruation studies mainly focus on endometrial regeneration and the role of ovarian steroids in orchestrating the process (Gaide Chevronnay et al. 2009, Guo et al. 2011, Coudyzer et al. 2015) and use the endometrial fragments model variant. To date, this mouse model has only been employed once to study the presence of hypoxia during menstruation (Coudyzer et al. 2013). In 2013, Coudyzer et al. subcutaneously implanted endometrial patches on SCID female mice and tested for signs of hypoxia in the resulting xenograft using several methods. First, they directly measured the local partial oxygen pressure (pO2) using electron paramagnetic resonance and OxyLite fluorescent probes. They also studied the presence of pimonidazole staining and HIF1A using immunohistochemistry (IHC). The authors did not detect hypoxia during endometrial breakdown or repair using any of these methods. These results contrast with findings in the mouse model of simulated menses and may be partially explained by the xenograft model itself. Endometrial tissue architecture and vasculature is severely compromised following transplantation and may impair vasoconstriction and prevent endometrial hypoxia. Moreover, endometrial breakdown and repair are considered inflammatory events, as they involve pro-inflammatory cytokine production and myeloid leukocyte recruitment (Finn 1986). Therefore, the necessary immunosuppressed state of the recipient mice may alter physiological menstrual endometrial events. The SCID model aims to suppress T and B-cell mediated transplant/xenograft rejection without substantially affecting the innate immune response and may be more relevant than other immunocompromised recipient mice (Guo et al. 2011, Donoghue et al. 2012).
Spiny mouse
The common spiny mouse (Acomys cahirinus) is, to date, the only known rodent to display spontaneous decidualisation and natural menstruation (Bellofiore et al. 2017, 2018).
Although anatomically different, the spiny mouse uterus has physiological similarities to the human endometrium. For example, the spiny mouse displays spiral arteriole remodelling in the perimenstrual phase (Bellofiore et al. 2018). In addition, endometrial decidualisation is tightly controlled, not compromising the structural integrity of the endometrial glands or the myometrium, as observed in other mouse models (Bellofiore et al. 2018). Hypoxia has not been examined in this rodent to date and these studies are awaited with interest.
Macaque models
Macaques have morphologically similar uteri to humans, a similar length of menstrual cycle and they display spontaneous decidualisation (Brenner & Slayden 2012). Macaques also experience menstrual abnormalities (e.g. heavy menstrual bleeding (HMB)) and can be fitted with tampons, hence they are exceptional candidates for evaluating therapies for menstrual disorders (reviewed in Brenner & Slayden 2012). Despite menstruating naturally, macaques are routinely ovariectomised and treated with oestradiol and progesterone to create artificial menstrual cycles and enable accurate timing of endometrial sampling. However, the need for larger experimental groups, longer experimental times and the increased cost of these experiments has meant many researchers are now preferentially using rodent models to study menstrual physiology.
As previously mentioned, the first indication of endometrial tissue hypoxia was observed in endometrial explants transplanted to the eye of rhesus macaques in the 1940s (Markee 1940). Rather than hypoxia, Markee observed pulses of intense vasoconstriction in the spiral arterioles that he associated with localised hypoxic ischemia. This hypothesis was later supported by the detection and increased expression of HIF1A in the functional layer of the macaque endometrium during menstruation (Brenner & Slayden 2012), consistent with the presence of endometrial hypoxia.
Ex vivo human endometrial studies
HIF1A protein has been identified, both by Western blot and IHC, in human endometrial biopsies collected during the late secretory and menstrual phases (Critchley et al. 2006, Maybin et al. 2018). HIF1A staining was localised in the glandular and stromal cells in the functional endometrium, whereas in the basal layer HIF1A staining was restricted to the glands.
In contrast, HIF2A is present exclusively during the early-mid secretory phase (Maybin et al. 2018). Downstream targets of HIF, such as VEGF and carbonic anhydrase IX (CA9), have also been shown to be increased during the menstrual and proliferative phases (Charnock-Jones et al. 1993, Sharkey et al. 2000, Punyadeera et al. 2006, Maybin et al. 2011b).
In vivo human endometrial studies
Detection of human endometrial hypoxia in vivo has been largely via measurements of perfusion, initially investigated using thermal heat dissipation (Prill & Götz 1961) and later by a Xenon-133 clearance technique (Fraser et al. 1987) (Fig. 2). Both methods are invasive, and results were conflicting as suffering from variable calibration and poor spatial and temporal resolution, respectively. The introduction of Doppler ultrasound allowed perfusion measurements in individual spiral arterioles (Kupesic & Kurjak 1993), but this showed an increase in flow the day before ovulation, in contrast with the 133Xe clearance study which found a fall at this time. Laser Doppler fluxmetry was able to assess endometrial perfusion using a fibre optic probe (Gannon et al. 1997), finding blood flow peaks in the early proliferative and early secretory phase, but spatial resolution was limited. The more sensitive three-dimensional power Doppler angiography (3D-PDA) was also used in spiral arterioles (Raine-Fenning et al. 2004) and revealed a significant pre-ovulatory peak in perfusion, followed by a post-ovulatory fall and gradual increase through early to mid-secretory phases. In general, there has been little consensus regarding changes in endometrial blood flow over the menstrual cycle and how to measure such changes. MRI methods may now offer a better alternative, although there has been little work on the application of these techniques to detect endometrial hypoxia.
To our knowledge, functional investigation of the normal endometrium has been limited to MR spectroscopy (Sarac et al. 2004, Celik et al. 2005). This technique detects the presence of specific metabolites in the body by examining the resonant frequencies of the hydrogen protons within them. In particular, lactate is a product of anaerobic respiration (and therefore a marker of hypoxia) and has been detected in normal secretory and proliferative endometrium (Sarac et al. 2004, Celik et al. 2005). Although lactate is arguably a more direct marker of hypoxia than measurement of perfusion, analysis and acquisition of spectroscopy data is technically challenging (Lange et al. 2006) and spatial resolution tends to be poor.
Dynamic contrast-enhanced (DCE) MRI is a technique that can detect hypoxia indirectly by measuring perfusion using an exogenous gadolinium-based contrast agent (CA) (Sourbron 2010). Passage of the CA through the tissue can be modelled to allow perfusion to be estimated as part of a model-fitting process (Sourbron & Buckley 2012). The technique has been applied in the normal endometrium (Majd et al. 2017) but showed no differences between the secretory and proliferative phases. The advantage of DCE-MRI for hypoxia imaging is its good spatial resolution, but imaging and analysis can be complex (Brix et al. 2004, 2009, Michaely et al. 2008) and there is no gold standard for validation of the technique. Use of DCE-MRI to detect a reduction in perfusion related to hypoxia in the menstrual cycle would require a specialised imaging protocol and robust data analysis using a complex model, including estimation of parameter uncertainties.
Other existing MRI techniques could be applied to measure endometrial hypoxia (Fig. 2). T2* is a characteristic tissue relaxation time that depends on inhomogeneities in the main magnetic field produced by the scanner as well as rapidly changing inhomogeneities induced by the presence of other nearby molecules. Detection of a reduction in T2* is commonly assumed to be due to the presence of deoxyhaemoglobin and therefore, tissue hypoxia. This technique has been used in the myometrium (Kido et al. 2007, Imaoka et al. 2012) and has a high spatial resolution necessary to investigate the endometrium. T2* can change for a number of other reasons (e.g. local haematocrit, hemosiderin, calcification and tissue iron deposition) therefore changes should be interpreted with caution. Similarly, a non-invasive perfusion technique known as arterial spin labelling (ASL) (Ferré et al. 2013) could be extended from existing work in the myometrium (Takahashi et al. 2016) to the endometrium, though it can be technically challenging. Finally, the extensive work on hypoxia measurements in cancer (Horsman et al. 2012) could be applied in the endometrium. Oxygen-enhanced (OE) MRI (O’Connor et al. 2019) allows a change in the tissue relaxation time T1 as a result of the patient breathing 100% oxygen through a mask to be related to the oxygen status of the tissue (O’Connor et al. 2016). These minimally invasive MRI techniques may provide key information on the presence of human endometrial hypoxia throughout the menstrual cycle, with potential diagnostic and therapeutic benefits for women.
Role of hypoxia throughout the menstrual cycle
Mice have the experimental advantage of genetic or pharmacological alteration to assess the role of hypoxia in endometrial function. Hif1a heterozygote mice have revealed that HIF1A is required for normal menstruation, and decreased HIF1A delays endometrial repair (Maybin et al. 2018). Pharmacological stabilisation and inhibition of HIF1A in mice have confirmed this role (Chen et al. 2015, Maybin et al. 2018). Mice placed in hyperoxic chambers (75% O2) during menses had reduced local endometrial hypoxia at menstruation and delayed endometrial repair (Maybin et al. 2018). Hif2a deficiency restricted to uterine stromal cells in a mouse implantation model revealed a key role in decidualisation, endometrial receptivity, embryonic implantation and survival (Matsumoto et al. 2018).
This emerging evidence for the presence and important role of hypoxia and HIF in endometrial function presents an exciting and developing research area (Fig. 3). The effects of hypoxia on the important menstrual processes of inflammation, proliferation and tissue remodelling remain to be elucidated.
Impact of hypoxia on inflammation
Inflammation is a key event during implantation, at menstruation and the subsequent endometrial repair. There is a peri-menstrual influx of leukocytes into the endometrium, in particular, neutrophils and macrophages (Armstrong et al. 2017). Interactions between the inflammatory response and hypoxia are well described at other tissue sites (Cramer et al. 2003, Taylor 2008, Taylor et al. 2016) but the impact of hypoxia on the endometrial inflammatory response is less well characterised.
Impact on neutrophils
Neutrophils comprise up to 15% of the total endometrial cell numbers during menstruation (Poropatich et al. 1987, Salamonsen & Lathbury 2000). Their influx is tightly regulated, displaying a rapid, short-lasting induction, which coincides with the upregulation of chemokines and cytokines. This temporal dynamic has been observed in both the mouse model of simulated menses and in human endometrial samples (Armstrong et al. 2017). Neutrophils are important mediators of endometrial breakdown, which has been confirmed by their depletion in the mouse model of menstruation (Kaitu’u-Lino et al. 2007b). However, the depleting agent used in this study also affects the monocytic cell lineage. Activated neutrophils release enzymes, such as neutrophil elastase and cathepsin G. These enzymes activate MMPs produced by endometrial stromal cells and cause degradation of the extracellular matrix (Salamonsen & Lathbury 2000). In airway inflammation, hypoxia boosts neutrophil degranulation and protease release (Hoenderdos et al. 2016). It would be informative to determine whether hypoxia has similar effects in the endometrial environment during menses.
Neutrophils also produce reactive oxygen species (ROS) that might participate in the endometrial breakdown. The potential role of ROS in menstruation has been reported (Sugino et al. 1996), suggesting that free oxygen radicals may contribute to endometrial shedding by causing tissue damage. Indeed, the inhibition of ROS generation in the mouse model of simulated menstruation has been shown to abrogate endometrial breakdown (Wu et al. 2014).
Neutrophil depletion in mouse models also affected endometrial regeneration (Kaitu’u-Lino et al. 2007b). Little is known about the impact of hypoxia on neutrophils during the endometrial repair. The concept that hypoxia has an effect on neutrophil number and function is derived from studies of tumour biology. In a mouse model of endometrial carcinoma, there was spatiotemporal correlation between hypoxia and neutrophil infiltration within the tumour (Blaisdell et al. 2015). Accumulation of pimonidazole and nuclear staining of HIF1A was detected slightly prior to neutrophil infiltration. These results are consistent with those observed in the mouse model of simulated menses, where pharmacological inhibition of HIF1A decreased the number of endometrial neutrophils present during active bleeding (Maybin et al. 2018). The role of hypoxia in promoting neutrophil recruitment in endometrial carcinoma was confirmed by placing mice in hyperoxic chambers (60% O2) (Mahiddine et al. 2020). This resulted in a dramatic reduction in neutrophil influx within the tumour and also improved the ability of these cells to oppose tumour growth through increased activation and expression of several MMPs and ROS production. This is consistent with hypoxia not only affecting the recruitment of neutrophils, but also their function. Determining the effects of hypoxia on neutrophil number and phenotype in the normal endometrium would be of great interest to advance our understanding of menstrual physiology.
Effects of hypoxia on neutrophils have also been observed in benign tissues. Airway inflammation studies have revealed that hypoxia, via HIF1A and HIF2A, prolonged neutrophil lifespan by inhibiting apoptosis (Walmsley et al. 2005, Thompson et al. 2014). Glucocorticoids have also been shown to delay neutrophil apoptosis in vitro, but this did not occur in the presence of hypoxia (Marwick et al. 2013). Neutrophil apoptosis has been identified in the menstrual endometrium of mice, when hypoxia is present (Armstrong et al. 2017). In addition, glucocorticoids have been identified as having an important role in the human menstrual endometrium (McDonald et al. 2006, Rae et al. 2009). The impact of hypoxia on endometrial myeloid apoptosis has not been examined to date.
Impact on macrophages
Macrophages have been detected in the endometrium throughout the menstrual cycle, both close to the endometrial glands and in the stromal compartment (Bonatz et al. 1992). They show a peri-menstrual peak in number, reaching up to 15% of the cell total number at the time of menses (Salamonsen & Woolley 1999). Like neutrophils, it is proposed that macrophages play a critical role in the onset of endometrial breakdown via production and release of MMPs (reviewed in Critchley et al. 2001, Thiruchelvam et al. 2013). There are also indications of their involvement in glandular remodelling (Garry et al. 2010) and endometrial regeneration (Maybin et al. 2012, Cousins et al. 2016a), including the regulation of angiogenesis (Thiruchelvam et al. 2016).
Macrophages are remarkably plastic cells, capable of shifting towards different phenotypes by sensing the surrounding microenvironment (Martinez et al. 2008). Thus, their microenvironment may affect their recruitment and function. Historically, macrophage polarisation has been categorised as classical (M1) or alternative (M2). M1 phenotype is associated with microbicidal properties and M2 reflects a more regulatory, anti-inflammatory phenotype. More recently, macrophage polarisation is understood to be a dynamic spectrum of macrophage transition in response to environmental cues (Martinez & Gordon 2014). As there is mounting evidence for hypoxia in the local endometrial environment at menstruation (Cousins et al. 2016b, Maybin et al. 2018), it is important to determine its effect on endometrial macrophages.
Under physiological conditions, M2 macrophages are involved in angiogenesis and cellular clearance, hence promote wound healing. However, tumour-infiltrating macrophages (TAMs) are often correlated with poor cancer prognosis (Kawanaka et al. 2008). TAMs have been shown to be retained in hypoxic regions of tumours through the Sema3A/Neuropilin-1signaling axis, which is regulated by HIF2A (Casazza et al. 2013). The influence of hypoxia on TAMs is not only limited to the macrophage number but also influences their phenotype. Indeed, specific TAM phenotypical subsets have been reported depending on intra-tumoral oxygen levels (Laoui et al. 2014).
Non-tumoral studies have also linked HIF to changes in macrophage phenotype. In a model of endotoxemia, HIF1A and HIF2A were differentially expressed in M1 and M2-macrophages, respectively (Takeda et al. 2010). In addition, in the context of obesity and adipose tissue inflammation, HIF1A has been proven to promote inflammation and insulin resistance through M1 macrophage polarisation whereas HIF2A ameliorated the effects via M2-macrophage induction (reviewed in Lin & Simon 2016). Interestingly, HIF1A was found to be decreased in mouse adipose tissue when glucocorticoid activation was suppressed, suggesting a crucial role of glucocorticoids in HIF-dependent macrophage polarisation (Chapman et al. 2013). Thus, different research fields converge around the concept that HIF1A may be required for M1 polarisation of macrophages, while HIF2A might promote M2 polarisation.
The menstrual endometrium presents a unique model of transient, physiological hypoxia in which to study macrophage number and phenotype. HIF2A may have a role in the recruitment and function of macrophages during implantation, when endometrial HIF2A was found to be present (Maybin et al. 2018). However, a recent study of mice with a targeted deletion of Hif1a in myeloid cells resulted in decreased pregnancy rates and increased miscarriage rates, suggesting that HIF1A dependent pathways in myeloid cells are also important for the maintenance of pregnancy (Köstlin-Gille et al. 2019). It would be informative to establish if the balance between HIF1A/HIF2A determines the pro-inflammatory or anti-inflammatory fate of the endometrium.
Impact of hypoxia on proliferation
After ‘injury’, fibroblasts must migrate and proliferate in the damaged area, where they produce extracellular matrix (ECM) components that contribute to repair (Gonzalez et al. 2016). This production must be tightly regulated to prevent excessive ECM growth, scar formation and fibrosis (Ruthenborg et al. 2014). In dermal tissue, hypoxia has been shown to stimulate macrophage growth factors that may contribute to fibroblast proliferation and tissue repair (Murdoch et al. 2005). Macrophage production of platelet-derived growth factor (PDGF) enhances fibroblast mitosis, while transforming growth factor β (TGFB) promotes the formation of the ECM (Ruthenborg et al. 2014). In addition, hypoxia has been proven to induce the transcription of VEGF, connective tissue growth factor and adrenomedullin in endometrial stromal tissue (Maybin et al. 2011a, Maybin et al. 2012). Hence, hypoxia may induce a pro-repair environment by modifying the secretome of endometrial cell populations.
To complete tissue restoration, reepithelialisation of the affected area must take place. In the skin, this is achieved through the migration and proliferation of keratinocytes towards the injury site (Ruthenborg et al. 2014). Stabilisation of HIF1A in a mouse model of skin wound healing revealed its role in promoting keratinocyte proliferation and migration to the injured area, accelerating wound closure (Kalucka et al. 2013). This is consistent with the findings of delayed endometrial repair with decreased HIF1A (Maybin et al. 2018).
Impact of hypoxia on vascular remodelling and angiogenesis
Angiogenesis and vascular remodelling are crucial events in the endometrium throughout the menstrual cycle. Optimal vascular function is necessary to support the repair of the functional endometrial layer and to supply the thickened endometrium required for successful implantation and placentation.
VEGF is a key mediator of both physiological and tumoral angiogenesis and may be induced by hypoxia (Carmeliet 2005). VEGF mRNA and protein have been detected during all phases of the menstrual cycle, both in the stromal compartment and the glandular epithelium ( Charnock-Jones et al. 1993, Shifren et al. 1996, Punyadeera et al. 2006) but was maximal during menses (Sharkey et al. 2000, Graubert et al. 2001, Maybin et al. 2011b). Studies in mouse and macaque models of menstruation have shown that blocking VEGF dramatically decreases reepithelialisation and new blood vessel formation in the endometrium (Fan et al. 2008), consistent with an essential role for VEGF in endometrial angiogenesis and repair.
Hypoxia has been detected in the mouse model of simulated menses (Chen et al. 2015, Cousins et al. 2016b, Maybin et al. 2018) and coincides with increased VEGF mRNA (Cousins et al. 2016b). Hypoxia and VEGF have also been detected in human perimenstrual endometrial biopsies (Punyadeera et al. 2006) highlighting their possible interrelation. In vitro studies have also shown that subjecting endometrial and epithelial stromal cells to hypoxia increases VEGF mRNA and protein (Popovici et al. 1999, Sharkey et al. 2000, Graubert et al. 2001) and that silencing of HIF1A abrogates this hypoxia-induced VEGF expression (Maybin et al. 2011b, Chen et al. 2015). Through a chromatin immunoprecipitation (ChIP) assay, Chen et al. detected the direct binding of HIF1A to the VEGF promoter, which was maximal during the endometrial breakdown of the mouse model of menses (Chen et al. 2015). Inhibition of HIF1A using 2-methoxyestradiol (2-ME) significantly suppressed VEGF levels during menses. Therefore, hypoxia, and more specifically HIF1A, seems to promote endometrial VEGF during menses.
In addition, VEGF expression is induced by different cytokines and chemokines (Li et al. 1995, Stavri et al. 1995, Zagzag et al. 2006), some of which contain hypoxic response elements. Optimal blood vessel formation requires the trafficking of endothelial progenitors cells through the interaction of the chemokine CXCL12 with its receptor CXCR4 (Ruthenborg et al. 2014). Both ligand and receptor have been found to be upregulated by HIF1A, contributing to angiogenesis and blood vessel repair partly through VEGF (Ceradini et al. 2004, Zagzag et al. 2006). CXCL12 and CXCR4 have been described in the human endometrium (Ruiz et al. 2010) and endometrial CXCR4 was found to be decreased in patients with heavy menstrual bleeding (Maybin et al. 2018). Hence, the interactions between hypoxia pathways and inflammatory processes may significantly influence endometrial vascular function.
During decidualisation there is in vitro evidence that endometrial stromal cells increase VEGF mRNA and protein (Popovici et al. 1999, Matsui et al. 2004) and that hypoxia induces further increases in VEGF (Popovici et al. 1999). This VEGF production may be responsible for macrophage recruitment and polarisation towards a pro-angiogenic M2 phenotype (Wheeler et al. 2018). Thus, the responsiveness of the decidualised stroma to hypoxia suggests a possible role in the preparation of the endometrial vasculature for implantation. Uterine HIF2A deficiency has been shown to impair decidualisation in mice, revealing a downregulation of prolactin-related factors which can compromise the maintenance of the corpus luteum and therefore endometrial receptivity (Matsumoto et al. 2018).
When studying implantation in mice, HIF factors were found to be differentially expressed at the time of peri-implantation: HIF1A was detected in the luminal epithelium, whereas HIF2A expression was limited to the stromal compartment and neither correlated with VEGF expression (Daikoku et al. 2003). Therefore, HIF effects on implantation seem to be more versatile than simply contributing to vessel formation, playing a substantial role in decidualisation, endometrial receptivity and embryo survival (Matsumoto et al. 2018). After implantation, HIF1A was found in the luminal epithelium and the decidual layer. However, the strongest signal came from HIF2A, with expression localised to stromal cells surrounding the blastocyst. This post-implantation HIF2A expression was correlated with VEGF induction, switching to a proangiogenic stimulus once implantation had taken place (Daikoku et al. 2003).
The role of hypoxia in endometrial pathology
As outlined above, the literature regarding the influence of hypoxia on inflammation, proliferation and vascular function is increasing (Fig. 3). The influence of oxygen levels on implantation, placentation and disorders such as pre-eclampsia has been comprehensively reviewed within this series by Burton (2009). The impact of hypoxia on embryo function has been covered in detail by Dunwoodie (2009). Therefore, this section is focused on the role of endometrial hypoxia during menstruation and its potential in the identification of novel diagnostic and therapeutic strategies.
Abnormal uterine bleeding
Abnormal uterine bleeding (AUB) affects 20–30% of pre-menopausal women and over 800,000 women seek treatment in the UK each year (National Heavy Menstrual Bleeding Audit 2011). Available medical treatments are often discontinued due to side effects or lack of efficacy. Research in this area was previously hindered by lack of a consistent classification system for the diagnosis of causes of AUB. This was rectified by the development of the FIGO classification system of structural and non-structural causes (Munro et al. 2011, 2018) (Fig. 4).
Structural causes of AUB
Structural causes of AUB can be detected on examination or investigation of the uterus, for example, polyps, adenomyosis, leiomyoma (fibroids) and malignancy (Munro et al. 2011, 2018). These conditions have previously been under-diagnosed, with clinicians often treating the symptom of AUB without identifying the underlying cause. This has limited our knowledge on why these conditions develop and why they result in AUB.
Adenomyosis is the presence of ectopic endometrial glands and stroma within the myometrial layer of the uterus. It occurs in 7–27% of reproductive-aged women and presents with painful, heavy menstrual bleeding (Naftalin et al. 2012, Mavrelos et al. 2017). The impact of the adenomyotic lesions on the eutopic endometrium and the mechanisms causing AUB is not well understood. AUB due to adenomyosis (AUB-A) is particularly challenging as it is often resistant to medical treatment and surgical options (ablation or hysterectomy) are unacceptable to those wishing to preserve their fertility.
There is some evidence that the hypoxic response is aberrant within adenomyotic lesions. A study of hysterectomy samples from 14 women with adenomyosis and 9 without revealed increased VEGF protein in the eutopic endometrium of women with adenomyosis and increased VEGF and HIF1A protein in ectopic vs eutopic endometrium (Goteri et al. 2009). This suggests that a hypoxic environment in the adenomyotic lesions could contribute to increased vessel formation. In endometriosis, where ectopic endometrium implants outside of the uterus, HIF1A was also found to be increased in ectopic vs eutopic endometrium (Wu et al. 2007, Young et al. 2014). Inhibition of HIF1 in a mouse model of endometriosis suppressed the growth of lesions (Becker et al. 2008), identifying the hypoxia pathway as a potential therapeutic target. The peritoneum is a common site for implantation of ectopic endometrial deposits in endometriosis. Women with endometriosis have been shown to have increased HIF1A in non-affected peritoneum compared to peritoneum from women without disease (Young et al. 2014), consistent with a role of the hypoxia pathway in the development of peritoneal disease. Studies examining the non-affected myometrium in women with adenomyosis are not yet available, but similar alterations in hypoxic response would highlight hypoxia pathways as a potential target for preventative and therapeutic interventions.
Leiomyomas (uterine fibroids) are common, benign tumours of the myometrium that form as a consequence of the proliferation of uterine smooth muscle cells and collagen matrix. They occur in approximately 70% of women (Stewart et al. 2017) and are extremely heterogeneous in size, location and pathophysiology. Leiomyomas are symptomatic in approximately 50% of women (Day Baird et al. 2003) and may cause symptoms of AUB, pressure, pelvic pain and be associated with subfertility.
Genome wide association studies have identified genetic subgroups that may predispose to leiomyoma formation (reviewed in Stewart et al. 2016) but local mechanisms regulating their development remain an area of active research. Uterine leiomyomas contain broad avascular areas and HIF1A protein was found to be increased in leiomyoma nuclear protein extracts when compared to adjacent myometrium (Ishikawa et al. 2019). However, it is not yet clear whether hypoxia is necessary for leiomyoma development and/or growth. In contrast, an in vivo study of women with leiomyoma using DCE-MRI has revealed increased Ktrans (a combination of perfusion and permeability) in fibroids compared with the normal uterus (Majd et al. 2017) which does not support the presence of hypoxia within fibroids. There is evidence that treatment of leiomyomas with gonadotrophin-releasing hormone (GnRH) analogues, often used pre-operatively to reduce fibroid size and decrease AUB, lead to a decrease in perfusion parameters (Munro et al. 2014). These contrasting in vitro and in vivo findings may reflect the heterogeneity of leiomyomas and it remains unclear if altered perfusion is associated with AUB.
The cause of AUB experienced by a proportion of women with leiomyomas is not understood. Vasoconstriction may be impaired at the time of menstruation in women with fibroids, with leiomyoma tissue expressing altered levels of endothelin receptors and prostaglandin F2α when compared to normal myometrium (Pekonen et al. 1994, Miura et al. 2006). A small decrease in spiral arteriole vasoconstriction can significantly increase menstrual blood flow, causing heavy menstrual bleeding. A greater understanding of the role of hypoxia in leiomyoma formation and growth may identify new, specific treatments to reduce their presence, size and symptoms.
Endometrial cancer
The importance of hypoxia in the tumour microenvironment is well established, including its influence on immune cell populations, angiogenesis, tumour progression and metastasis (De Bock et al. 2011, Casazza et al. 2014, Schito & Semenza 2016, Semenza 2016). The accuracy of translation of these principles to patients with endometrial cancer is less well determined. In a quest to identify a robust biomarker that would predict tumour behaviour, Chang et al. identified an eight-gene set of lymphocyte and tumour hypoxia markers and validated its performance in predicting overall survival in six cancers, including 370 women with endometrial cancer (Chang et al. 2019). They found a superior performance over current tumour staging parameters, highlighting the importance of hypoxia in determining risk and aiding clinical decision making.
Assessment of endometrial tissues from 386 patients with endometrial carcinoma using CA9 as a hypoxia marker and CD34 to determine vascular density, revealed that patients with the presence of both hypoxia and high vascular density (16.4%) had reduced disease-specific survival and distant disease-free survival (Reijnen et al. 2019). In vivo imaging with DCE-MRI revealed that a poor prognosis was associated with low microvascular blood flow to the endometrial tumour (Haldorsen et al. 2013, 2014, Berg et al. 2016). This was thought to reflect disorganised angiogenesis with coexisting vascular proliferation and hypoxia. These studies highlight normalisation of the vasculature to limit hypoxia as a potential therapeutic target in endometrial cancer.
Non-structural causes of AUB
These non-structural disorders are not usually identified by routine pelvic imaging. They include coagulopathies, ovulatory dysfunction, endometrial and iatrogenic causes (Munro et al. 2018). Evidence for a role of hypoxia in these disorders is limited but its contribution to AUB of endometrial origin (AUB-E) is discussed below.
AUB-E includes disorders of local endometrial haemostasis, vascular function and/or inflammation (Fig. 4). Women with objectively defined heavy menstrual bleeding (HMB; >80 mL/cycle) had reduced levels of HIF1A protein and downstream target genes in menstrual phase endometrial biopsies when compared to those from women with normal blood loss (Maybin et al. 2018). Examination of endometrial repair in mice where hypoxia was prevented during simulated menses, or where HIF1A was pharmacologically or genetically reduced, revealed delayed repair (Maybin et al. 2018). This is consistent with hypoxia having a key role in the rapid endometrial repair necessary to limit menstrual blood loss. The delayed repair in a non-hypoxic mouse menstruation model could be rescued with a pharmacological compound that stabilises HIF1, identifying a potential non-hormonal therapeutic target for women with AUB-E.
The cause of the endometrial tissue hypoxia observed at menstruation is unknown. It is likely that spiral arteriole vasoconstriction limits blood supply to the functional layer of the endometrium following progesterone withdrawal (Markee 1940). Hence, factors that limit the ability of the specialised endometrial arterioles to constrict will have a significant impact on the presence of endometrial hypoxia. Women with the symptom of HMB have been shown to have significantly decreased smooth muscle myosin heavy chain in their spiral arterioles and also reduced vascular smooth muscle cell proliferation during the mid-late secretory phase compared to those with normal menstrual blood loss (Abberton et al. 1999). Another study showed that endometrial vessel wall circumference and endothelial cell focal discontinuities were both significantly larger in women with HMB compared to normal controls (Mints et al. 2007). Furthermore, calponin (a vascular smooth muscle cell contractile protein) was found to be significantly lower in endometrial blood vessels in women with HMB (Biswas Shivhare et al. 2014). This evidence is all consistent with an aberrant vasculature within the pre-menstrual endometrium of women with AUB-E, leading to a suboptimal hypoxic response during menstruation.
Conclusions
Herein, we have reviewed the mounting evidence for the presence of endometrial hypoxia and its potential impact on endometrial function. Furthering our understanding of hypoxia in endometrial physiology and pathology using the tools described in this review may provide novel preventative and therapeutic strategies for those suffering from endometrial disorders, including abnormal uterine bleeding (AUB). Furthermore, a complete understanding of optimal endometrial physiology may inform the management of other disorders where aberrant hypoxia is a prominent feature, such as tumour biology and chronic obstructive pulmonary disorder. Addressing the gaps in our knowledge of how hypoxia influences endometrial function represents an exciting area with huge translational potential.
Declaration of interest
R M-A, L K, J J R and J A M have nothing to disclose. H O D C has clinical research support for laboratory consumables and staff from Bayer AG and provides consultancy advice (but with no personal remuneration) for Bayer AG, PregLem SA, Gedeon Richter, Vifor Pharma UK Ltd, AbbVie Inc; Myovant Sciences GmbH. H O D C receives royalties from UpToDate for article on abnormal uterine bleeding. J A Maybin is a Guest Editor of this special issue of Reproduction. J A Maybin was not involved in the review or editorial process for this paper, on which she is listed as an author.
Funding
Some aspects of studies described herein were undertaken in the MRC Centre for Reproductive Health and have been supported by funding from the Medical Research Council: MRC Project Grant G0600048 and Centre Grants G1002033 and MR/N022556/1. This work was also supported by Wellcome Trust Grants 209589/Z/17/Z and 100646/Z/21/Z; Wellbeing of Women Grant RG1820; and Academy of Medical Sciences Grant AMS-SGCL13.
Author contribution statement
R M-A, L K and J A M wrote and edited the manuscript. J J R and H O D C modified the manuscript. All authors have read and approved the final version of this manuscript.
Acknowledgements
We are grateful to Alison Murray for manuscript review and Sheila Milne for assistance with manuscript formatting.
References
Abberton KM, Taylor NH, Healy DL & Rogers PAW 1999 Vascular smooth muscle cell proliferation in arterioles of the human endometrium. Human Reproduction 14 1072–1079. (https://doi.org/10.1093/humrep/14.4.1072)
Armstrong GM, Maybin JA, Murray AA, Nicol M, Walker C, Saunders PTK, Rossi AG & Critchley HOD 2017 Endometrial apoptosis and neutrophil infiltration during menstruation exhibits spatial and temporal dynamics that are recapitulated in a mouse model. Scientific Reports 7 17416. (https://doi.org/10.1038/s41598-017-17565-x)
Baird DT, Cameron ST, Critchley HOD, Drudy TA, Howe A, Jones RL, Lea RG & Kelly RW 1996 Prostaglandins and menstruation. European Journal of Obstetrics, Gynecology, and Reproductive Biology 70 15–17. (https://doi.org/10.1016/s0301-2115(9602568-7)
Becker CM, Rohwer N, Funakoshi T, Cramer T, Bernhardt W, Birsner A, Folkman J & D’Amato RJ 2008 2-Methoxyestradiol inhibits hypoxia-inducible factor-1α and suppresses growth of lesions in a mouse model of endometriosis. American Journal of Pathology 172 534–544. (https://doi.org/10.2353/ajpath.2008.061244)
Bellofiore N, Ellery SJ, Mamrot J, Walker DW, Temple-Smith P & Dickinson H 2017 First evidence of a menstruating rodent: the spiny mouse (Acomys cahirinus). American Journal of Obstetrics and Gynecology 216 40.e1–40.e11. (https://doi.org/10.1016/j.ajog.2016.07.041)
Bellofiore N, Rana S, Dickinson H, Temple-Smith P & Evans J 2018 Characterization of human-like menstruation in the spiny mouse: comparative studies with the human and induced mouse model. Human Reproduction 33 1715–1726. (https://doi.org/10.1093/humrep/dey247)
Berg A, Fasmer KE, Mauland KK, Ytre-Hauge S, Hoivik EA, Husby JA, Tangen IL, Trovik J, Halle MK & Woie K et al. 2016 Tissue and imaging biomarkers for hypoxia predict poor outcome in endometrial cancer. Oncotarget 7 69844–69856. (https://doi.org/10.18632/oncotarget.12004)
Biswas Shivhare S, Bulmer JN, Innes BA, Hapangama DK & Lash GE 2014 Altered vascular smooth muscle cell differentiation in the endometrial vasculature in menorrhagia. Human Reproduction 29 1884–1894. (https://doi.org/10.1093/humrep/deu164)
Blaisdell A, Crequer A, Columbus D, Daikoku T, Mittal K, Dey SK & Erlebacher A 2015 Neutrophils oppose uterine epithelial carcinogenesis via debridement of hypoxic tumor cells. Cancer Cell 28 785–799. (https://doi.org/10.1016/j.ccell.2015.11.005)
Bonatz G, Hansmann ML, Buchholz F, Mettler L, Radzun HJ & Semm K 1992 Macrophage- and lymphocyte-subtypes in the endometrium during different phases of the ovarian cycle. International Journal of Gynaecology and Obstetrics 37 29–36. (https://doi.org/10.1016/0020-7292(9290974-n)
Brasted M, White CA, Kennedy TG & Salamonsen LA 2003 Mimicking the events of menstruation in the murine uterus. Biology of Reproduction 69 1273–1280. (https://doi.org/10.1095/biolreprod.103.016550)
Brenner RM & Slayden OD 2012 Molecular and functional aspects of menstruation in the macaque. Reviews in Endocrine and Metabolic Disorders 13 309–318. (https://doi.org/10.1007/s11154-012-9225-5)
Brix G, Kiessling F, Lucht R, Darai S, Wasser K, Delorme S & Griebel J 2004 Microcirculation and microvasculature in breast tumors: pharmacokinetic analysis of dynamic MR image series. Magnetic Resonance in Medicine 52 420–429. (https://doi.org/10.1002/mrm.20161)
Brix G, Zwick S, Kiessling F & Griebel J 2009 Pharmacokinetic analysis of tissue microcirculation using nested models: multimodel inference and parameter identifiability. Medical Physics 36 2923–2933. (https://doi.org/10.1118/1.3147145)
Burton GJ 2009 Oxygen, the Janus gas; its effects on human placental development and function. Journal of Anatomy 215 27–35. (https://doi.org/10.1111/j.1469-7580.2008.00978.x)
Carmeliet P 2005 VEGF as a key mediator of angiogenesis in cancer. Oncology 69 (Supplement 3) 4–10. (https://doi.org/10.1159/000088478)
Casazza A, Laoui D, Wenes M, Rizzolio S, Bassani N, Mambretti M, Deschoemaeker S, Van Ginderachter JA, Tamagnone L & Mazzone M 2013 Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell 24 695–709. (https://doi.org/10.1016/j.ccr.2013.11.007)
Casazza A, Di Conza G, Wenes M, Finisguerra V, Deschoemaeker S & Mazzone M 2014 Tumor stroma: a complexity dictated by the hypoxic tumor microenvironment. Oncogene 33 1743–1754. (https://doi.org/10.1038/onc.2013.121)
Celik O, Hascalik S, Sarac K, Meydanli MM, Alkan A & Mizrak B 2005 Magnetic resonance spectroscopy of premalignant and malignant endometrial disorders: a feasibility of in vivo study. European Journal of Obstetrics, Gynecology, and Reproductive Biology 118 241–245. (https://doi.org/10.1016/j.ejogrb.2004.06.030)
Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, Capla JM, Galiano RD, Levine JP & Gurtner GC 2004 Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nature Medicine 10 858–864. (https://doi.org/10.1038/nm1075)
Chang WH, Forde D & Lai AG 2019 A novel signature derived from immunoregulatory and hypoxia genes predicts prognosis in liver and five other cancers. Journal of Translational Medicine 17 14. (https://doi.org/10.1186/s12967-019-1775-9)
Chapman KE, Coutinho AE, Zhang Z, Kipari T, Savill JS & Seckl JR 2013 Changing glucocorticoid action: 11β-hydroxysteroid dehydrogenase type 1 in acute and chronic inflammation. Journal of Steroid Biochemistry and Molecular Biology 137 82–92. (https://doi.org/10.1016/j.jsbmb.2013.02.002)
Charnock-Jones DS, Sharkey AM, Rajput-Williams J, Burch D, Schofield JP, Fountain SA, Boocock CA & Smith SK 1993 Identification and localization of alternately spliced mRNAs for vascular endothelial growth factor in human uterus and estrogen regulation in endometrial carcinoma cell lines. Biology of Reproduction 48 1120–1128. (https://doi.org/10.1095/biolreprod48.5.1120)
Chen X, Liu J, He B, Li Y, Liu S, Wu B, Wang S, Zhang S, Xu X & Wang J 2015 Vascular endothelial growth factor (VEGF) regulation by hypoxia inducible factor-1 alpha (HIF1A) starts and peaks during endometrial breakdown, not repair, in a mouse menstrual-like model. Human Reproduction 30 2160–2170. (https://doi.org/10.1093/humrep/dev156)
Chen X, Wu B, Wang S, Liu J, Gao H, Zhou F, Nan N, Zhang B, Wang J & Xu X et al. 2020 Hypoxia: involved but not essential for endometrial breakdown in mouse menstural-like model. Reproduction 159 133–144. (https://doi.org/10.1530/REP-18-0562)
Corker CS, Michie E, Hobson B & Parboosingh J 1976 Hormonal patterns in conceptual cycles and early pregnancy. British Journal of Obstetrics and Gynaecology 83 489–494. (https://doi.org/10.1111/j.1471-0528.1976.tb00871.x)
Coudyzer P, Lemoine P, Jordan BF, Gallez B, Galant C, Nisolle M, Courtoy PJ, Henriet P & Marbaix E 2013 Hypoxia is not required for human endometrial breakdown or repair in a xenograft model of menstruation. FASEB Journal 27 3711–3719. (https://doi.org/10.1096/fj.13-232074)
Coudyzer P, Lemoine P, Po C, Jordan BF, Van Der Smissen P, Courtoy PJ, Henriet P & Marbaix E 2015 Induction of post-menstrual regeneration by ovarian steroid withdrawal in the functionalis of xenografted human endometrium. Human Reproduction 30 1156–1168. (https://doi.org/10.1093/humrep/dev043)
Cousins FL, Murray A, Esnal A, Gibson DA, Critchley HOD & Saunders PTK 2014 Evidence from a mouse model that epithelial cell migration and mesenchymal-epithelial transition contribute to rapid restoration of uterine tissue integrity during menstruation. PLoS ONE 9 e86378. (https://doi.org/10.1371/journal.pone.0086378)
Cousins FL, Kirkwood PM, Saunders PTK & Gibson DA 2016a Evidence for a dynamic role for mononuclear phagocytes during endometrial repair and remodelling. Scientific Reports 6 36748. (https://doi.org/10.1038/srep36748)
Cousins FL, Murray AA, Scanlon JP & Saunders PTK 2016b HypoxyprobeTM reveals dynamic spatial and temporal changes in hypoxia in a mouse model of endometrial breakdown and repair. BMC Research Notes 9 30. (https://doi.org/10.1186/s13104-016-1842-8)
Cramer T, Yamanishi Y, Clausen BE, Förster I, Pawlinski R, Mackman N, Haase VH, Jaenisch R, Corr M & Nizet V et al. 2003 HIF-1α is essential for myeloid cell-mediated inflammation. Cell 112 645–657. (https://doi.org/10.1016/s0092-8674(0300154-5)
Critchley HOD, Jones RL, Lea RG, Drudy TA, Kelly RW, Williams ARW & Baird DT 1999 Role of inflammatory mediators in human endometrium during progesterone withdrawal and early pregnancy. Journal of Clinical Endocrinology and Metabolism 84 240–248. (https://doi.org/10.1210/jcem.84.1.5380)
Critchley HOD, Kelly RW, Brenner RM & Baird DT 2001 The endocrinology of menstruation - a role for the immune system. Clinical Endocrinology 55 701–710. (https://doi.org/10.1046/j.1365-2265.2001.01432.x)
Critchley HOD, Henderson TA, Kelly RW, Scobie GS, Evans LR, Groome NP & Saunders PTK 2002 Wild-type estrogen receptor (ERβ1) and the splice variant (ERβcx/β2) are both expressed within the human endometrium throughout the normal menstrual cycle. Journal of Clinical Endocrinology and Metabolism 87 5265–5273. (https://doi.org/10.1210/jc.2002-020502)
Critchley HOD, Osei J, Henderson TA, Boswell L, Sales KJ, Jabbour HN & Hirani N 2006 Hypoxia-inducible factor-1α expression in human endometrium and its regulation by prostaglandin E-series prostanoid receptor 2 (EP2). Endocrinology 147 744–753. (https://doi.org/10.1210/en.2005-1153)
Critchley HOD, Maybin JA, Armstrong GM & Williams ARW 2020 Physiology of the endometrium and regulation of menstruation. Physiological Reviews 100 1149–1179. (https://doi.org/10.1152/physrev.00031.2019)
Daikoku T, Matsumoto H, Gupta RA, Das SK, Gassmann M, DuBois RN & Dey SK 2003 Expression of hypoxia-inducible factors in the peri-implantation mouse uterus is regulated in a cell-specific and ovarian steroid hormone-dependent manner. Evidence for differential function of HIFs during early pregnancy. Journal of Biological Chemistry 278 7683–7691. (https://doi.org/10.1074/jbc.M211390200)
Day Baird DD, Dunson DB, Hill MC, Cousins D & Schectman JM 2003 High cumulative incidence of uterine leiomyoma in black and white women: ultrasound evidence. American Journal of Obstetrics and Gynecology 188 100–107. (https://doi.org/10.1067/mob.2003.99)
De Bock K, Mazzone M & Carmeliet P 2011 Antiangiogenic therapy, hypoxia, and metastasis: risky liaisons, or not? Nature Reviews: Clinical Oncology 8 393–404. (https://doi.org/10.1038/nrclinonc.2011.83)
Donoghue JF, McGavigan CJ, Lederman FL, Cann LM, Fu L, Dimitriadis E, Girling JE & Rogers PAW 2012 Dilated thin-walled blood and lymphatic vessels in human endometrium: a potential role for VEGF-D in progestin-induced break-through bleeding. PLoS ONE 7 e30916. (https://doi.org/10.1371/journal.pone.0030916)
Dunn CL, Kelly RW & Critchley HO 2003 Decidualization of the human endometrial stromal cell: an enigmatic transformation. Reproductive Biomedicine Online 7 151–161. (https://doi.org/10.1016/s1472-6483(1061745-2)
Dunwoodie SL 2009 The role of hypoxia in development of the mammalian embryo. Developmental Cell 17 755–773. (https://doi.org/10.1016/j.devcel.2009.11.008)
Evans J, Kaitu’u-Lino T & Salamonsen LA 2011 Extracellular matrix dynamics in scar-free endometrial repair: perspectives from mouse in vivo and human in vitro studies. Biology of Reproduction 85 511–523. (https://doi.org/10.1095/biolreprod.111.090993)
Fan X, Krieg S, Kuo CJ, Wiegand SJ, Rabinovitch M, Druzin ML, Brenner RM, Giudice LC & Nayak NR 2008 VEGF blockade inhibits angiogenesis and reepithelialization of endometrium. FASEB Journal 22 3571–3580. (https://doi.org/10.1096/fj.08-111401)
Ferré JC, Bannier E, Raoult H, Mineur G, Carsin-Nicol B & Gauvrit JY 2013 Arterial spin labeling (ASL) perfusion: techniques and clinical use. Diagnostic and Interventional Imaging 94 1211–1223. (https://doi.org/10.1016/j.diii.2013.06.010)
Finn CA 1986 Implantation, menstruation and inflammation. Biological Reviews of the Cambridge Philosophical Society 61 313–328. (https://doi.org/10.1111/j.1469-185x.1986.tb00657.x)
Finn CA & Pope M 1984 Vascular and cellular changes in the decidualized endometrium of the ovariectomized mouse following cessation of hormone treatment: a possible model for menstruation. Journal of Endocrinology 100 295–300. (https://doi.org/10.1677/joe.0.1000295)
Fraser IS, McCarron G, Hutton B & Macey D 1987 Endometrial blood flow measured by xenon 133 clearance in women with normal menstrual cycles and dysfunctional uterine bleeding. American Journal of Obstetrics and Gynecology 156 158–166. (https://doi.org/10.1016/0002-9378(8790229-8)
Gaide Chevronnay HP, Galant C, Lemoine P, Courtoy PJ, Marbaix E & Henriet P 2009 Spatiotemporal coupling of focal extracellular matrix degradation and reconstruction in the menstrual human endometrium. Endocrinology 150 5094–5105. (https://doi.org/10.1210/en.2009-0750)
Gannon BJ, Carati CJ & Verco CJ 1997 Endometrial perfusion across the normal human menstrual cycle assessed by laser Doppler fluxmetry. Human Reproduction 12 132–139. (https://doi.org/10.1093/humrep/12.1.132)
Garry R, Hart R, Karthigasu KA & Burke C 2010 Structural changes in endometrial basal glands during menstruation. BJOG 117 1175–1185. (https://doi.org/10.1111/j.1471-0528.2010.02630.x)
Gellersen B & Brosens JJ 2014 Cyclic decidualization of the human endometrium in reproductive health and failure. Endocrine Reviews 35 851–905. (https://doi.org/10.1210/er.2014-1045)
Gonzalez AC, Costa TF, Andrade ZA & Medrado AR 2016 Wound healing – a literature review. Anais brasileiros de dermatologia 91 614–620. (https://doi.org/10.1590/abd1806-4841.20164741)
Goteri G, Lucarini G, Montik N, Zizzi A, Stramazzotti D, Fabris G, Tranquilli AL & Ciavattini A 2009 Expression of vascular endothelial growth factor (VEGF), hypoxia inducible factor-1α (HIF-1α), and microvessel density in endometrial tissue in women with adenomyosis. International Journal of Gynecological Pathology 28 157–163. (https://doi.org/10.1097/PGP.0b013e318182c2be)
Graubert MD, Asuncion Ortega MA, Kessel B, Mortola JF & Iruela-Arispe ML 2001 Vascular repair after menstruation involves regulation of vascular endothelial growth factor-receptor phosphorylation by sFLT-1. American Journal of Pathology 158 1399–1410. (https://doi.org/10.1016/s0002-9440(1064091-6)
Guo Y, He B, Xu X & Wang J 2011 Comprehensive analysis of leukocytes, vascularization and matrix metalloproteinases in human menstrual xenograft model. PLoS ONE 6 e16840. (https://doi.org/10.1371/journal.pone.0016840)
Haldorsen IS, Grüner R, Husby JA, Magnussen IJ, Werner HM, Salvesen OO, Bjørge L, Stefansson I, Akslen LA & Trovik J et al. 2013 Dynamic contrast-enhanced MRI in endometrial carcinoma identifies patients at increased risk of recurrence. European Radiology 23 2916–2925. (https://doi.org/10.1007/s00330-013-2901-3)
Haldorsen IS, Stefansson I, Grüner R, Husby JA, Magnussen IJ, Werner HM, Salvesen ØO, Bjørge L, Trovik J & Taxt T et al. 2014 Increased microvascular proliferation is negatively correlated to tumour blood flow and is associated with unfavourable outcome in endometrial carcinomas. British Journal of Cancer 110 107–114. (https://doi.org/10.1038/bjc.2013.694)
Hamlett GWD 1934 Uterine bleeding in a bat, Glossophaga soricina. Anatomical Record 60 9–17. (https://doi.org/10.1002/ar.1090600104)
Hoenderdos K, Lodge KM, Hirst RA, Chen C, Palazzo SG, Emerenciana A, Summers C, Angyal A, Porter L & Juss JK et al. 2016 Hypoxia upregulates neutrophil degranulation and potential for tissue injury. Thorax 71 1030–1038. (https://doi.org/10.1136/thoraxjnl-2015-207604)
Horsman MR, Mortensen LS, Petersen JB, Busk M & Overgaard J 2012 Imaging hypoxia to improve radiotherapy outcome. Nature Reviews: Clinical Oncology 9 674–687. (https://doi.org/10.1038/nrclinonc.2012.171)
Imaoka I, Nakatsuka T, Araki T, Katsube T, Okada M, Kumano S, Ishii K, Ashikaga R, Okuaki T & Van Cauteren M et al. 2012 T2* Relaxometry mapping of the uterine zones. Acta Radiologica 53 473–477. (https://doi.org/10.1258/ar.2012.110342)
Ishikawa H, Xu L, Sone K, Kobayashi T, Wang G & Shozu M 2019 Hypoxia induces hypoxia-inducible factor 1α and potential HIF-responsive gene expression in uterine leiomyoma. Reproductive Sciences 26 428–435. (https://doi.org/10.1177/1933719118776793)
Kaitu’u-Lino TJ, Morison NB & Salamonsen LA 2007a Estrogen is not essential for full endometrial restoration after breakdown: lessons from a mouse model. Endocrinology 148 5105–5111. (https://doi.org/10.1210/en.2007-0716)
Kaitu’u-Lino TJ, Morison NB & Salamonsen LA 2007b Neutrophil depletion retards endometrial repair in a mouse model. Cell and Tissue Research 328 197–206. (https://doi.org/10.1007/s00441-006-0358-2)
Kalucka J, Ettinger A, Franke K, Mamlouk S, Singh RP, Farhat K, Muschter A, Olbrich S, Breier G & Katschinski DM et al. 2013 Loss of epithelial hypoxia-inducible factor prolyl hydroxylase 2 accelerates skin wound healing in mice. Molecular and Cellular Biology 33 3426–3438. (https://doi.org/10.1128/MCB.00609-13)
Kawanaka T, Kubo A, Ikushima H, Sano T, Takegawa Y & Nishitani H 2008 Prognostic significance of HIF-2α expression on tumor infiltrating macrophages in patients with uterine cervical cancer undergoing radiotherapy. Journal of Medical Investigation 55 78–86. (https://doi.org/10.2152/jmi.55.78)
Kelly RW, King AE & Critchley HO 2001 Cytokine control in human endometrium. Reproduction 121 3–19. (https://doi.org/10.1530/rep.0.1210003)
Kido A, Koyama T, Kataoka M, Yamamoto A, Saga T, Turner R & Togashi K 2007 Physiological changes of the human uterine myometrium during menstrual cycle: preliminary evaluation using Bold MR imaging. Journal of Magnetic Resonance Imaging 26 695–700. (https://doi.org/10.1002/jmri.21061)
King AE, Critchley HO & Kelly RW 2001 The NF-kappaB pathway in human endometrium and first trimester decidua. Molecular Human Reproduction 7 175–183. (https://doi.org/10.1093/molehr/7.2.175)
Köstlin-Gille N, Dietz S, Schwarz J, Spring B, Pauluschke-Fröhlich J, Poets CF & Gille C 2019 HIF-1α-deficiency in myeloid cells leads to a disturbed accumulation of myeloid derived suppressor cells (MDSC) during pregnancy and to an increased abortion rate in mice. Frontiers in Immunology 10 161. (https://doi.org/10.3389/fimmu.2019.00161)
Kuokkanen S, Zhu L & Pollard JW 2017 Xenografted tissue models for the study of human endometrial biology. Differentiation: Research in Biological Diversity 98 62–69. (https://doi.org/10.1016/j.diff.2017.11.004)
Kupesic S & Kurjak A 1993 Uterine and ovarian perfusion during the periovulatory period assessed by transvaginal color Doppler. Fertility and Sterility 60 439–443. (https://doi.org/10.1016/s0015-0282(1656157-3)
Lange T, Dydak U, Roberts TPL, Rowley HA, Bjeljac M & Boesiger P 2006 Pitfalls in lactate measurements at 3T. American Journal of Neuroradiology 27 895–901.
Laoui D, Van Overmeire E, Di Conza G, Aldeni C, Keirsse J, Morias Y, Movahedi K, Houbracken I, Schouppe E & Elkrim Y et al. 2014 Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer Research 74 24–30. (https://doi.org/10.1158/0008-5472.CAN-13-1196)
Lessey BA, Killam AP, Metzger DA, Haney AF, Greene GL & McCarty KS 1988 Immunohistochemical analysis of human uterine estrogen and progesterone receptors throughout the menstrual cycle. Journal of Clinical Endocrinology and Metabolism 67 334–340. (https://doi.org/10.1210/jcem-67-2-334)
Li J, Perrella MA, Tsai JC, Yet SF, Hsieh CM, Yoshizumi M, Patterson C, Endege WO, Zhou F & Lee ME 1995 Induction of vascular endothelial growth factor gene expression by interleukin-1 in rat aortic smooth muscle cells. Journal of Biological Chemistry 270 308–312. (https://doi.org/10.1074/jbc.270.1.308)
Lin N & Simon MC 2016 Hypoxia-inducible factors: key regulators of myeloid cells during inflammation. Journal of Clinical Investigation 126 3661–3671. (https://doi.org/10.1172/JCI84426)
Mahiddine K, Blaisdell A, Ma S, Créquer-Grandhomme A, Lowell CA & Erlebacher A 2020 Relief of tumor hypoxia unleashes the tumoricidal potential of neutrophils. Journal of Clinical Investigation 130 389–403. (https://doi.org/10.1172/JCI130952)
Majd S, Johan Jeet Abdullah B & Ahmad Sarji S 2017 Quantitative dynamic contrast-enhanced magnetic resonance imaging in symptomatic uterine fibroids and normal uterus: a feasibility study. Iranian Journal of Radiology 14 1–10. (https://doi.org/10.5812/iranjradiol.36213)
Markee JE 1940 Menstruation in intraocular endometrial transplants in the rhesus monkey. Contributions to Embryology 219–308.
Marsh MM, Malakooti N, Taylor NH, Findlay JK & Salamonsen LA 1997 Endothelin and neutral endopeptidase in the endometrium of women with menorrhagia. Human Reproduction 12 2036–2040. (https://doi.org/10.1093/humrep/12.9.2036)
Martinez FO & Gordon S 2014 The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Reports 6 13. (https://doi.org/10.12703/P6-13)
Martinez FO, Sica A, Mantovani A & Locati M 2008 Macrophage activation and polarization. Frontiers in Bioscience 13 453–461. (https://doi.org/10.2741/2692)
Marwick JA, Dorward DA, Lucas CD, Jones KO, Sheldrake TA, Fox S, Ward C, Murray J, Brittan M & Hirani N et al. 2013 Oxygen levels determine the ability of glucocorticoids to influence neutrophil survival in inflammatory environments. Journal of Leukocyte Biology 94 1285–1292. (https://doi.org/10.1189/jlb.0912462)
Masuda H, Maruyama T, Hiratsu E, Yamane J, Iwanami A, Nagashima T, Ono M, Miyoshi H, Okano HJ & Ito M et al. 2007 Noninvasive and real-time assessment of reconstructed functional human endometrium in NOD/SCID/gamma c(null) immunodeficient mice. PNAS 104 1925–1930. (https://doi.org/10.1073/pnas.0604310104)
Matsui N, Kawano Y, Nakamura S & Miyakawa I 2004 Changes in vascular endothelial growth factor production associated with decidualization by human endometrial stromal cells in vitro. Acta Obstetricia et Gynecologica Scandinavica 83 138–143. (https://doi.org/10.1111/j.0001-6349.2004.00256.x)
Matsumoto L, Hirota Y, Saito-Fujita T, Takeda N, Tanaka T, Hiraoka T, Akaeda S, Fujita H, Shimizu-Hirota R & Igaue S et al. 2018 HIF2α in the uterine stroma permits embryo invasion and luminal epithelium detachment. Journal of Clinical Investigation 128 3186–3197. (https://doi.org/10.1172/JCI98931)
Matsuura-Sawada R, Murakami T, Ozawa Y, Nabeshima H, Akahira J, Sato Y, Koyanagi Y, Ito M, Terada Y & Okamura K 2005 Reproduction of menstrual changes in transplanted human endometrial tissue in immunodeficient mice. Human Reproduction 20 1477–1484. (https://doi.org/10.1093/humrep/deh783)
Mavrelos D, Holland TK, O’Donovan O, Khalil M, Ploumpidis G, Jurkovic D & Khalaf Y 2017 The impact of adenomyosis on the outcome of IVF–embryo transfer. Reproductive Biomedicine Online 35 549–554. (https://doi.org/10.1016/j.rbmo.2017.06.026)
Maybin JA, Battersby S, Hirani N, Nikitenko LL, Critchley HOD & Jabbour HN 2011a The expression and regulation of adrenomedullin in the human endometrium: a candidate for endometrial repair. Endocrinology 152 2845–2856. (https://doi.org/10.1210/en.2010-1256)
Maybin JA, Hirani N, Brown P, Jabbour HN & Critchley HOD 2011b The regulation of vascular endothelial growth factor by hypoxia and prostaglandin F2α during human endometrial repair. Journal of Clinical Endocrinology and Metabolism 96 2475–2483. (https://doi.org/10.1210/jc.2010-2971)
Maybin JA, Barcroft J, Thiruchelvam U, Hirani N, Jabbour HN & Critchley HOD 2012 The presence and regulation of connective tissue growth factor in the human endometrium. Human Reproduction 27 1112–1121. (https://doi.org/10.1093/humrep/der476)
Maybin JA, Murray AA, Saunders PTK, Hirani N, Carmeliet P & Critchley HOD 2018 Hypoxia and hypoxia inducible factor-1α are required for normal endometrial repair during menstruation. Nature Communications 9 295. (https://doi.org/10.1038/s41467-017-02375-6)
McDonald SE, Henderson TA, Gomez-Sanchez CE, Critchley HOD & Mason JI 2006 11β-Hydroxysteroid dehydrogenases in human endometrium. Molecular and Cellular Endocrinology 248 72–78. (https://doi.org/10.1016/j.mce.2005.12.010)
Michaely HJ, Sourbron SP, Buettner C, Lodemann KP, Reiser MF & Schoenberg SO 2008 Temporal constraints in renal perfusion imaging with a 2-compartment model. Investigative Radiology 43 120–128. (https://doi.org/10.1097/RLI.0b013e3181583b0c)
Mints M, Hultenby K, Zetterberg E, Blomgren B, Falconer C, Rogers R & Palmblad J 2007 Wall discontinuities and increased expression of vascular endothelial growth factor-A and vascular endothelial growth factor receptors 1 and 2 in endometrial blood vessels of women with menorrhagia. Fertility and Sterility 88 691–697. (https://doi.org/10.1016/j.fertnstert.2006.11.190)
Miura S, Khan KN, Kitajima M, Hiraki K, Moriyama S, Masuzaki H, Samejima T, Fujishita A & Ishimaru T 2006 Differential infiltration of macrophages and prostaglandin production by different uterine leiomyomas. Human Reproduction 21 2545–2554. (https://doi.org/10.1093/humrep/del205)
Mole DR, Blancher C, Copley RR, Pollard PJ, Gleadle JM, Ragoussis J & Ratcliffe PJ 2009 Genome-wide association of hypoxia-inducible factor (HIF)-1α and HIF-2α DNA binding with expression profiling of hypoxia-inducible transcripts. Journal of Biological Chemistry 284 16767–16775. (https://doi.org/10.1074/jbc.M901790200)
Munro MG, Critchley HOD, Fraser IS & FIGO Menstrual Disorders Working Group 2011 The FIGO classification of causes of abnormal uterine bleeding in the reproductive years. Fertility and Sterility 95 2204.e1–2208.e1. (https://doi.org/10.1016/j.fertnstert.2011.03.079)
Munro KI, Thrippleton MJ, Williams AR, McKillop G, Walker J, Horne AW, Newby DE, Anderson RA, Semple SI & Marshall I et al. 2014 Quantitative serial MRI of the treated fibroid uterus. PLoS ONE 9 e89809. (https://doi.org/10.1371/journal.pone.0089809)
Munro MG, Critchley HOD, Fraser IS & FIGO Menstrual Disorders Committee 2018 The two FIGO systems for normal and abnormal uterine bleeding symptoms and classification of causes of abnormal uterine bleeding in the reproductive years: 2018 revisions. International Journal of Gynaecology and Obstetrics 143 393–408. (https://doi.org/10.1002/ijgo.12666)
Murdoch C, Muthana M & Lewis CE 2005 Hypoxia regulates macrophage functions in inflammation. Journal of Immunology 175 6257–6263. (https://doi.org/10.4049/jimmunol.175.10.6257)
Mutsaers SE, Birnie K, Lansley S, Herrick SE, Lim CB & Prêle CM 2015 Mesothelial cells in tissue repair and fibrosis. Frontiers in Pharmacology 6 113. (https://doi.org/10.3389/fphar.2015.00113)
Naftalin J, Hoo W, Pateman K, Mavrelos D, Holland T & Jurkovic D 2012 How common is adenomyosis? A prospective study of prevalence using transvaginal ultrasound in a gynaecology clinic. Human Reproduction 27 3432–3439. (https://doi.org/10.1093/humrep/des332)
Noyes RW, Hertig AT & Rock J 1950 Dating the endometrial biopsy. Fertility and Sterility 1 3–25.
O’Connor JPB, Boult JK, Jamin Y, Babur M, Finegan KG, Williams KJ, Little RA, Jackson A, Parker GJ & Reynolds AR et al. 2016 Oxygen-enhanced MRI accurately identifies, quantifies, and maps tumor hypoxia in preclinical cancer models. Cancer Research 76 787–795. (https://doi.org/10.1158/0008-5472.CAN-15-2062)
O’Connor JPB, Robinson SP & Waterton JC 2019 Imaging tumour hypoxia with oxygen-enhanced MRI and BOLD MRI. British Journal of Radiology 92 20180642. (https://doi.org/10.1259/bjr.20180642)
Pasanen A, Heikkilä M, Rautavuoma K, Hirsilä M, Kivirikko KI & Myllyharju J 2010 Hypoxia-inducible factor (HIF)-3α is subject to extensive alternative splicing in human tissues and cancer cells and is regulated by HIF-1 but not HIF-2. International Journal of Biochemistry and Cell Biology 42 1189–1200. (https://doi.org/10.1016/j.biocel.2010.04.008)
Pekonen F, Nyman T & Rutanen EM 1994 Differential expression of mRNAs for endothelin-related proteins in human endometrium, myometrium and leiomyoma. Molecular and Cellular Endocrinology 103 165–170. (https://doi.org/10.1016/0303-7207(9490084-1)
Polotsky AJ, Zhu L, Santoro N & Pollard JW 2009 Lithium chloride treatment induces epithelial cell proliferation in xenografted human endometrium. Human Reproduction 24 1960–1967. (https://doi.org/10.1093/humrep/dep115)
Popovici RM, Irwin JC, Giaccia AJ & Giudice LC 1999 Hypoxia and cAMP stimulate vascular endothelial growth factor (VEGF) in human endometrial stromal cells: potential relevance to menstruation and endometrial regeneration. Journal of Clinical Endocrinology and Metabolism 84 2245–2248. (https://doi.org/10.1210/jcem.84.6.5886)
Poropatich C, Rojas M & Silverberg SG 1987 Polymorphonuclear leukocytes in the endometrium during the normal menstrual cycle. International Journal of Gynecological Pathology 6 230–234. (https://doi.org/10.1097/00004347-198709000-00004)
Prill HJ & Götz F 1961 Blood flow in the myometrium and endometrium of the uterus. American Journal of Obstetrics and Gynecology 82 102–108. (https://doi.org/10.1016/s0002-9378(1636102-6)
Pringle KG, Kind KL, Thompson JG & Roberts CT 2007 Complex interactions between hypoxia inducible factors, insulin-like growth factor-II and oxygen in early murine trophoblasts. Placenta 28 1147–1157. (https://doi.org/10.1016/j.placenta.2007.05.009)
Punyadeera C, Thijssen VL, Tchaikovski S, Kamps R, Delvoux B, Dunselman GA, de Goeij AF, Griffioen AW & Groothuis PG 2006 Expression and regulation of vascular endothelial growth factor ligands and receptors during menstruation and post-menstrual repair of human endometrium. Molecular Human Reproduction 12 367–375. (https://doi.org/10.1093/molehr/gal027)
Rae M, Mohamad A, Price D, Hadoke PWF, Walker BR, Mason JI, Hillier SG & Critchley HOD 2009 Cortisol inactivation by 11β-hydroxysteroid dehydrogenase-2 may enhance endometrial angiogenesis via reduced thrombospondin-1 in heavy menstruation. Journal of Clinical Endocrinology and Metabolism 94 1443–1450. (https://doi.org/10.1210/jc.2008-1879)
Raine-Fenning NJ, Campbell BK, Kendall NR, Clewes JS & Johnson IR 2004 Quantifying the changes in endometrial vascularity throughout the normal menstrual cycle with three-dimensional power Doppler angiography. Human Reproduction 19 330–338. (https://doi.org/10.1093/humrep/deh056)
Rasweiler JJ & de Bonilla H 1992 Menstruation in short-tailed fruit bats (Carollia spp.). Journal of Reproduction and Fertility 95 231–248. (https://doi.org/10.1530/jrf.0.0950231)
Reijnen C, van Weelden WJ, Arts MSJP, Peters JP, Rijken PF, van de Vijver K, Santacana M, Bronsert P, Bulten J & Hirschfeld M et al. 2019 Poor outcome in hypoxic endometrial carcinoma is related to vascular density. British Journal of Cancer 120 1037–1044. (https://doi.org/10.1038/s41416-019-0461-2)
Royal College of Obstreticians and Gynaecologists 2014 National Heavy Menstrual Bleeding Audit.
Rudolph M, Döcke WD, Müller A, Menning A, Röse L, Zollner TM & Gashaw I 2012 Induction of overt menstruation in intact mice. PLoS ONE 7 e32922. (https://doi.org/10.1371/journal.pone.0032922)
Ruiz A, Salvo VA, Ruiz LA, Báez P, García M & Flores I 2010 Basal and steroid hormone-regulated expression of CXCR4 in human endometrium and endometriosis. Reproductive Sciences 17 894–903. (https://doi.org/10.1177/1933719110379920)
Ruthenborg RJ, Ban JJ, Wazir A, Takeda N & Kim JW 2014 Regulation of wound healing and fibrosis by hypoxia and hypoxia-inducible factor-1. Molecules and Cells 37 637–643. (https://doi.org/10.14348/molcells.2014.0150)
Salamonsen LA & Lathbury LJ 2000 Endometrial leukocytes and menstruation. Human Reproduction Update 6 16–27. (https://doi.org/10.1093/humupd/6.1.16)
Salamonsen LA & Woolley DE 1999 Menstruation: induction by matrix metalloproteinases and inflammatory cells. Journal of Reproductive Immunology 44 1–27. (https://doi.org/10.1016/s0165-0378(9900002-9)
Salceda S & Caro J 1997 Hypoxia-inducible factor 1α (HIF-1α) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions. Journal of Biological Chemistry 272 22642–22647. (https://doi.org/10.1074/jbc.272.36.22642)
Sarac K, Celik O, Hascalik S, Alkan A & Mizrak B 2004 In vivo proton magnetic resonance spectroscopy in the evaluation of the endometrium. Acta Obstetricia et Gynecologica Scandinavica 83 751–757. (https://doi.org/10.1111/j.0001-6349.2004.00435.x)
Schito L & Semenza GL 2016 Hypoxia-inducible factors: master regulators of cancer progression. Trends in Cancer 2 758–770. (https://doi.org/10.1016/j.trecan.2016.10.016)
Semenza GL 2000 HIF-1: mediator of physiological and pathophysiological responses to hypoxia. Journal of Applied Physiology 88 1474–1480. (https://doi.org/10.1152/jappl.2000.88.4.1474)
Semenza GL 2012 Hypoxia-inducible factors in physiology and medicine. Cell 148 399–408. (https://doi.org/10.1016/j.cell.2012.01.021)
Semenza GL 2016 The hypoxic tumor microenvironment: a driving force for breast cancer progression. Biochimica et Biophysica Acta 1863 382–391. (https://doi.org/10.1016/j.bbamcr.2015.05.036)
Sharkey AM, Day K, McPherson A, Malik S, Licence D, Smith SK & Charnock-Jones DS 2000 Vascular endothelial growth factor expression in human endometrium is regulated by hypoxia. Journal of Clinical Endocrinology and Metabolism 85 402–409. (https://doi.org/10.1210/jcem.85.1.6229)
Shifren JL, Tseng JF, Zaloudek CJ, Ryan IP, Meng YG, Ferrara N, Jaffe RB & Taylor RN 1996 Ovarian steroid regulation of vascular endothelial growth factor in the human endometrium: implications for angiogenesis during the menstrual cycle and in the pathogenesis of endometriosis. Journal of Clinical Endocrinology and Metabolism 81 3112–3118. (https://doi.org/10.1210/jcem.81.8.8768883)
Short RV 1976 The evolution of human reproduction. Proceedings of the Royal Society of London: Series B, Biological Sciences 195 3–24. (https://doi.org/10.1098/rspb.1976.0095)
Sourbron S 2010 Technical aspects of MR perfusion. European Journal of Radiology 76 304–313. (https://doi.org/10.1016/j.ejrad.2010.02.017)
Sourbron SP & Buckley DL 2012 Tracer kinetic modelling in MRI: estimating perfusion and capillary permeability. Physics in Medicine and Biology 57 R1–R33. (https://doi.org/10.1088/0031-9155/57/2/R1)
Stavri GT, Zachary IC, Baskerville PA, Martin JF & Erusalimsky JD 1995 Basic fibroblast growth factor upregulates the expression of vascular endothelial growth factor in vascular smooth muscle cells. Synergistic interaction with hypoxia. Circulation 92 11–14. (https://doi.org/10.1161/01.cir.92.1.11)
Stewart EA, Cookson CL, Gandolfo RA & Schulze-Rath R 2017 Epidemiology of uterine fibroids: a systematic review. BJOG 124 1501–1512. (https://doi.org/10.1111/1471-0528.14640)
Stewart EA, Laughlin-Tommaso SK, Catherino WH, Lalitkumar S, Gupta D & Vollenhoven B 2016 Uterine fibroids. Nature Reviews: Disease Primers 2 16043. (https://doi.org/10.1038/nrdp.2016.43)
Suda T, Takubo K & Semenza GL 2011 Metabolic regulation of hematopoietic stem cells in the hypoxic niche. Cell Stem Cell 9 298–310. (https://doi.org/10.1016/j.stem.2011.09.010)
Sugino N, Shimamura K, Takiguchi S, Tamura H, Ono M, Nakata M, Nakamura Y, Ogino K, Uda T & Kato H 1996 Changes in activity of superoxide dismutase in the human endometrium throughout the menstrual cycle and in early pregnancy. Human Reproduction 11 1073–1078. (https://doi.org/10.1093/oxfordjournals.humrep.a019299)
Takahashi N, Yoshino O, Hiraike O, Maeda E, Nakamura M, Hori M, Harada M, Koga K, Saito S & Fujii T et al. 2016 The assessment of myometrium perfusion in patients with uterine fibroid by arterial spin labeling MRI. SpringerPlus 5 1907. (https://doi.org/10.1186/s40064-016-3596-0)
Takeda N, O’Dea EL, Doedens A, Kim JW, Weidemann A, Stockmann C, Asagiri M, Simon MC, Hoffmann A & Johnson RS 2010 Differential activation and antagonistic function of HIF- isoforms in macrophages are essential for NO homeostasis. Genes and Development 24 491–501. (https://doi.org/10.1101/gad.1881410)
Taylor CT 2008 Interdependent roles for hypoxia inducible factor and nuclear factor-κB in hypoxic inflammation. Journal of Physiology 586 4055–4059. (https://doi.org/10.1113/jphysiol.2008.157669)
Taylor CT, Doherty G, Fallon PG & Cummins EP 2016 Hypoxia-dependent regulation of inflammatory pathways in immune cells. Journal of Clinical Investigation 126 3716–3724. (https://doi.org/10.1172/JCI84433)
Thiruchelvam U, Dransfield I, Saunders PTK & Critchley HOD 2013 The importance of the macrophage within the human endometrium. Journal of Leukocyte Biology 93 217–225. (https://doi.org/10.1189/jlb.0712327)
Thiruchelvam U, Maybin JA, Armstrong GM, Greaves E, Saunders PTK & Critchley HOD 2016 Cortisol regulates the paracrine action of macrophages by inducing vasoactive gene expression in endometrial cells. Journal of Leukocyte Biology 99 1165–1171. (https://doi.org/10.1189/jlb.5A0215-061RR)
Thompson AAR, Elks PM, Marriott HM, Eamsamarng S, Higgins KR, Lewis A, Williams L, Parmar S, Shaw G & McGrath EE et al. 2014 Hypoxia-inducible factor 2α regulates key neutrophil functions in humans, mice, and zebrafish. Blood 123 366–376. (https://doi.org/10.1182/blood-2013-05-500207)
Van der Horst CJ & Gillman J 1940 Mechanism of ovulation and corpus luteum formation in Elephantulus. Nature 145 974–974. (https://doi.org/10.1038/145974a0)
Velnar T, Bailey T & Smrkolj V 2009 The wound healing process: an overview of the cellular and molecular mechanisms. Journal of International Medical Research 37 1528–1542. (https://doi.org/10.1177/147323000903700531)
Walmsley SR, Print C, Farahi N, Peyssonnaux C, Johnson RS, Cramer T, Sobolewski A, Condliffe AM, Cowburn AS & Johnson N et al. 2005 Hypoxia-induced neutrophil survival is mediated by HIF-1α-dependent NF-κB activity. Journal of Experimental Medicine 201 105–115. (https://doi.org/10.1084/jem.20040624)
Wang GL, Jiang BH, Rue EA & Semenza GL 1995 Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. PNAS 92 5510–5514. (https://doi.org/10.1073/pnas.92.12.5510)
Wang Q, Xu X, He B, Li Y, Chen X & Wang J 2013 A critical period of progesterone withdrawal precedes endometrial breakdown and shedding in mouse menstrual-like model. Human Reproduction 28 1670–1678. (https://doi.org/10.1093/humrep/det052)
Wheeler KC, Jena MK, Pradhan BS, Nayak N, Das S, Hsu CD, Wheeler DS, Chen K & Nayak NR 2018 VEGF may contribute to macrophage recruitment and M2 polarization in the decidua. PLoS ONE 13 e0191040. (https://doi.org/10.1371/journal.pone.0191040)
Wu MH, Chen KF, Lin SC, Lgu CW & Tsai SJ 2007 Aberrant expression of leptin in human endometriotic stromal cells is induced by elevated levels of hypoxia inducible factor-1α. American Journal of Pathology 170 590–598. (https://doi.org/10.2353/ajpath.2007.060477)
Wu B, Chen X, He B, Liu S, Li Y, Wang Q, Gao H, Wang S, Liu J & Zhang S et al. 2014 ROS are critical for endometrial breakdown via NF-κB–COX-2 signaling in a female mouse menstrual-like model. Endocrinology 155 3638–3648. (https://doi.org/10.1210/en.2014-1029)
Young VJ, Brown JK, Maybin J, Saunders PTK, Duncan WC & Horne AW 2014 Transforming growth factor-β induced warburg-like metabolic reprogramming may underpin the development of peritoneal endometriosis. Journal of Clinical Endocrinology and Metabolism 99 3450–3459. (https://doi.org/10.1210/jc.2014-1026)
Zagzag D, Lukyanov Y, Lan L, Ali MA, Esencay M, Mendez O, Yee H, Voura EB & Newcomb EW 2006 Hypoxia-inducible factor 1 and VEGF upregulate CXCR4 in glioblastoma: implications for angiogenesis and glioma cell invasion. Laboratory Investigation 86 1221–1232. (https://doi.org/10.1038/labinvest.3700482)
Zhang J & Salamonsen LA 2002 Expression of hypoxia-inducible factors in human endometrium and suppression of matrix metalloproteinases under hypoxic conditions do not support a major role for hypoxia in regulating tissue breakdown at menstruation. Human Reproduction 17 265–274. (https://doi.org/10.1093/humrep/17.2.265)
Zhang X, Zhu C, Lin H, Yang Q, Ou Q, Li Y, Chen Z, Racey P, Zhang S & Wang H 2007 Wild fulvous fruit bats (Rousettus leschenaulti) exhibit human-like menstrual cycle. Biology of Reproduction 77 358–364. (https://doi.org/10.1095/biolreprod.106.058958)
Zhang P, Yao Q, Lu L, Li Y, Chen PJ & Duan C 2014 Hypoxia-inducible factor 3 is an oxygen-dependent transcription activator and regulates a distinct transcriptional response to hypoxia. Cell Reports 6 1110–1121. (https://doi.org/10.1016/j.celrep.2014.02.011)
Zheng L, Kelly CJ & Colgan SP 2015 Physiologic hypoxia and oxygen homeostasis in the healthy intestine. A review in the theme: cellular responses to hypoxia. American Journal of Physiology: Cell Physiology 309 C350–C360. (https://doi.org/10.1152/ajpcell.00191.2015)