Next Article in Journal
Systematical Screening of Intracellular Protein Targets of Polyphemusin-I Using Escherichia coli Proteome Microarrays
Next Article in Special Issue
The Impact of Hypoxia in Early Pregnancy on Placental Cells
Previous Article in Journal
Role of the Novel Peptide Phoenixin in Stress Response and Possible Interactions with Nesfatin-1
Previous Article in Special Issue
Uteroplacental Circulation in Normal Pregnancy and Preeclampsia: Functional Adaptation and Maladaptation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

HIV Associated Preeclampsia: A Multifactorial Appraisal

1
Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
2
Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban 4041, South Africa
3
Women’s Health and HIV Research Group, Department of Obstetrics and Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(17), 9157; https://doi.org/10.3390/ijms22179157
Submission received: 14 July 2021 / Revised: 16 August 2021 / Accepted: 18 August 2021 / Published: 25 August 2021

Abstract

:
Introduction: This review explores angiogenesis, vascular dysfunction, the complement system, RAAS, apoptosis and NETosis as potential pathways that are dysregulated during preeclampsia, HIV infection and ART usage. Results: HIV-1 accessory and matrix proteins are protagonists for the elevation of oxidative stress, apoptosis, angiogenesis, and elevation of adhesion markers. Despite the immunodeficiency during HIV-1 infection, HIV-1 exploits our cellular defence arsenal by escaping cell-mediated lysis, yet HIV-1 infectivity is enhanced via C5a release of TNF-α and IL-6. This review demonstrates that PE is an oxidatively stressed microenvironment associated with increased apoptosis and NETosis, but with a decline in angiogenesis. Immune reconstitution in the duality of HIV-1 and PE by protease inhibitors, HAART and nucleoside reverse transcriptase, affect similar cellular pathways that eventuate in loss of endothelial cell integrity and, hence, its dysfunction. Conclusions: HIV-1 infection, preeclampsia and ARTs differentially affect endothelial cell function. In the synergy of both conditions, endothelial dysfunction predominates. This knowledge will help us to understand the effect of HIV infection and ART on immune reconstitution in preeclampsia.

Graphical Abstract

1. Introduction

Low and middle-income countries have higher maternal mortality rates (462 per 100,000 live births) compared to high-income countries (11 per 100,000 live births) [1]. A systematic analysis conducted by the World Health Organisation (WHO) indicates that haemorrhage and hypertensive disorders of pregnancy are the major causes of direct maternal deaths worldwide [1,2]. Preeclampsia (PE) is a hypertensive disorder of pregnancy and one of the main direct causes of maternal morbidity and mortality, whereas human immunodeficiency virus (HIV) infection in pregnancy remains the leading indirect cause of maternal deaths [3].
Women of reproductive age account for 52% of all adult HIV infections worldwide [1,4]. In poor income countries, HIV infected women have 5.2–8 times the risk of direct maternal mortality compared to HIV uninfected women [5,6]. One in three pregnant women attending antenatal care in South Africa is HIV infected [7,8]. Despite antiretroviral therapy (ART) improving life expectancy and reducing mother to child HIV transmission, a significant number of maternal mortality rates from HIV infection still prevail [9,10]. In addition, adverse pregnancy outcomes, low birthweight and preterm births are reported in individuals receiving ART [11].
The COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is also associated with an increased risk of severe outcomes in pregnancy than the general population [12,13,14]. The SARS-CoV-2 infection superimposed on HIV-associated pregnancy is complex as both impact the inflammatory response and influence endothelial function [15,16].
The idiopathic condition of PE is diagnosed by the presence of de novo hypertension after 20 weeks of gestation with/without proteinuria and/or evidence of maternal acute kidney injury, liver dysfunction, neurological features, haemolysis or thrombocytopenia, or foetal growth restriction [17]. The pathogenesis of PE remains unknown; however, it involves defective placentation and inadequate spiral artery transformation in early pregnancy, resulting in reduced blood flow and inadequate nutrients and oxygen supply to the neonate [18]. The consequent hypoxic conditions promote widespread oxidative stress, angiogenic imbalance and an amplified immune and inflammatory response [19]. Endothelial dysfunction induces the maternal syndrome of PE [16].

2. Immune Response and the Complement System

In a healthy pregnancy, several immunological adaptations support immunologic tolerance to the semi-allogenic foetus and placenta and simultaneously protects the mother and foetus against pathogens [20]. The inhibitory action of maternal immune cells to paternal antigens in the developing foetus is key to a successful pregnancy. The evolving theory encompassing immune modulation during pregnancy suggests that placentation synergistically develops with the immune system to capitalize on the crosstalk linking trophoblast and immune cells, instead of circumventing the maternal decidual immune responses [21]. Maternal decidual natural killer cells (dNK) and foetal trophoblasts confer immunity at the maternal-foetal interface via receptor-signalling. Immune tolerance occurs via human leukocyte antigens (HLA), albeit their absence on syncytiotrophoblasts precludes them from detecting paternal antigens [21]. Stable forms of HLA-E and HLA-G are expressed by the human foetal extravillous trophoblasts (EVT), whilst HLA-C allotypes express an affinity to either stimulatory or inhibitory maternal dNK receptors [21].
A breakdown of immune tolerance or immunological incompatibility between the mother and foetus may be associated with PE development [22]. Preeclampsia is dominated by a proinflammatory (Th1) response [23]; however, in combination with HIV infection and usage of HAART, the Th1 response is exacerbated [24]. Placental perfusion and invasion are enhanced by the stimulatory effects of foetal HLA-C genetic variants, in contrast to their inhibitory invasive effects in PE [21].
The complement system, a part of the innate immunity, is associated with the host defence and removal of apoptotic cells, injured tissue debris and immune complexes, and mediates one’s adaptive response [25]. Activation of the complement cascade results in inflammation, opsonisation and lysis by forming the membrane attack complex [25,26].
In pregnancy, a degree of complement activation is required during placentation for development, and to support the removal of placental apoptotic debris [22,27]. Foetal protection at the maternal-foetal interface is enhanced via complement regulators [27]. Excessive activation or dysregulation, however, can result in tissue damage, anaphylatoxin release with consequent inflammation, vascular leakage and thrombosis [28]. Previous studies demonstrate a dysregulation of the circulatory and placental complement system in PE [29,30,31,32], suggestive that a fine balance must be maintained between complement activation and regulation.
HIV-1 infection causes immunodeficiency, hyperactivation of the immune system and chronic inflammation [33]. Nonetheless, activation of the complement system is an essential arsenal in the defence against HIV; however, it may also enhance HIV-1 infectivity [34]. The membrane spikes of HIV-1 consist of 3 glycoprotein (gp) 120 molecules linked together and anchored to the transmembrane protein, gp41. The glycoprotein 41 binds to complement component 1q (C1q) and activates the complement classical pathway, whereas the HIV-1 envelope gp120 binds to mannose-binding lectins and neutralises the virus [35]. This action triggers the activation of the complement lectin pathway, preventing viral entry [36]. Nonetheless, C5a increases TNF-α and IL-6 release, thereby promoting HIV-1 infection, whilst inhibition of its receptor C5aR reverses this action [37].
Additionally, HIV-1 incorporates the host cell complement regulatory proteins (CD59 and CD55) on its viral envelope to escape complement-mediated lysis [35]. The subsequent complement opsonized HIV-1 enhances viral infection of T and B cell lines [38]. Both HIV and PE are characterised by chronic inflammation, which may be aggravated by excessive complement activation [39,40].
People living with HIV and receiving ART still encounter a high rate of non-AIDS related comorbidities, which may be related to increased levels of systemic immune activation [40,41]. A 54% increase in complement activation in ART-treated adults versus healthy controls has been observed [40]. A recent study revealed a significant elevation in several complement factors in ART-treated HIV infected individuals vs. seronegative controls [41]. In addition, an upregulation in complement regulatory proteins CD35 and CD55 was demonstrated in ART-treated HIV infected PE compared to normotensive pregnant women [42], suggesting that HIV confers a protective effect in PE. All pregnant HIV infected women were on ART [42]. Furthermore, activation of the complement system may lead to endothelial cell injury and subsequent activation of the clotting cascade [43].

3. HIV Infection and Endothelial Dysfunction

Similar to PE [44], HIV infected individuals display increased systemic oxidative stress because it is able to suppress endogenous antioxidant enzymatic mechanisms [45,46]. In addition, syncytin, a captured retroviral gene, is expressed by placental syncytiotrophoblasts [47]. The dysregulation of this viral fusion protein of endosymbiotic endogenous retroviruses may be implicated in PE pathogenesis [47]. Recent studies suggest a causative role of the HIV-1-proteins tat, gp120 and nef on endothelial dysfunction (Figure 1) [48].
The endothelium plays an important role in the pathogenesis of viral infection [33]. In HIV infection, endothelial cell (EC) injury and loss of its integrity is due to changes in blood flow and increased circulation of molecules such as IL-6, D-dimer, fibrinogen, C-reactive protein, TNF-α, soluble intercellular adhesion molecule (sICAM), soluble vascular cell adhesion molecule (sVCAM) and endothelial microvesicles [49]. The HIV-1 tat protein is a powerful angiogenic factor, and it binds to cell adhesion molecules (integrins αvβ5, α5β1 and αvβ3, fibronectin and vitronectin) [50]. It also binds to Flk-1/KDR, FLt-1 and MAP kinase, thereby influencing EC growth, invasion and angiogenesis [33,51]. The Tat protein is released from infected cells and causes endothelial cell dysfunction (Figure 1) even when the patient is on ART [52].
The HIV-1 nef protein moves into endothelial cells via nanotube-like conduits mediated by a cytoskeletal rearrangement that affects their permeability and reduces tight junction proteins [53,54,55]. Additionally, nef induces apoptosis via the mitochondrial and Fas/FasL pathways and decreases nitric oxide production and promotes ROS production and consequent oxidative stress and apoptosis [56].
The accessory protein gp120 promotes apoptosis and the production of ET-1, reducing the nitric oxide (NO) synthesized by the NO synthase, hence promoting EC dysfunction [57]. Glycoprotein 120 mediates apoptosis via CXCR4-dependent caspase and p38 MAPK promotion [58]. Post EC injury, an elevated expression of the potent vasoconstrictor ET-1, occurs [57,59]. The HIV-1 matrix protein, p17, interacts with CXCR1 and CXCR2 on endothelial cells to activate the MAPK/ERK and PI3K/Akt signalling pathways, thereby prompting angiogenesis and lymphangiogenesis [60].

4. Vascular Endothelial Growth Factors: Pivotal Angiogenesis Regulators

The vascular endothelial growth factor (VEGF) family are potent regulators of the angiogenic signalling and includes VEGF-A, VEGF-B, VEGF-C, VEGF-D and placental growth factor (PlGF), as well as VEGF receptors: (VEGFR) VEGFR-1, VEGFR-2 and VEGFR-3 [61,62,63]. Specifically, VEGFs regulate cell proliferation, invasion and migration during angiogenesis [64]. Signal transduction of VEGF-A occurs via VEGFR-1 and VEGFR-2, whilst VEGF-B and PlGF activate VEGFR-1 [65], and VEGF-C and VEGF-D show an affinity for VEGFR-3 (Figure 2) [66].

5. Angiogenic Imbalance in Preeclampsia

Several studies have reported a decrease of VEGF-A in maternal circulation with a concomitant increase of soluble fms-like tyrosine kinase (sFlt-1), the soluble form of VEGFR-1 [69]. Gene expression profiling confirms an upregulation of sFlt-1 mRNA in preeclamptic placentae [70]. Notably, an elevation in sFlt-1 and sEng was observed in maternal circulation almost five weeks prior to the onset of PE development [69,70,71,72]. Excess sFlt-1 in PE pregnancies reduces the circulating levels of VEGF and PlGF and antagonises their biological function [71,73], resulting in endothelial dysfunction [74,75]. In view of this, the sFlt-1/PlGF ratio is a highly recommended clinical tool for differentiating between diverse pregnancy-related hypertensive syndromes [76,77,78,79]. Furthermore, soluble endoglin (sEng) and sFlt-1 exert inhibitory effects on VEGF, PlGF and TGF-β signalling, thus activating the endothelial nitric oxide synthase (eNOS) system and vasomotor effects [80]. Similar to sFlt-1, elevated serum sEng levels diminish circulating TGF-β levels and disrupt subsequent signalling [81].
Moreover, our group demonstrated increased circulating levels of sFlt-1 and sEng in PE compared to normotensive pregnancies, irrespective of the HIV status, albeit with a downward trend of circulating sFlt-1 and sEng in the HIV-infected vs. HIV uninfected groups [82,83]. HIV-infected pregnant women develop PE at lower rates compared to HIV uninfected women [8]; however, their risk of PE development is debatable [84]. Govender et al. reported no significant difference between sFlt-1, sEng and PlGF levels in HIV-associated PE [83]. An overview of the role of these angiogenic and antiangiogenic factors in PE development is illustrated in Table 1.
Our group also demonstrated the role of various other angiogenic factors in PE development [16,85,86,87,88,89,90]. We have shown an elevation of TN-C [86], Ang-2 and Eng [87], sVEGFR-1 with concurrent reduction in PECAM-1 and sVEGFR-2 levels [88] and a downregulation in plasma NF-κB and IκB-α expression [90] in PE versus normotensive pregnancies, regardless of HIV status, indicative of their potential role in PE development. However, HIV status did not affect the expression of sTie2 and sHER2 [85] nor sE-selectin [16] in our cohorts, regardless of pregnancy type, which is suggestive that HAART reconstitutes the immune system.
Table 1. Angiogenic and antiangiogenic markers in Preeclampsia development.
Table 1. Angiogenic and antiangiogenic markers in Preeclampsia development.
Angiogenic/
Antiangiogenic/
Lymphangiogenic Factors
Main FindingsReferences
VEGFR-2
  • Downregulation of VEGFR-2 in PE compared to normotensive.
  • Downregulation of VEGF-dependent cell proliferation when the VEGFR-2 signalling pathway was blocked in mice.
[70,91,92,93]
VEGFR-3
  • Decidual VEGFR-3 is significantly downregulated in PE placentae vs. normotensive; indicative of lymphangiogenic inhibition on PE.
[94,95]
VEGF-A
  • Reduced VEGF-A expression in PE compared to normotensive pregnancies.
  • Sequestering and binding of VEGF-A by its soluble receptors.
[71,96,97,98]
VEGF-C
  • Significant increase in VEGF-C expression in preeclampsia compared to normotensives; the pro-lymphangiogenic state evident in PE is linked with oedema and hypertension.
  • No dysregulation in VEGF-C expression noted in HIV infected preeclamptic women vs. uninfected women; may be due to immune reconstitution in response to ARV use.
[99,100]
PlGF
  • Downregulation in PlGF levels noted in PE compared to normotensive pregnancies.
  • Greater PlGF reduction seen in EOPE, due to binding and sequestering of PlGF by the soluble VEGF receptors.
[70,71,96,98]
sFlt-1
  • Circulating sFlt-1 levels upregulated in preeclampsia prior to delivery and is reduced to baseline 48–72 h postdelivery.
  • High sFlt-1 levels correlate inversely with decreased free PlGF levels during preeclampsia.
  • Circulating PlGF levels are downregulated in PE and correlated elevation in total VEGF-A with soluble VEGFR-1.
  • Membrane-bound VEGFR1 is reduced in the placental bed and correlates with poor uteroplacental progression.
  • Upregulation of new sFlt-1 isoforms with differing mRNA lengths in preeclamptic placentas.
  • Circulating sFlt-1-14 identified as a major VEGF inhibitor in PE.
[71,96,101,102,103,104]
sEng
  • Upregulation of sEng in confirmed preeclampsia and in pregnancies prior to symptom onset.
  • Midpregnancy urinary PlGF levels correlates with the consequent preterm PE development.
  • Thrombocytopenia, haemolysis and HELLP was noted as a severe PE outcome in animals treated with both sFLT1 and sEng.
  • sEng upregulated in PE patients with severe complications including placental abruption, HELLP syndrome, eclampsia and IUGR.
[69,70,73,80,105,106,107,108,109]
Our research group has recently established that, in PE, a pre-delivery sFlt-1/PIGF proportion (<181.5) is an encouraging predictor value for excluding the need for >3 slow-acting and/or a rapid-acting antihypertensive drug management in the immediate postpartum period [89]. To our knowledge, we are the first group to provide the cut-off thresholds of pre-delivery sFlt-1/PIGF ratio that can be used to foresee antihypertensive use in the postpartum period in PE and normotensive pregnancy. These results implicate the usefulness of angiogenic factors in examining the manifestation and progression of PE development. Future prospective studies using larger sample sizes that encompass all three trimesters will assist in determining the clinical usefulness of these biomarkers in advancing the clinical management strategies for PE.

6. Angiogenesis and Lymphangiogenesis

Neuropilin-1 (NRP-1), a co-receptor for VEGFR-1 [61], binds to VEGF-2 in the presence of VEGF165, which stimulates phosphatidylinositol 3-kinase (PI3K) and activate protein kinase B (Akt) to promote angiogenesis [110,111]. Activation of VEGFR-3 enables lymphangiogenic regulation, which is also regulated by neuropilin-2 (NRP-2) [65,112]. Moreover, VEGF-A and PlGF binds to NRP-1 and enhances its affinity for VEGFR-2 [113]. A disruption to the equilibrium of these factors will inevitably result in an imbalance in angiogenesis and lymphangiogenesis. Thus, PE may be molecularly defined by a reduction in sVEGFR-3, decreased sVEGFR-2 and increased VEGF-C; however, the involvement of VEGF-B, VEGF-C and VEGF-D in PE development remains debatable.
The immunoexpression of NRP-1 is decreased in the syncytiotrophoblasts in PE compared to normotensive pregnancies, implicating NRP-1 in PE development [114]. Likewise, significantly lower NRP-1 and VEGF levels were observed in both PE and in homocysteine-induced PE mice, which is indicative of endothelial injury and consequent PE development [115]. Moreover, the downregulation of placental NRP-1 expression in foetal growth-restricted pregnancies complicated with absent end-diastolic flow in the umbilical artery correlates with PE development [116]. However, a lack of noticeable differences in NRP-1 and NRP-2 mRNA expression was noted between preeclamptic and normotensive human placentas [117]. Notably, NRP-1 was recently implicated in binding SARS-CoV-2 and showed direct involvement in SARS-CoV-2 entry by potentiating increased viral internalization and infectivity [118]. Investigations linked to NRP-1 and NRP-2 in HIV infection and PE in the HIV-COVID-19 pandemic thus requires urgent and intensive scrutiny.

7. Antiretroviral Therapy and Preeclampsia

The higher prevalence and mortality rates observed among PE cases are associated with ART use in comparison to HIV infection alone [16]. In the event of HIV infection, a pregnant women’s adherence to ART increases the risk of pre-term birth [119,120]. Additionally, hyper-vascularity of the placental terminal villi has been documented as a compensatory response to maternal ARV therapy [119]. Further large-scale studies are urgently required to investigate the effect of the duration of highly active antiretroviral treatment/therapy (HAART) on PE development.
Dysregulated angiogenesis, inflammation and oxidative/nitrosative stress resulting from placental maladaptation promote widespread endothelial dysfunction in PE. HIV accessory and matrix proteins also upregulate these pathways, which intensify endothelial injury. A summary of the impact of ART on various indicators of endothelial dysfunction is shown in Table 2.

8. Renin-Angiotensin-Aldosterone System in HIV Associated Preeclamptic Women on ART

The Renin-Angiotensin-Aldosterone System (RAAS) depends on several receptors for signal transduction of the vasoconstrictor angiotensin II (ANGII), which is liable for increasing blood pressure in PE. Irregular renin-angiotensin stimulation may be linked to the pathophysiology of hypertension and insulin resistance in HIV infection [131], with elevated plasma renin activity resulting from increased RAAS upstream activation [132]. This anomaly is elaborated by the similarity between the structure of HIV-1 protease and that of renin [133]. An increased renin production in immune cells occurs due to the presence of HIV, thereby activating RAAS. Furthermore, renin contributes to HIV replication via the renin signalling cascade and cleavage of the HIV Gag polyproteins [131].
Bouba et al., (2003) reported a significant association between circulating angiotensin (AGT) gene polymorphisms (M235T) and PE [134]. Similarly, Aung et al. (2017) reported a significant increase in the AGT variant (M235T) in PE compared to controls [135]. In contrast to AGT gene polymorphisms, the ATR1 variant (A1166C) showed no association with PE, despite the upregulation reported in both the circulation and the placentae of women with PE compared to controls [135,136]. However, ATR2 (C4599A) showed a significant association with PE in women with a BMI of ≥25 kg/m2 [132]. Moreover, a significant reduction of AT4R in the circulation and the placentae of women with PE was reported [136,137,138], albeit a non-significant association of genetic variant (rs18059) with the pathogenesis of PE [139].
The association of HIV treatment (HAART, protease inhibitors and/or nucleoside reverse transcriptase inhibitors) and RAAS is currently unclear. However, evidence shows that HIV treatment increases the risk of hypertension. A study conducted in Cameroon that was comprised of HIV infected non-pregnant patients on treatment reported the development of hypertension in 36.4% of the population, in comparison to 13.3% HIV positive patients without treatment [140]. Furthermore, the development of PE has been observed in patients on HAART [141,142]. Reports suggest that HIV infected pregnant women are protected against PE but develop the disease on ART initiation [142]. The precise pathogenesis underlying this mechanism and the effect of HAART on RAAS gene expression and RAAS genetic variants is poorly understood and thus requires further investigation. Notably, SARS-CoV-2 infection exploits ACE2 to induce endothelial dysfunction and hypertension, thereby mimicking angiotensin II-mediated PE in severe cases of infection [143]. Upregulated ACE2 in pregnancy is a possible risk factor for SARS-CoV-2 infection and subsequent PE development [16].

9. Cell Death-Neutrophil Extracellular Traps (NETs): Immune Defence

Several studies have implicated a dysregulation of apoptosis on PE development [144,145]. The former study demonstrates increased trophoblast apoptosis in the placental bed of preeclamptic compared to normotensive pregnancies with a concurrent absence of proliferation at term; this enhancement interferes with the process of placentation and eventuates in systemic endothelial damage. In contrast to apoptosis, NETosis is the formation of neutrophil extracellular traps (NETs), a novel neutrophil cell death mechanism [146]. It is triggered in response to pathogenic and pro-inflammatory stimuli, which activates neutrophils to expel their intra-nuclear and intra-cytoplasmic contents in NETs [147]. These NETs are a sticky web of chromatic decorated with histones and granules to trap invading pathogens or pro-inflammatory molecules before eliciting cell death [146,147,148,149]. A hyper-activation or defective homeostatic clearance of NETs result in various adverse outcomes such as SARS-CoV-2, rheumatoid arthritis and PE [150,151,152,153].

10. NETs in HIV-Associated Preeclampsia

In normotensive pregnancies, NETs are expressed in the placental decidua within the conducting villi circulation, surrounding the conducting and exchange villi in the intervillous space [153,154]. In PE, syncytiotrophoblasts (STB) release elevated levels of pro-inflammatory microparticles and interleukin-8 (IL-8), which activate neutrophils to undergo excessive NETosis [153,154]. These NETs contribute to the hyper-inflammatory state observed in PE and exacerbate the underlying PE-compromised endothelial dysfunction [155]. Evidently, elevated levels of maternal cell-free DNA correlate with the severity of PE [156] and further contributes to placental malfunction and foetal rejection by autoimmune activation [157]. NETs are elevated in HIV infection [154,158] and function in trapping and inactivating the virions at a transcriptional level [158,159]. However, as a viral evasion mechanism, HIV stimulates the release of anti-inflammatory IL-10 from dendritic cells to suppress the levels of NETs [158]. Because NETs are individually elevated in PE and HIV infections, it is surprisingly evident that NETs are suppressed in the co-infection of HIV-associated PE; however, this suppression may be attributed to ART usage [154]. This synergy is plausibly attributed to the enhanced expression of dendritic cells in PE [160]—serving as a reservoir for HIV-mediated NETs suppression [154].

11. Recommendations and Therapy Guidelines: HIV Infection and Preeclampsia

In view of the scarcity of available data on drug-to-drug inventions in HIV associated pregnancy, current recommendations for the use of anti-hypertensive drugs in PE are valid. There is no long-term data on the use of anti-hypertensive drugs in HIV infected patients receiving HAART, therefore clinical practice should be based on a case by case scenario. If the hypertension is severe, then anti-hypertensive agents must be used to lower the immediate threat to the mother and baby. Viral replication may be controlled by an antibody response without HAART in pregnancy [161]; however, pregnancy itself does weaken the immune system. Therefore, it is not advisable to temporarily stop ARV usage in pregnant women to prevent PE development as the immune system may not be strong enough to fight viral replication. Further studies in a pregnant cohort are required to investigate antibody neutralization without ARV usage.
From a clinical perspective, it is advisable that patients with HIV infection evaluate their viral load prior to planning a pregnancy, as viral load is not an indication for termination of pregnancy. There is a paucity of data on pregnancy that compares the efficacy of anti-hypertensive drugs in pregnancy; however, Labetalol, an alpha beta blocker may be an appropriate choice in treating HIV-infected PE patients. Notably, no data is currently available to suggest that PE increases the risk of maternal-foetal HIV transmission.

12. Conclusions

This review explores research evidence that may aid in reducing the main direct (preeclampsia) and indirect (HIV infection) causes of global maternal mortality. Both Preeclampsia and HIV infection have opposing immune responses; however, this is offset by the immune reconstitution that occurs with ART usage. Moreover, both conditions culminate in endothelial cell dysfunction (Figure 1). The accessory and matrix proteins of HIV-1 differentially contribute to elevated oxidative stress, apoptosis and the secretion of pro-inflammatory cytokines, cell adhesion molecules and angiogenesis. These events also occur in the hypoxic microenvironment of PE; hence, in the synergy of HIV associated PE, there is an exacerbation of these events, thereby affecting downstream targets. It is possible that HIV-1 has developed mechanisms to escape complement-mediated lysis whilst simultaneously increasing its infectivity of host cells, thereby disturbing foetal defence at the maternal-foetal interface.

Author Contributions

Conceptualization, T.N., J.M. and N.G.; original draft preparation, T.N., J.M., N.G., T.A., N.N., M.M., Y.P., S.S. and O.P.K.; review and editing, T.N., N.G. and J.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest relevant to this manuscript.

References

  1. World Health Organization Maternal Mortality Ratio. Available online: https://www.who.int/news-room/fact-sheets/detail/maternal-mortality (accessed on 16 August 2021).
  2. Say, L.; Chou, D.; Gemmill, A.; Tunçalp, Ö.; Moller, A.B.; Daniels, J.; Gülmezoglu, A.M.; Temmerman, M.; Alkema, L. Global causes of maternal death: A WHO systematic analysis. Lancet Glob. Health 2014, 2, e323–e333. [Google Scholar] [CrossRef] [Green Version]
  3. National Committee for Confidential Enquiry into Maternal Deaths. Saving Mothers Report 2017; Department of Health: Pretoria, South Africa, 2017. [Google Scholar]
  4. UNAIDS Fact Sheet—Global AIDS Update. Available online: https://www.unaids.org/en/resources/fact-sheet (accessed on 21 December 2019).
  5. Calvert, C.; Marston, M.; Slaymaker, E.; Crampin, A.C.; Price, A.J.; Klein, N.; Herbst, K.; Michael, D.; Urassa, M.; Clark, S.J.; et al. Direct maternal deaths attributable to HIV in the era of antiretroviral therapy: Evidence from three population-based HIV cohorts with verbal autopsy. Aids 2020, 34, 1397–1405. [Google Scholar] [CrossRef]
  6. Zaba, B.; Calvert, C.; Marston, M.; Isingo, R.; Nakiyingi-Miiro, J.; Lutalo, T.; Crampin, A.; Robertson, L.; Herbst, K.; Newell, M.L.; et al. Effect of HIV infection on pregnancy-related mortality in sub-Saharan Africa: Secondary analyses of pooled community-based data from the network for Analysing Longitudinal Population-based HIV/AIDS data on Africa (ALPHA). Lancet 2013, 381, 1763–1771. [Google Scholar] [CrossRef] [Green Version]
  7. Clouse, K.; Malope-Kgokong, B.; Bor, J.; Nattey, C.; Mudau, M.; Maskew, M. The South African National HIV Pregnancy Cohort: Evaluating continuity of care among women living with HIV. BMC Public Health 2020, 20, 1662. [Google Scholar] [CrossRef] [PubMed]
  8. Kalumba, V.M.; Moodley, J.; Naidoo, T.D. Is the prevalence of pre-eclampsia affected by HIV/AIDS? A retrospective case-control study. Cardiovasc. J. Afr. 2013, 24, 24–27. [Google Scholar] [CrossRef] [PubMed]
  9. Harris, T.G.; Rabkin, M.; El-Sadr, W.M. Achieving the fourth 90: Healthy aging for people living with HIV. Aids 2018, 32, 1563–1569. [Google Scholar] [CrossRef]
  10. The Antiretroviral Therapy Cohort Collaboration. Survival of HIV-positive patients starting antiretroviral therapy between 1996 and 2013: A collaborative analysis of cohort studies. Lancet HIV 2017, 4, e349–e356. [Google Scholar] [CrossRef] [Green Version]
  11. Harris, K.; Yudin, M.H. HIV Infection in Pregnant Women: A 2020 Update. Prenat. Diagn. 2020, 40, 1715–1721. [Google Scholar] [CrossRef]
  12. Fauci, A.S.; Lane, H.C.; Redfield, R.R. Covid-19—Navigating the Uncharted. N. Engl. J. Med. 2020, 382, 1268–1269. [Google Scholar] [CrossRef]
  13. Villar, J.; Ariff, S.; Gunier, R.B.; Thiruvengadam, R.; Rauch, S.; Kholin, A.; Roggero, P.; Prefumo, F.; do Vale, M.S.; Cardona-Perez, J.A.; et al. Maternal and Neonatal Morbidity and Mortality among Pregnant Women with and without COVID-19 Infection: The INTERCOVID Multinational Cohort Study. JAMA Pediatr. 2021, 175, 817–826. [Google Scholar] [CrossRef]
  14. Wei, S.Q.; Bilodeau-Bertrand, M.; Liu, S.; Auger, N. The impact of COVID-19 on pregnancy outcomes: A systematic review and meta-analysis. Cmaj 2021, 193, E540–E548. [Google Scholar] [CrossRef] [PubMed]
  15. Govender, R.; Moodley, J.; Naicker, T. The COVID-19 Pandemic: An Appraisal of its Impact on Human Immunodeficiency Virus Infection and Pre-Eclampsia. Curr. Hypertens. Rep. 2021, 23, 9. [Google Scholar] [CrossRef] [PubMed]
  16. Naidoo, N.; Moodley, J.; Naicker, T. Maternal endothelial dysfunction in HIV-associated preeclampsia comorbid with COVID-19: A review. Hypertens. Res. 2021, 44, 386–398. [Google Scholar] [CrossRef] [PubMed]
  17. Brown, M.A.; Magee, L.A.; Kenny, L.C.; Karumanchi, S.A.; McCarthy, F.P.; Saito, S.; Hall, D.R.; Warren, C.E.; Adoyi, G.; Ishaku, S. Hypertensive Disorders of Pregnancy: ISSHP Classification, Diagnosis, and Management Recommendations for International Practice. Hypertension 2018, 72, 24–43. [Google Scholar] [CrossRef] [Green Version]
  18. Jena, M.K.; Sharma, N.R.; Petitt, M.; Maulik, D.; Nayak, N.R. Pathogenesis of Preeclampsia and Therapeutic Approaches Targeting the Placenta. Biomolecules 2020, 10, 6. [Google Scholar] [CrossRef] [PubMed]
  19. Liang, X.; Arullampalam, P.; Yang, Z.; Ming, X.F. Hypoxia Enhances Endothelial Intercellular Adhesion Molecule 1 Protein Level through Upregulation of Arginase Type II and Mitochondrial Oxidative Stress. Front. Physiol. 2019, 10, 1003. [Google Scholar] [CrossRef] [Green Version]
  20. Burwick, R.M.; Feinberg, B.B. Complement activation and regulation in preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome. Am. J. Obstet. Gynecol. 2020. [Google Scholar] [CrossRef]
  21. Colucci, F. The immunological code of pregnancy. Science 2019, 365, 862–863. [Google Scholar] [CrossRef]
  22. Lokki, A.I.; Heikkinen-Eloranta, J.K.; Laivuori, H. The Immunogenetic Conundrum of Preeclampsia. Science 2018, 9, 2630. [Google Scholar] [CrossRef] [Green Version]
  23. Hu, W.; Wang, H.; Wang, Z.; Huang, H.; Dong, M. Elevated serum levels of interleukin-15 and interleukin-16 in preeclampsia. J. Reprod. Immunol. 2007, 73, 166–171. [Google Scholar] [CrossRef]
  24. Maharaj, N.R.; Phulukdaree, A.; Nagiah, S.; Ramkaran, P.; Tiloke, C.; Chuturgoon, A.A. Pro-Inflammatory Cytokine Levels in HIV Infected and Uninfected Pregnant Women with and without Preeclampsia. PLoS ONE 2017, 12, e0170063. [Google Scholar] [CrossRef] [Green Version]
  25. Noris, M.; Remuzzi, G. Overview of Complement Activation and Regulation. Semin. Nephrol. 2013, 33, 479–492. [Google Scholar] [CrossRef] [Green Version]
  26. Merle, N.S.; Church, S.E.; Fremeaux-Bacchi, V.; Roumenina, L.T. Complement System Part I—Molecular Mechanisms of Activation and Regulation. Front. Immunol. 2015, 6, 262. [Google Scholar] [CrossRef] [Green Version]
  27. Girardi, G.; Lingo, J.J.; Fleming, S.D.; Regal, J.F. Essential Role of Complement in Pregnancy: From Implantation to Parturition and Beyond. Front. Immunol. 2020, 11, 1681. [Google Scholar] [CrossRef] [PubMed]
  28. Teirilä, L.; Heikkinen-Eloranta, J.; Kotimaa, J.; Meri, S.; Lokki, A.I. Regulation of the complement system and immunological tolerance in pregnancy. Semin. Immunol. 2019, 45, 101337. [Google Scholar] [CrossRef] [PubMed]
  29. Derzsy, Z.; Prohaszka, Z.; Rigo, J., Jr.; Fust, G.; Molvarec, A. Activation of the complement system in normal pregnancy and preeclampsia. Mol. Immunol. 2010, 47, 1500–1506. [Google Scholar] [CrossRef] [PubMed]
  30. Lokki, A.I.; Heikkinen-Eloranta, J.; Jarva, H.; Saisto, T.; Lokki, M.-L.; Laivuori, H.; Meri, S. Complement Activation and Regulation in Preeclamptic Placenta. Front. Immunol. 2014, 5, 312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Lynch, A.M.; Wagner, B.D.; Giclas, P.C.; West, N.A.; Gibbs, R.S.; Holers, V.M. The relationship of longitudinal levels of complement bb during pregnancy with preeclampsia. Am. J. Reprod. Immunol. 2016, 75, 104–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Burwick, R.M.; Fichorova, R.N.; Dawood, H.Y.; Yamamoto, H.S.; Feinberg, B.B. Urinary excretion of C5b-9 in severe preeclampsia: Tipping the balance of complement activation in pregnancy. Hypertension 2013, 62, 1040–1045. [Google Scholar] [CrossRef] [Green Version]
  33. Mazzuca, P.; Caruso, A.; Caccuri, F. HIV-1 infection, microenvironment and endothelial cell dysfunction. New Microbiol. 2016, 39, 163–173. [Google Scholar]
  34. Pillay, Y.; Moodley, J.; Naicker, T. The role of the complement system in HIV infection and preeclampsia. Inflamm. Res. 2019, 68, 459–469. [Google Scholar] [CrossRef]
  35. Yu, Q.; Yu, R.; Qin, X. The good and evil of complement activation in HIV-1 infection. Cell. Mol. Immunol. 2010, 7, 334–340. [Google Scholar] [CrossRef] [Green Version]
  36. Eisen, S.; Dzwonek, A.; Klein, N.J. Mannose-binding lectin in HIV infection. Future Virol. 2008, 3, 225–233. [Google Scholar] [CrossRef] [Green Version]
  37. Kacani, L.; Bánki, Z.; Zwirner, J.; Schennach, H.; Bajtay, Z.; Erdei, A.; Stoiber, H.; Dierich, M.P. C5a and C5a(desArg) enhance the susceptibility of monocyte-derived macrophages to HIV infection. J. Immunol. 2001, 166, 3410–3415. [Google Scholar] [CrossRef] [Green Version]
  38. Bouhlal, H.; Chomont, N.; Haeffner-Cavaillon, N.; Kazatchkine, M.D.; Belec, L.; Hocini, H. Opsonization of HIV-1 by Semen Complement Enhances Infection of Human Epithelial Cells. J. Immunol. 2002, 169, 3301. [Google Scholar] [CrossRef] [Green Version]
  39. Harmon, A.C.; Cornelius, D.C.; Amaral, L.M.; Faulkner, J.L.; Cunningham, M.W., Jr.; Wallace, K.; LaMarca, B. The role of inflammation in the pathology of preeclampsia. Clin. Sci. 2016, 130, 409–419. [Google Scholar] [CrossRef] [Green Version]
  40. Rossheim, A.E.B.; Cunningham, T.D.; Hair, P.S.; Shah, T.; Cunnion, K.M.; Troy, S.B. Effects of Well-Controlled HIV Infection on Complement Activation and Function. J. Acquir. Immune Defic. Syndr. 2016, 73, 20–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Vujkovic-Cvijin, I.; Sortino, O.; Verheij, E.; Wit, F.W.; Kootstra, N.A.; Sellers, B.; Schim van der Loeff, M.; Belkaid, Y.; Reiss, P.; Sereti, I. The Complement Pathway Is Activated in People with Human Immunodeficiency Virus and Is Associated with Non-AIDS Comorbidities. J. Infect. Dis. 2021. [Google Scholar] [CrossRef]
  42. Khan, R.; Maduray, K.; Moodley, J.; Naicker, T. Activation of CD35 and CD55 in HIV associated normal and pre-eclamptic pregnant women. Eur J. Obs. Gynecol. Reprod. Biol. 2016, 204, 51–56. [Google Scholar] [CrossRef] [PubMed]
  43. Perico, L.; Benigni, A.; Casiraghi, F.; Ng, L.F.P.; Renia, L.; Remuzzi, G. Immunity, endothelial injury and complement-induced coagulopathy in COVID-19. Nat. Rev. Nephrol. 2020, 17, 46–64. [Google Scholar] [CrossRef] [PubMed]
  44. Verma, S.; Pillay, P.; Naicker, T.; Moodley, J.; Mackraj, I. Placental hypoxia inducible factor -1α & CHOP immuno-histochemical expression relative to maternal circulatory syncytiotrophoblast micro-vesicles in preeclamptic and normotensive pregnancies. Eur. J. Obstet. Gynecol. Reprod. Biol. 2018, 220, 18–24. [Google Scholar]
  45. Masiá, M.; Padilla, S.; Fernández, M.; Rodríguez, C.; Moreno, A.; Oteo, J.A.; Antela, A.; Moreno, S.; Del Amo, J.; Gutiérrez, F. Oxidative Stress Predicts All-Cause Mortality in HIV-Infected Patients. PLoS ONE 2016, 11, e0153456. [Google Scholar] [CrossRef]
  46. Baliga, R.S.; Chaves, A.A.; Jing, L.; Ayers, L.W.; Bauer, J.A. AIDS-related vasculopathy: Evidence for oxidative and inflammatory pathways in murine and human AIDS. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, H1373–H1380. [Google Scholar] [CrossRef]
  47. Mi, S.; Lee, X.; Li, X.P.; Veldman, G.M.; Finnerty, H.; Racie, L.; LaVallie, E.; Tang, X.Y.; Edouard, P.; Howes, S.; et al. Syncytin is a captive retroviral envelope protein involved in human placental morphogenesis. Nature 2000, 403, 785–789. [Google Scholar] [CrossRef]
  48. Anand, A.R.; Rachel, G.; Parthasarathy, D. HIV Proteins and Endothelial Dysfunction: Implications in Cardiovascular Disease. Front. Cardiovasc. Med. 2018, 5, 185. [Google Scholar] [CrossRef] [PubMed]
  49. Blann, A.D.; Woywodt, A.; Bertolini, F.; Bull, T.M.; Buyon, J.P.; Clancy, R.M.; Haubitz, M.; Hebbel, R.P.; Lip, G.Y.; Mancuso, P.; et al. Circulating endothelial cells. Biomarker of vascular disease. Thromb. Haemost. 2005, 93, 228–235. [Google Scholar] [PubMed]
  50. Ganju, R.K.; Munshi, N.; Nair, B.C.; Liu, Z.Y.; Gill, P.; Groopman, J.E. Human immunodeficiency virus tat modulates the Flk-1/KDR receptor, mitogen-activated protein kinases, and components of focal adhesion in Kaposi’s sarcoma cells. J. Virol. 1998, 72, 6131–6137. [Google Scholar] [CrossRef] [Green Version]
  51. Albini, A.; Soldi, R.; Giunciuglio, D.; Giraudo, E.; Benelli, R.; Primo, L.; Noonan, D.; Salio, M.; Camussi, G.; Rockl, W.; et al. The angiogenesis induced by HIV-1 tat protein is mediated by the Flk-1/KDR receptor on vascular endothelial cells. Nat. Med. 1996, 2, 1371–1375. [Google Scholar] [CrossRef] [PubMed]
  52. Mediouni, S.; Darque, A.; Baillat, G.; Ravaux, I.; Dhiver, C.; Tissot-Dupont, H.; Mokhtari, M.; Moreau, H.; Tamalet, C.; Brunet, C.; et al. Antiretroviral therapy does not block the secretion of the human immunodeficiency virus tat protein. Infect. Disord. Drug Targets 2012, 12, 81–86. [Google Scholar] [CrossRef]
  53. Shiu, C.; Barbier, E.; Di Cello, F.; Choi, H.J.; Stins, M. HIV-1 gp120 as well as alcohol affect blood-brain barrier permeability and stress fiber formation: Involvement of reactive oxygen species. Alcohol. Clin. Exp. Res. 2007, 31, 130–137. [Google Scholar] [CrossRef]
  54. Wang, T.; Green, L.A.; Gupta, S.K.; Kim, C.; Wang, L.; Almodovar, S.; Flores, S.C.; Prudovsky, I.A.; Jolicoeur, P.; Liu, Z.; et al. Transfer of intracellular HIV Nef to endothelium causes endothelial dysfunction. PLoS ONE 2014, 9, e91063. [Google Scholar] [CrossRef] [Green Version]
  55. Kanmogne, G.D.; Primeaux, C.; Grammas, P. HIV-1 gp120 proteins alter tight junction protein expression and brain endothelial cell permeability: Implications for the pathogenesis of HIV-associated dementia. J. Neuropathol. Exp. Neurol. 2005, 64, 498–505. [Google Scholar] [CrossRef] [Green Version]
  56. Acheampong, E.A.; Parveen, Z.; Muthoga, L.W.; Kalayeh, M.; Mukhtar, M.; Pomerantz, R.J. Human Immunodeficiency virus type 1 Nef potently induces apoptosis in primary human brain microvascular endothelial cells via the activation of caspases. J. Virol. 2005, 79, 4257–4269. [Google Scholar] [CrossRef] [Green Version]
  57. Jiang, J.; Fu, W.; Wang, X.; Lin, P.H.; Yao, Q.; Chen, C. HIV gp120 induces endothelial dysfunction in tumour necrosis factor-alpha-activated porcine and human endothelial cells. Cardiovasc. Res. 2010, 87, 366–374. [Google Scholar] [CrossRef] [Green Version]
  58. Huang, M.B.; Khan, M.; Garcia-Barrio, M.; Powell, M.; Bond, V.C. Apoptotic effects in primary human umbilical vein endothelial cell cultures caused by exposure to virion-associated and cell membrane-associated HIV-1 gp120. J. Acquir. Immune Defic. Syndr. 2001, 27, 213–221. [Google Scholar] [CrossRef] [PubMed]
  59. Didier, N.; Banks, W.A.; Créminon, C.; Dereuddre-Bosquet, N.; Mabondzo, A. HIV-1-induced production of endothelin-1 in an in vitro model of the human blood-brain barrier. Neuroreport 2002, 13, 1179–1183. [Google Scholar] [CrossRef] [PubMed]
  60. Caccuri, F.; Giagulli, C.; Bugatti, A.; Benetti, A.; Alessandri, G.; Ribatti, D.; Marsico, S.; Apostoli, P.; Slevin, M.A.; Rusnati, M.; et al. HIV-1 matrix protein p17 promotes angiogenesis via chemokine receptors CXCR1 and CXCR2. Proc. Natl. Acad. Sci. USA 2012, 109, 14580–14585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Holmes, D.I.R.; Zachary, I. The vascular endothelial growth factor (VEGF) family: Angiogenic factors in health and disease. Genome Biol. 2005, 6, 209–209.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Karaman, S.; Leppänen, V.-M.; Alitalo, K. Vascular endothelial growth factor signaling in development and disease. Development 2018, 145, 151019. [Google Scholar] [CrossRef] [Green Version]
  63. Roy, H.; Bhardwaj, S.; Ylä-Herttuala, S. Biology of vascular endothelial growth factors. FEBS Lett. 2006, 580, 2879–2887. [Google Scholar] [CrossRef] [Green Version]
  64. Rana, S.; Powe, C.E.; Salahuddin, S.; Verlohren, S.; Perschel, F.H.; Levine, R.J.; Lim, K.-H.; Wenger, J.B.; Thadhani, R.; Karumanchi, S.A. Angiogenic factors and the risk of adverse outcomes in women with suspected preeclampsia. Circulation 2012, 125, 911–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Pajusola, K.; Aprelikova, O.; Korhonen, J.; Kaipainen, A.; Pertovaara, L.; Alitalo, R.; Alitalo, K. FLT4 receptor tyrosine kinase contains seven immunoglobulin-like loops and is expressed in multiple human tissues and cell lines. Cancer Res. 1992, 52, 5738–5743. [Google Scholar] [PubMed]
  66. Oplawski, M.; Dziobek, K.; Zmarzły, N.; Grabarek, B.; Halski, T.; Januszyk, P.; Kuś-Kierach, A.; Adwent, I.; Dąbruś, D.; Kiełbasiński, K.; et al. Expression Profile of VEGF-C, VEGF-D, and VEGFR-3 in Different Grades of Endometrial Cancer. Curr Pharm. Biotechnol. 2019, 20, 1004–1010. [Google Scholar] [CrossRef]
  67. Padayachee, S.; Moodley, J.; Naicker, T. A Review of Angiogenic Imbalance in HIV-Infected Hypertensive Disorders of Pregnancy. Curr. Hypertens. Rep. 2019, 21, 69. [Google Scholar] [CrossRef]
  68. Pandey, A.K.; Singhi, E.K.; Arroyo, J.P.; Ikizler, T.A.; Gould, E.R.; Brown, J.; Beckman, J.A.; Harrison, D.G.; Moslehi, J. Mechanisms of VEGF (Vascular Endothelial Growth Factor) Inhibitor-Associated Hypertension and Vascular Disease. Hypertension 2018, 71, e1–e8. [Google Scholar] [CrossRef] [PubMed]
  69. Levine, R.J.; Lam, C.; Qian, C.; Yu, K.F.; Maynard, S.E.; Sachs, B.P.; Sibai, B.M.; Epstein, F.H.; Romero, R.; Thadhani, R. Soluble endoglin and other circulating antiangiogenic factors in preeclampsia. N. Engl. J. Med. 2006, 355, 992–1005. [Google Scholar] [CrossRef]
  70. Maynard, S.E.; Min, J.Y.; Merchan, J.; Lim, K.H.; Li, J.; Mondal, S.; Libermann, T.A.; Morgan, J.P.; Sellke, F.W.; Stillman, I.E.; et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J. Clin. Investig. 2003, 111, 649–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Levine, R.J.; Maynard, S.E.; Qian, C.; Lim, K.H.; England, L.J.; Yu, K.F.; Schisterman, E.F.; Thadhani, R.; Sachs, B.P.; Epstein, F.H.; et al. Circulating angiogenic factors and the risk of preeclampsia. N. Engl. J. Med. 2004, 350, 672–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ohkuchi, A.; Hirashima, C.; Takahashi, K.; Suzuki, H.; Matsubara, S.; Suzuki, M. Onset threshold of the plasma levels of soluble fms-like tyrosine kinase 1/placental growth factor ratio for predicting the imminent onset of preeclampsia within 4 weeks after blood sampling at 19–31 weeks of gestation. Hypertens. Res. Off. J. Jpn. Soc. Hypertens. 2013, 36, 1073–1080. [Google Scholar] [CrossRef]
  73. Chaiworapongsa, T.; Romero, R.; Kim, Y.M.; Kim, G.J.; Kim, M.R.; Espinoza, J.; Bujold, E.; Gonçalves, L.; Gomez, R.; Edwin, S.; et al. Plasma soluble vascular endothelial growth factor receptor-1 concentration is elevated prior to the clinical diagnosis of pre-eclampsia. J. Matern. Fetal Neonatal Med. 2005, 17, 3–18. [Google Scholar] [CrossRef]
  74. Eremina, V.; Jefferson, J.A.; Kowalewska, J.; Hochster, H.; Haas, M.; Weisstuch, J.; Richardson, C.; Kopp, J.B.; Kabir, M.G.; Backx, P.H.; et al. VEGF inhibition and renal thrombotic microangiopathy. N. Engl. J. Med. 2008, 358, 1129–1136. [Google Scholar] [CrossRef]
  75. Patel, T.V.; Morgan, J.A.; Demetri, G.D.; George, S.; Maki, R.G.; Quigley, M.; Humphreys, B.D. A preeclampsia-like syndrome characterized by reversible hypertension and proteinuria induced by the multitargeted kinase inhibitors sunitinib and sorafenib. J. Natl. Cancer Inst. 2008, 100, 282–284. [Google Scholar] [CrossRef] [Green Version]
  76. Verlohren, S.; Herraiz, I.; Lapaire, O.; Schlembach, D.; Moertl, M.; Zeisler, H.; Calda, P.; Holzgreve, W.; Galindo, A.; Engels, T.; et al. The sFlt-1/PlGF ratio in different types of hypertensive pregnancy disorders and its prognostic potential in preeclamptic patients. Am. J. Obs. Gynecol. 2012, 206, e1–e8. [Google Scholar] [CrossRef]
  77. Stepan, H.; Hund, M.; Andraczek, T. Combining Biomarkers to Predict Pregnancy Complications and Redefine Preeclampsia: The Angiogenic-Placental Syndrome. Hypertension 2020, 75, 918–926. [Google Scholar] [CrossRef]
  78. Schrey-Petersen, S.; Stepan, H. Anti-angiogenesis and Preeclampsia in 2016. Curr. Hypertens. Rep. 2017, 19, 6. [Google Scholar] [CrossRef]
  79. Zeisler, H.; Llurba, E.; Chantraine, F.; Vatish, M.; Staff, A.C.; Sennström, M.; Olovsson, M.; Brennecke, S.P.; Stepan, H.; Allegranza, D. Predictive value of the sFlt-1: PlGF ratio in women with suspected preeclampsia. N. Engl. J. Med. 2016, 374, 13–22. [Google Scholar] [CrossRef] [PubMed]
  80. Venkatesha, S.; Toporsian, M.; Lam, C.; Hanai, J.; Mammoto, T.; Kim, Y.M.; Bdolah, Y.; Lim, K.H.; Yuan, H.T.; Libermann, T.A.; et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat. Med. 2006, 12, 642–649. [Google Scholar] [CrossRef] [PubMed]
  81. Maharaj, A.S.; Walshe, T.E.; Saint-Geniez, M.; Venkatesha, S.; Maldonado, A.E.; Himes, N.C.; Matharu, K.S.; Karumanchi, S.A.; D’Amore, P.A. VEGF and TGF-beta are required for the maintenance of the choroid plexus and ependyma. J. Exp. Med. 2008, 205, 491–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Govender, N.; Naicker, T.; Moodley, J. Maternal imbalance between pro-angiogenic and anti-angiogenic factors in HIV-infected women with pre-eclampsia. Cardiovasc. J. Afr. 2013, 24, 174–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Govender, N.; Naicker, T.; Rajakumar, A.; Moodley, J. Soluble fms-like tyrosine kinase-1 and soluble endoglin in HIV-associated preeclampsia. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 170, 100–105. [Google Scholar] [CrossRef] [PubMed]
  84. Sansone, M.; Sarno, L.; Saccone, G.; Berghella, V.; Maruotti, G.M.; Migliucci, A.; Capone, A.; Martinelli, P. Risk of Preeclampsia in Human Immunodeficiency Virus-Infected Pregnant Women. Obstet. Gynecol. 2016, 127, 1027–1032. [Google Scholar] [CrossRef]
  85. Mazibuko, M.; Moodley, J.; Naicker, T. Dysregulation of circulating sTie2 and sHER2 in HIV-infected women with preeclampsia. Hypertens. Pregnancy 2019, 38, 89–95. [Google Scholar] [CrossRef] [PubMed]
  86. Mafuika, S.N.; Naicker, T. The role of Tenascin-C in HIV associated pre-eclampsia. Pregnancy Hypertens. 2021, 25, 156–160. [Google Scholar] [CrossRef] [PubMed]
  87. Mbhele, N.; Moodley, J.; Naicker, T. Role of angiopoietin-2, endoglin, and placental growth factor in HIV-associated preeclampsia. Hypertens. Pregnancy 2017, 36, 240–246. [Google Scholar] [CrossRef]
  88. Thakoordeen, S.; Moodley, J.; Naicker, T. Serum levels of platelet endothelial cell adhesion molecule-1 (PECAM-1) and soluble vascular endothelial growth factor receptor (sVEGFR)-1 and -2 in HIV associated preeclampsia. Hypertens. Pregnancy 2017, 36, 168–174. [Google Scholar] [CrossRef]
  89. Ngene, N.C.; Moodley, J.; Naicker, T. The performance of pre-delivery serum concentrations of angiogenic factors in predicting postpartum antihypertensive drug therapy following abdominal delivery in severe preeclampsia and normotensive pregnancy. PLoS ONE 2019, 14, e0215807. [Google Scholar]
  90. Zozo, B.; Govender, N.; Moodley, J.; Naicker, T. Expression of plasma nuclear factor-kappa B cells (NF-κB) and Inhibitory subunit kappa B alpha (IκB-α) in HIV-associated pre-eclampsia. Hypertens. Pregnancy 2021, 40, 15–20. [Google Scholar] [CrossRef] [PubMed]
  91. Helske, S.; Vuorela, P.; Carpén, O.; Hornig, C.; Weich, H.; Halmesmäki, E. Expression of vascular endothelial growth factor receptors 1, 2 and 3 in placentas from normal and complicated pregnancies. Mol. Hum. Reprod. 2001, 7, 205–210. [Google Scholar] [CrossRef] [Green Version]
  92. Munaut, C.; Lorquet, S.; Pequeux, C.; Coulon, C.; Le Goarant, J.; Chantraine, F.; Noël, A.; Goffin, F.; Tsatsaris, V.; Subtil, D. Differential expression of Vegfr-2 and its soluble form in preeclampsia. PLoS ONE 2012, 7, 3. [Google Scholar] [CrossRef] [Green Version]
  93. Takahashi, T.; Yamaguchi, S.; Chida, K.; Shibuya, M. A single autophosphorylation site on KDR/Flk-1 is essential for VEGF-A-dependent activation of PLC-gamma and DNA synthesis in vascular endothelial cells. EMBO J. 2001, 20, 2768–2778. [Google Scholar] [CrossRef] [Green Version]
  94. Liu, H.; Li, Y.; Zhang, J.; Rao, M.; Liang, H.; Liu, G. The defect of both angiogenesis and lymphangiogenesis is involved in preeclampsia. Placenta 2015, 36, 279–286. [Google Scholar] [CrossRef]
  95. Marini, M.; Vichi, D.; Toscano, A.; Zappoli Thyrion, G.D.; Parretti, E.; Mello, G.; Gheri, G.; Pacini, A.; Sgambati, E. Expression of vascular endothelial growth factor receptor types 1, 2 and 3 in placenta from pregnancies complicated by hypertensive disorders. Reprod. Fertil. Dev. 2007, 19, 641–651. [Google Scholar] [CrossRef] [Green Version]
  96. Tsatsaris, V.; Goffin, F.; Munaut, C.; Brichant, J.F.; Pignon, M.R.; Noel, A.; Schaaps, J.P.; Cabrol, D.; Frankenne, F.; Foidart, J.M. Overexpression of the soluble vascular endothelial growth factor receptor in preeclamptic patients: Pathophysiological consequences. J. Clin. Endocrinol. Metab. 2003, 88, 5555–5563. [Google Scholar] [CrossRef] [Green Version]
  97. Zhou, Q.; Liu, H.; Qiao, F.; Wu, Y.; Xu, J. VEGF deficit is involved in endothelium dysfunction in preeclampsia. J. Huazhong Univ. Sci. Technol. Med. Sci. 2010, 30, 370–374. [Google Scholar] [CrossRef] [PubMed]
  98. Lecarpentier, E.; Tsatsaris, V. Angiogenic balance (sFlt-1/PlGF) and preeclampsia. Ann. Endocrinol. 2016, 77, 97–100. [Google Scholar] [CrossRef] [PubMed]
  99. Lely, A.T.; Salahuddin, S.; Holwerda, K.M.; Karumanchi, S.A.; Rana, S. Circulating lymphangiogenic factors in preeclampsia. Hypertens. Pregnancy 2013, 32, 42–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Shange, G.P.; Moodley, J.; Naicker, T. Effect of vascular endothelial growth factors A, C, and D in HIV-associated pre-eclampsia. Hypertens. Pregnancy 2017, 36, 196–203. [Google Scholar] [CrossRef] [PubMed]
  101. Risau, W. Development and differentiation of endothelium. Kidney Int. Suppl. 1998, 67, S3–S6. [Google Scholar] [CrossRef] [Green Version]
  102. Taylor, R.N.; Grimwood, J.; Taylor, R.S.; McMaster, M.T.; Fisher, S.J.; North, R.A. Longitudinal serum concentrations of placental growth factor: Evidence for abnormal placental angiogenesis in pathologic pregnancies. Am. J. Obs. Gynecol. 2003, 188, 177–182. [Google Scholar] [CrossRef]
  103. Sela, S.; Itin, A.; Natanson-Yaron, S.; Greenfield, C.; Goldman-Wohl, D.; Yagel, S.; Keshet, E. A novel human-specific soluble vascular endothelial growth factor receptor 1: Cell-type-specific splicing and implications to vascular endothelial growth factor homeostasis and preeclampsia. Circ. Res. 2008, 102, 1566–1574. [Google Scholar] [CrossRef] [Green Version]
  104. Thomas, C.P.; Andrews, J.I.; Liu, K.Z. Intronic polyadenylation signal sequences and alternate splicing generate human soluble Flt1 variants and regulate the abundance of soluble Flt1 in the placenta. FASEB J. 2007, 21, 3885–3895. [Google Scholar] [CrossRef]
  105. Chaiworapongsa, T.; Romero, R.; Espinoza, J.; Bujold, E.; Mee Kim, Y.; Gonçalves, L.F.; Gomez, R.; Edwin, S. Evidence supporting a role for blockade of the vascular endothelial growth factor system in the pathophysiology of preeclampsia. Young Investigator Award. Am. J. Obs. Gynecol. 2004, 190, 1541–1547. [Google Scholar] [CrossRef] [PubMed]
  106. Levine, R.J.; Thadhani, R.; Qian, C.; Lam, C.; Lim, K.H.; Yu, K.F.; Blink, A.L.; Sachs, B.P.; Epstein, F.H.; Sibai, B.M.; et al. Urinary placental growth factor and risk of preeclampsia. JAMA 2005, 293, 77–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Wallace, K.; Morris, R.; Kyle, P.B.; Cornelius, D.; Darby, M.; Scott, J.; Moseley, J.; Chatman, K.; Lamarca, B. Hypertension, inflammation and T lymphocytes are increased in a rat model of HELLP syndrome. Hypertens. Pregnancy 2014, 33, 41–54. [Google Scholar] [CrossRef] [PubMed]
  108. Noori, M.; Donald, A.E.; Angelakopoulou, A.; Hingorani, A.D.; Williams, D.J. Prospective study of placental angiogenic factors and maternal vascular function before and after preeclampsia and gestational hypertension. Circulation 2010, 122, 478–487. [Google Scholar] [CrossRef] [Green Version]
  109. Schmella, M.J.; Assibey-Mensah, V.; Parks, W.T.; Roberts, J.M.; Jeyabalan, A.; Hubel, C.A.; Catov, J.M. Plasma concentrations of soluble endoglin in the maternal circulation are associated with maternal vascular malperfusion lesions in the placenta of women with preeclampsia. Placenta 2019, 78, 29–35. [Google Scholar] [CrossRef]
  110. Zachary, I.; Gliki, G. Signaling transduction mechanisms mediating biological actions of the vascular endothelial growth factor family. Cardiovasc Res 2001, 49, 568–581. [Google Scholar] [CrossRef] [Green Version]
  111. Chen, Y.-L.; Hong, T.-M.; Yang, P.-C. Neuropilin 1 and Lung Cancer Progression: Potential Role of Targeting Neuropilin 1 as an Antitumor Strategy. Clin. Cancer Res. 2007, 13, 4759–4768. [Google Scholar]
  112. Apte, R.S.; Chen, D.S.; Ferrara, N. VEGF in Signaling and Disease: Beyond Discovery and Development. Cell 2019, 176, 1248–1264. [Google Scholar] [CrossRef] [Green Version]
  113. Soker, S.; Takashima, S.; Miao, H.Q.; Neufeld, G.; Klagsbrun, M. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell 1998, 92, 735–745. [Google Scholar] [CrossRef] [Green Version]
  114. Arad, A.; Nammouz, S.; Nov, Y.; Ohel, G.; Bejar, J.; Vadasz, Z. The Expression of Neuropilin-1 in Human Placentas from Normal and Preeclamptic Pregnancies. Int. J. Gynecol. Pathol. 2017, 36, 42–49. [Google Scholar] [CrossRef]
  115. Xu, X.; Yang, X.Y.; He, B.W.; Yang, W.J.; Cheng, W.W. Placental NRP1 and VEGF expression in pre-eclamptic women and in a homocysteine-treated mouse model of pre-eclampsia. Eur J. Obs. Gynecol. Reprod. Biol. 2016, 196, 69–75. [Google Scholar] [CrossRef]
  116. Maulik, D.; De, A.; Ragolia, L.; Evans, J.; Grigoryev, D.; Lankachandra, K.; Mundy, D.; Muscat, J.; Gerkovich, M.M.; Ye, S.Q. Down-regulation of placental neuropilin-1 in fetal growth restriction. Am. J. Obs. Gynecol. 2016, 214, e1–e279. [Google Scholar] [CrossRef] [Green Version]
  117. Chung, J.Y.; Song, Y.; Wang, Y.; Mazagness, R.R.; Zheng, J. Differential expression of vascular endothelial growth factor (VEGF), endocrine gland derived-VEGF, and VEGF receptors in human placentas from normal and preeclamptic pregnancies. J. Clin. Endocrinol. Metab. 2004, 89, 2484–2490. [Google Scholar] [CrossRef] [Green Version]
  118. Coutard, B.; Valle, C.; de Lamballerie, X.; Canard, B.; Seidah, N.G.; Decroly, E. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antivir. Res. 2020, 176, 104742. [Google Scholar] [CrossRef]
  119. Obimbo, M.M.; Zhou, Y.; McMaster, M.T.; Cohen, C.R.; Qureshi, Z.; Ong’ech, J.; Ogeng’o, J.A.; Fisher, S.J. Placental structure in preterm birth among HIV-positive versus HIV-negative women in Kenya. J. Acquir. Immune Defic. Syndr. 2019, 80, 94. [Google Scholar] [CrossRef] [PubMed]
  120. Machado, E.S.; Krauss, M.R.; Megazzini, K.; Coutinho, C.M.; Kreitchmann, R.; Melo, V.H.; Pilotto, J.H.; Ceriotto, M.; Hofer, C.B.; Siberry, G.K. Hypertension, preeclampsia and eclampsia among HIV-infected pregnant women from Latin America and Caribbean countries. J. Infect. 2014, 68, 572–580. [Google Scholar] [CrossRef] [Green Version]
  121. Hernández, S.; Catalán-García, M.; Morén, C.; García-Otero, L.; López, M.; Guitart-Mampel, M.; Milisenda, J.; Coll, O.; Cardellach, F.; Gratacós, E. Placental mitochondrial toxicity, oxidative stress, apoptosis, and adverse perinatal outcomes in HIV pregnancies under antiretroviral treatment containing zidovudine. JAIDS J. Acquir. Immune Defic. Syndr. 2017, 75, e113–e119. [Google Scholar] [CrossRef]
  122. Song, L.; Ding, S.; Ge, Z.; Zhu, X.; Qiu, C.; Wang, Y.; Lai, E.; Yang, W.; Sun, Y.; Chow, S.A.; et al. Nucleoside/nucleotide reverse transcriptase inhibitors attenuate angiogenesis and lymphangiogenesis by impairing receptor tyrosine kinases signalling in endothelial cells. Br. J. Pharmacol. 2018, 175, 1241–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Autran, B.; Carcelaint, G.; Li, T.S.; Gorochov, G.; Blanc, C.; Renaud, M.; Durali, M.; Mathez, D.; Calvez, V.; Leibowitch, J.; et al. Restoration of the immune system with anti-retroviral therapy. Immunol. Lett. 1999, 66, 207–211. [Google Scholar] [CrossRef]
  124. Powis, K.M.; Shapiro, R.L. Protease Inhibitors and Adverse Birth Outcomes: Is Progesterone the Missing Piece to the Puzzle? Oxford University Press: Oxford, UK, 2015. [Google Scholar]
  125. Kala, S.; Dunk, C.; Acosta, S.; Serghides, L. Periconceptional exposure to lopinavir, but not darunavir, impairs decidualization: A potential mechanism leading to poor birth outcomes in HIV-positive pregnancies. Hum. Reprod. 2020, 35, 1781–1796. [Google Scholar] [CrossRef] [PubMed]
  126. Francisci, D.; Giannini, S.; Baldelli, F.; Leone, M.; Belfiori, B.; Guglielmini, G.; Malincarne, L.; Gresele, P. HIV type 1 infection, and not short-term HAART, induces endothelial dysfunction. Aids 2009, 23, 589–596. [Google Scholar] [CrossRef] [PubMed]
  127. Powis, K.M.; McElrath, T.F.; Hughes, M.D.; Ogwu, A.; Souda, S.; Datwyler, S.A.; von Widenfelt, E.; Moyo, S.; Nádas, M.; Makhema, J.; et al. High viral load and elevated angiogenic markers associated with increased risk of preeclampsia among women initiating highly active antiretroviral therapy in pregnancy in the Mma Bana study, Botswana. J. Acquir. Immune Defic. Syndr. 2013, 62, 517–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Mattar, R.; Amed, A.M.; Lindsey, P.C.; Sass, N.; Daher, S. Preeclampsia and HIV infection. Eur. J. Obstet. Gynecol. Reprod. Biol. 2004, 117, 240–241. [Google Scholar] [CrossRef]
  129. Sgadari, C.; Barillari, G.; Toschi, E.; Carlei, D.; Bacigalupo, I.; Baccarini, S.; Palladino, C.; Leone, P.; Bugarini, R.; Malavasi, L.; et al. HIV protease inhibitors are potent anti-angiogenic molecules and promote regression of Kaposi sarcoma. Nat. Med. 2002, 8, 225–232. [Google Scholar] [CrossRef]
  130. Govender, N.; Moodley, J.; Gathiram, P.; Naicker, T. Soluble fms-like tyrosine kinase-1 in HIV infected pre-eclamptic South African Black women. Placenta 2014, 35, 618–624. [Google Scholar] [CrossRef]
  131. Srinivasa, S.; Fitch, K.V.; Wong, K.; Torriani, M.; Mayhew, C.; Stanley, T.; Lo, J.; Adler, G.K.; Grinspoon, S.K. RAAS activation is associated with visceral adiposity and insulin resistance among HIV-infected patients. J. Clin. Endocrinol. Metab. 2015, 100, 2873–2882. [Google Scholar] [CrossRef]
  132. Chandel, N.; Ayasolla, K.; Lan, X.; Rai, P.; Mikulak, J.; Husain, M.; Malhotra, A.; McGowan, J.; Singhal, P.C. Renin modulates HIV replication in T cells. J. Leukoc. Biol. 2014, 96, 601–609. [Google Scholar] [CrossRef] [Green Version]
  133. Tzoupis, H.; Leonis, G.; Megariotis, G.; Supuran, C.T.; Mavromoustakos, T.; Papadopoulos, M.G. Dual inhibitors for aspartic proteases HIV-1 PR and renin: Advancements in AIDS–hypertension–diabetes linkage via molecular dynamics, inhibition assays, and binding free energy calculations. J. Med. Chem. 2012, 55, 5784–5796. [Google Scholar] [CrossRef] [PubMed]
  134. Bouba, I.; Makrydimas, G.; Kalaitzidis, R.; Lolis, D.E.; Siamopoulos, K.C.; Georgiou, I. Interaction between the polymorphisms of the renin–angiotensin system in preeclampsia. Eur. J. Obstet. Gynecol. Reprod. Biol. 2003, 110, 8–11. [Google Scholar] [CrossRef] [Green Version]
  135. Aung, M.; Konoshita, T.; Moodley, J.; Gathiram, P. Association of gene polymorphisms of four components of renin-angiotensin-aldosterone system and preeclampsia in South African black women. Eur. J. Obstet. Gynecol. Reprod. Biol. 2017, 215, 180–187. [Google Scholar] [CrossRef]
  136. Williams, P.; Mistry, H.; Innes, B.; Bulmer, J.; Pipkin, F.B. Expression of AT1R, AT2R and AT4R and their roles in extravillous trophoblast invasion in the human. Placenta 2010, 31, 448–455. [Google Scholar] [CrossRef]
  137. Zhou, A. Renin Angiotensin System Polymorphisms and Pregnancy Complications. Ph.D. Thesis, University of Adelaide, Adelaide, Australia, 2012. [Google Scholar]
  138. Khaliq, O.P.; Konoshita, T.; Moodley, J.; Naicker, T. Soluble angiotensin IV receptor levels in preeclampsia: Is there a variation? J. Matern. Fetal. Neonatal. Med. 2020, 1–6. [Google Scholar] [CrossRef]
  139. Khaliq, O.P.; Konoshita, T.; Moodley, J.; Naicker, T. The role of LNPEP and ANPEP gene polymorphisms in the pathogenesis of pre-eclampsia. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 252, 160–165. [Google Scholar] [CrossRef] [PubMed]
  140. Pangmekeh, P.J.; Awolu, M.M.; Gustave, S.; Gladys, T.; Cumber, S.N. Association between highly active antiretroviral therapy (HAART) and hypertension in persons living with HIV/AIDS at the Bamenda regional hospital, Cameroon. Pan Afr. Med. J. 2019, 33, 87. [Google Scholar] [CrossRef] [PubMed]
  141. Santos, R.A.S.; Sampaio, W.O.; Alzamora, A.C.; Motta-Santos, D.; Alenina, N.; Bader, M.; Campagnole-Santos, M.J. The ACE2/angiotensin-(1–7)/MAS axis of the renin-angiotensin system: Focus on angiotensin-(1–7). Physiol. Rev. 2017. [Google Scholar] [CrossRef] [Green Version]
  142. Woldesenbet, S.A.; Kufa, T.; Barron, P.; Ayalew, K.; Cheyip, M.; Chirombo, B.C.; Lombard, C.; Manda, S.; Pillay, Y.; Puren, A.J. Assessment of readiness to transition from antenatal HIV surveillance surveys to PMTCT programme data-based HIV surveillance in South Africa: The 2017 Antenatal Sentinel HIV Survey. Int. J. Infect. Dis. 2020, 91, 50–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Abel, T.; Moodley, J.; Naicker, T. The Involvement of MicroRNAs in SARS-CoV-2 Infection Comorbid with HIV-Associated Preeclampsia. Curr. Hypertens. Rep. 2021, 23, 20. [Google Scholar] [CrossRef] [PubMed]
  144. Naicker, T.; Dorsamy, E.; Ramsuran, D.; Burton, G.J.; Moodley, J. The role of apoptosis on trophoblast cell invasion in the placental bed of normotensive and preeclamptic pregnancies. Hypertens. Pregnancy 2013, 32, 245–256. [Google Scholar] [CrossRef] [PubMed]
  145. Raguema, N.; Moustadraf, S.; Bertagnolli, M. Immune and Apoptosis Mechanisms Regulating Placental Development and Vascularization in Preeclampsia. Front. Physiol. 2020, 11, 98. [Google Scholar] [CrossRef]
  146. Fuchs, T.A.; Abed, U.; Goosmann, C.; Hurwitz, R.; Schulze, I.; Wahn, V.; Weinrauch, Y.; Brinkmann, V.; Zychlinsky, A. Novel cell death program leads to neutrophil extracellular traps. J. Cell Biol. 2007, 176, 231–241. [Google Scholar] [CrossRef]
  147. Brinkmann, V.; Zychlinsky, A. Neutrophil extracellular traps: Is immunity the second function of chromatin? J. Cell Biol. 2012, 198, 773–783. [Google Scholar] [CrossRef] [Green Version]
  148. Brinkmann, V. Neutrophil Extracellular Traps in the Second Decade. J. Innate Immun. 2018, 10, 414–421. [Google Scholar] [CrossRef]
  149. Moodley, M.; Moodley, J.; Naicker, T. The Role of Neutrophils and Their Extracellular Traps in the Synergy of Pre-eclampsia and HIV Infection. Curr. Hypertens. Rep. 2020, 22, 1–9. [Google Scholar] [CrossRef]
  150. Mutua, V.; Gershwin, L.J. A review of neutrophil extracellular traps (NETs) in disease: Potential anti-NETs therapeutics. Clin. Rev. Allergy Immunol. 2020, 1–18. [Google Scholar] [CrossRef]
  151. Thierry, A.; Benoit, R. NETs by-products and extracellular DNA may play a key role in COVID-19 pathogenesis: Incidence on patient monitoring and therapy. Preprints 2020. [Google Scholar] [CrossRef] [Green Version]
  152. Song, W.; Ye, J.; Pan, N.; Tan, C.; Herrmann, M. Neutrophil Extracellular Traps Tied to Rheumatoid Arthritis: Points to Ponder. Front. Immunol. 2021, 11, 3668. [Google Scholar] [CrossRef]
  153. Gupta, A.K.; Hasler, P.; Holzgreve, W.; Gebhardt, S.; Hahn, S. Induction of neutrophil extracellular DNA lattices by placental microparticles and IL-8 and their presence in preeclampsia. Hum. Immunol. 2005, 66, 1146–1154. [Google Scholar] [CrossRef] [PubMed]
  154. Moodley, M.; Moodley, J.; Naicker, T. Neutrophil extracellular traps: The synergy source in the placentae of HIV infected women with pre-eclampsia. Pregnancy Hypertens. 2020, 20, 69–74. [Google Scholar] [CrossRef] [PubMed]
  155. Gupta, A.K.; Joshi, M.B.; Philippova, M.; Erne, P.; Hasler, P.; Hahn, S.; Resink, T.J. Activated endothelial cells induce neutrophil extracellular traps and are susceptible to NETosis--mediated cell death. FEBS Lett. 2010, 584, 3193–3197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Zhong, X.Y.; Laivuori, H.; Livingston, J.C.; Ylikorkala, O.; Sibai, B.M.; Holzgreve, W.; Hahn, S. Elevation of both maternal and fetal extracellular circulating deoxyribonucleic acid concentrations in the plasma of pregnant women with preeclampsia. Am. J. Obstet. Gynecol. 2001, 184, 414–419. [Google Scholar] [CrossRef]
  157. Konečná, B.; Lauková, L.; Vlková, B. Immune activation by nucleic acids: A role in pregnancy complications. Scand. J. Immunol. 2018, 87, e12651. [Google Scholar] [CrossRef] [PubMed]
  158. Saitoh, T.; Komano, J.; Saitoh, Y.; Misawa, T.; Takahama, M.; Kozaki, T.; Uehata, T.; Iwasaki, H.; Omori, H.; Yamaoka, S. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microbe 2012, 12, 109–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Kozlowski, H.N.; Lai, E.T.; Havugimana, P.C.; White, C.; Emili, A.; Sakac, D.; Binnington, B.; Neschadim, A.; McCarthy, S.D.; Branch, D.R. Extracellular histones identified in crocodile blood inhibit in-vitro HIV-1 infection. Aids 2016, 30, 2043–2052. [Google Scholar] [CrossRef] [PubMed]
  160. Huang, S.; Chen, C.P.; Schatz, F.; Rahman, M.; Abrahams, V.; Lockwood, C. Pre--eclampsia is associated with dendritic cell recruitment into the uterine decidua. J. Pathol. 2008, 214, 328–336. [Google Scholar] [CrossRef] [PubMed]
  161. Mothe, B.; Brander, C. Considerations for successful therapeutic immunization in HIV cure. Curr. Opin. HIV AIDS 2021, 16, 257–261. [Google Scholar] [CrossRef]
Figure 1. The homology between HIV-1 and PE response to endothelial dysfunction. HIV-1 accessory proteins gp120, tat and nef, as well as matrix proteins, are protagonists of the cellular machinery that culminate in endothelial cell dysfunction. Adapted from [30].
Figure 1. The homology between HIV-1 and PE response to endothelial dysfunction. HIV-1 accessory proteins gp120, tat and nef, as well as matrix proteins, are protagonists of the cellular machinery that culminate in endothelial cell dysfunction. Adapted from [30].
Ijms 22 09157 g001
Figure 2. HIV-1 accessory protein Tat is released from infected cells, decreases bioavailability of VEGF and increases cell adhesion expression, thereby promoting endothelial cell dysfunction. Adapted from [67,68].
Figure 2. HIV-1 accessory protein Tat is released from infected cells, decreases bioavailability of VEGF and increases cell adhesion expression, thereby promoting endothelial cell dysfunction. Adapted from [67,68].
Ijms 22 09157 g002
Table 2. Antiretroviral Therapy and HIV associated PE.
Table 2. Antiretroviral Therapy and HIV associated PE.
Antiretroviral TypeMain FindingsReferences
Nucleoside/nucleotide reverse transcriptase inhibitors
  • Immune reconstitution
  • Decreased endothelial cell proliferation, migration
  • Elevated mitochondrial oxidative stress
  • Defective tyrosine kinase receptor and VEGFR-2 signalling
  • Increased reactive oxygen species (ROS) generation in trophoblast apoptosis predisposing PE and/or IUGR
[121,122]
Protease inhibitors
  • Immune reconstitution preluding PE development
  • Decreased progesterone in trophoblast cells impede invasion
  • Dysregulated uterine decidualization and spiral artery remodelling
  • Decreased VEGF, placental growth factor (PlGF), angiopoietin-2, interferon-gamma and matrix metalloproteinase 9 (MMP-9) in decidual cell
  • Uterine natural killer (uNK) cell depletion
  • Incomplete trophoblast cell invasion following decreased expression of the transcription factor STAT3
[123,124,125]
HAART
  • Immune reconstitution
  • Dysregulated nuclear factor kappa B (NF-κB) transcription factors and, hence, decreased MMP and VEGF expression
  • Increased sFlt-1 and sEng
  • Decreased PlGF and VCAM-1
[83,88,126,127,128,129,130]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Naicker, T.; Govender, N.; Abel, T.; Naidoo, N.; Moodley, M.; Pillay, Y.; Singh, S.; Khaliq, O.P.; Moodley, J. HIV Associated Preeclampsia: A Multifactorial Appraisal. Int. J. Mol. Sci. 2021, 22, 9157. https://doi.org/10.3390/ijms22179157

AMA Style

Naicker T, Govender N, Abel T, Naidoo N, Moodley M, Pillay Y, Singh S, Khaliq OP, Moodley J. HIV Associated Preeclampsia: A Multifactorial Appraisal. International Journal of Molecular Sciences. 2021; 22(17):9157. https://doi.org/10.3390/ijms22179157

Chicago/Turabian Style

Naicker, Thajasvarie, Nalini Govender, Tashlen Abel, Nitalia Naidoo, Merantha Moodley, Yazira Pillay, Shoohana Singh, Olive Pearl Khaliq, and Jagidesa Moodley. 2021. "HIV Associated Preeclampsia: A Multifactorial Appraisal" International Journal of Molecular Sciences 22, no. 17: 9157. https://doi.org/10.3390/ijms22179157

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

Article Metrics

Back to TopTop