Next Article in Journal
Quality Evaluation of Dietary Supplements for Weight Loss Based on Garcinia cambogia
Previous Article in Journal
Vitamin D and Visceral Obesity in Humans: What Should Clinicians Know?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Potential of Spirulina platensis to Ameliorate the Adverse Effects of Highly Active Antiretroviral Therapy (HAART)

Discipline of Medical Biochemistry and Chemical Pathology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Howard College Campus, Durban 4013, South Africa
*
Author to whom correspondence should be addressed.
Submission received: 6 April 2022 / Revised: 31 May 2022 / Accepted: 3 June 2022 / Published: 27 July 2022
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

:
The human immunodeficiency virus (HIV) is one of the most prevalent diseases globally. It is estimated that 37.7 million people are infected with HIV globally, and 8.2 million persons are infected with the virus in South Africa. The highly active antiretroviral therapy (HAART) involves combining various types of antiretroviral drugs that are dependent on the infected person’s viral load. HAART helps regulate the viral load and prevents its associated symptoms from progressing into acquired immune deficiency syndrome (AIDS). Despite its success in prolonging HIV-infected patients’ lifespans, the use of HAART promotes metabolic syndrome (MetS) through an inflammatory pathway, excess production of reactive oxygen species (ROS), and mitochondrial dysfunction. Interestingly, Spirulina platensis (SP), a blue-green microalgae commonly used as a traditional food by Mexican and African people, has been demonstrated to mitigate MetS by regulating oxidative and inflammatory pathways. SP is also a potent antioxidant that has been shown to exhibit immunological, anticancer, anti-inflammatory, anti-aging, antidiabetic, antibacterial, and antiviral properties. This review is aimed at highlighting the biochemical mechanism of SP with a focus on studies linking SP to the inhibition of HIV, inflammation, and oxidative stress. Further, we propose SP as a potential supplement for HIV-infected persons on lifelong HAART.

1. Introduction

The human immunodeficiency virus (HIV) has continued to be a global public concern due to its widespread infection rate and alarming mortality rate [1]. The Joint United Nations Programme on HIV/AIDS (UNAIDS), in its most recent report in November 2021, estimated that 37.7 million people globally are living with HIV. It was also reported that about 1.5 million new HIV infections and 680,000 AIDS-related deaths have occurred in the year 2020 [1,2,3,4]. In South Africa, approximately 8.2 million people were living with HIV in the year 2021 [4]. According to the South African mid-year population statistics 2021, there has been an unprecedented increase from 79,420 to 85,154 HIV-AIDS-related deaths in 2021 [4]. Recently, the easy availability of antiretrovirals (ARVs) has tremendously changed the pattern of death. ARVs have also helped prolong the lifespan of HIV-infected people in South Africa. Globally, about 27.5 million HIV-infected persons had access to ARVs in 2020, while approximately 5.6 million infected South Africans accessed ARVs in 2020 [1,4,5].
The highly active antiretroviral therapy (HAART) entails combining three or more antiretroviral drugs that are subject to the HIV-infected person’s viral load. HAART assists in regulating viral loads and preventing the progression to AIDS. Despite its success in prolonging HIV-infected patients’ lifespans, the use of HAART promotes metabolic syndrome (MetS) through an inflammatory pathway, excess production of reactive oxygen species (ROS), and mitochondrial dysfunction. Over three decades since its discovery, HAART has significantly improved the diagnosis and management of persons with HIV [6,7,8,9,10,11,12]. The persistence of MetS before and during HAART treatment further highlights the need for more studies on MetS inhibitory compounds, such as Spirulina platensis (SP) [13], Moringa oleifera [14,15], Curcumin [16], and Mangiferin [17]. However, there are other alternatives to combat MetS and these include exercise training [18], life style changes, and properly balanced healthy food choices [19,20].
SP is a blue-green microalgae commonly used as a traditional food by some Mexican and African people [21,22]. SP is mostly found in the alkaline water of volcanic lakes. In addition to its popular nutritional value, SP possesses various medicinal properties. It can induce both the humoral and cellular mechanisms of the immune system when consumed [22]. Interestingly, SP was linked with MetS-lowering properties, such as hypoglycemia [23], hypolipidemia [24], and hypotension [13]. Studies in rodents suggested that SP is particularly useful in preventing MetS [13]. SP contains oxidative stress inhibitors, phycocyanin and phycocyanobilin [25,26,27]. Previous studies have also demonstrated that SP inhibits oxidative stress [21,25,28,29] and promotes mitochondrial health [30,31,32,33], thereby inhibiting inflammation [25,34]. Furthermore, SP can prevent the development of atherosclerosis [28] and diabetes [25]. The Food and Agriculture Organization (FAO) of the United Nations recommends Spirulina as a daily dietary supplement [35]. Microalgaes including Spirulina are environmentally friendly and have a high rate of yield in large-scale production under controlled conditions [36]. Spirulina, in addition to its nutritional and medicinal security, has the potential to eliminate poverty. The considerable potential for sustainable financial development in a small-scale crop for nutritional enhancement was evident in China, where the production increase resulted in a dramatic increase in profit from USD 7.6 million to USD 16.6 million. Spirulina production is possible for small and marginal farmers as well as enthusiastic urban gardeners; this makes it easily accessible to the population [37]. Moreover, the health system can provide SP as medication to control adverse effects in people living with HIV on HAART with minimal costs.
Due to the increasing number of HIV-infected people and their high dependence on HAART, it is imperative to explore the anti-inflammatory and antioxidant mechanisms of SP against HIV and MetS. This review explores the anti-inflammatory and antioxidant mechanisms of SP in the inhibition of MetS and its potential as a supplement against HIV and ARV-induced MetS.

2. The Roles of HIV and HAART in MetS

HIV has often been associated with MetS [38], which results in cardiovascular diseases. Recent reports have linked some HIV-related features to MetS. These characteristic features include escalated cases of cardiovascular diseases, type 2 diabetes mellitus, dyslipidemia, immunodeficiency, high viral load, and atherosclerosis [7]. Recently, studies have suggested that HAART, in addition to the above-mentioned HIV-related features, actively induces MetS in persons with HIV [7,39,40,41]. Earlier studies by Palios et al. (2011) on arterial stiffness, displayed by pulse wave velocity (PWV), and markers of MetS, reported that persons with HIV exhibited an increased degree of PWV when compared with the healthy controls. Subsequently, persons on HAART have been shown to exhibit a similar PWV as persons with hypertension [39,40]. The prevalence of MetS in HIV-infected people receiving antiretroviral (ARV) treatment was higher when compared to the general population. This prevalence was attributed to age, physical inactivity, and a low cluster of differentiation 4 (CD4) count [42,43,44]. The patient response to HAART varies; some antiviral drugs can successfully suppress the plasma viral load without increasing the CD4 count; this allows the risks of opportunistic infections and abnormalities. Failure to increase the CD4 count during HAART may be due to several factors, including drug resistance, low CD4+ T-cell count at the initiation of HAART, the advanced stage of the disease, and a low adherence to HAART [45].
The nuclear factor-kappa-light chain-enhancer of the activated B cell (NF-κB) is a protein complex that is responsible for DNA and cytokine (IL-1β, IL-6, and TNF-α) transcription, chemokine activation, and the survival of cells [7]. NF-κB is also a well-known mediator of inflammation that promotes HIV transcription [7,46].
Inflammation linked to HAART is evidenced by the persistently high levels of interleukin 6 (IL-6), C-reactive proteins, and D-dimers [47]. HIV creates chronic pro-inflammatory conditions that promote MetS [7]. Studies have shown that HIV is associated with inflammation, apoptosis, and mitochondrial dysfunction [7,48]; however, the mechanism that links HIV with MetS remains unclear [7]. Herein, we highlight the significance of SP as an anti-inflammatory supplement for HIV-infected people on lifelong HAART and its mechanism of inhibition on MetS.

3. Spirulina Species

Spirulina has three commonly investigated species due to their potential therapeutic nature and high nutritional content. These Spirulina species include Spirulina platensis (SP) (otherwise known as Arthrospira platensis), Spirulina maxima (Arthrospira maxima), and Spirulina fusiformis (Arthrospira fusiformis). These Spirulina species are also classified as oxygenic photosynthetic bacteria under Cyanobacteria and Prochlorales [49,50,51,52,53,54]. SP is found in alkaline water with abundant bicarbonate and saline [22,55]. Spirulina species are generally three-dimensional helix microstructures [56] protected by a cell wall composed of complex sugars and proteins [22]; however, helical transformation results after mature trichomes divide into hormogonia, binary fission, and undergo length elongation [57]. SP is considered an antioxidant and anti-inflammatory agent [58]. It is also considered as a nutraceutical food supplement due to its high content of proteins, vitamins, and minerals. Moreover, its composition includes chlorophyll, phycocyanin, and carotenoid. Chlorophyll has antioxidant and antimutagenic properties [59,60], carotenoids are vitally important antioxidants with cancer-inhibiting abilities [53], and phycocyanin is a Bili protein with antioxidant and radical scavenging properties [61]. Moreso, SP has also been credited for its cancer- and viral infection-suppressing abilities [62].

3.1. Role of Spirulina in the Inhibition of Oxidative Stress

Recently, research has unveiled the important roles of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in the production of reactive oxygen species (ROS). NADPH is contained in the nervous system and its assemblage and activation generate free radicals (FR) which subsequently destroy cells. Spirulina is a potent inhibitor of NADPH oxidase, which has been one of the proteins responsible for the production of ROS and subsequent oxidative stress [54,63,64]. SP has the potential to inhibit oxidative stress by blocking NADPH oxidase [21,25,28,29] and to enhance (Figure 1) mitochondrial health by promoting an antioxidant response [30,31,32,33]. Furthermore, SP prevents FR-induced apoptotic cell death through the inhibition of oxidative stress [65].

3.2. Multitargeted Therapeutic Roles of SP

SP has a therapeutic effect against vascular diseases, cancer, diabetes, neurodegenerative diseases, and inflammatory disorders [66]. Spirulina treatment enhances the NF-kappa B-directed luciferase expression [67]. It has antiallergic effects [54], including against allergic rhinitis in humans [68]. Spirulina is an immune booster [22]. It prevents cellular aging, infectious diseases, and promotes a strong immune system [57]. Spirulina has central neuroprotective effects in rodents [69]. It is also associated with the inhibitory effects against numerous viruses, such as HIV-1, herpes simplex virus 1 and 2 (HSV-1 and HSV-2), human cytomegalovirus (HCMV), influenza type A, measles, and other enveloped viruses [53,70,71,72,73]. Moreover, it has antimutagenic and anticancer effects [57]. Spirulina is an effective treatment against chronic arsenic poisoning with melanosis and keratosis [74]. SP shares similar chemical structures and physiological activities with bilirubin [25,26,27,28,75]. SP antioxidant properties are due to its composition and the presence of phycobilins, phycocyanin, and phycocyanobilin [25,26,27] (Figure 2). Phycobilins are similar in structure to bile pigments such as bilirubin, a well-known ROS scavenger [22,76]. Phycocyanin has been proven to possess antioxidant and anti-inflammatory activities [34,54,63,64,77,78]. Phycocyanin is also structurally similar to biliverdin, a strong inhibitor of NADPH oxidase and inflammation-induced radicals [25,34]. A study conducted by Zheng et al. (2013) indicated that phycocyanin normalized urinary and renal oxidative stress markers and the expression of NADPH oxidase components. Furthermore, phycocyanobilin, bilirubin, and biliverdin inhibited NADPH-dependent superoxide production in renal mesangial cells [25]. The study also demonstrated that SP may be used in a therapeutic approach to prevent diabetic nephropathy through the inhibition of oxidative stress [25]. Thus far, phycocyanine, the most powerful natural antioxidant, is only present in cyanobacteria and thus in spirulina.
SP exhibited neuroprotective activities through antioxidant and anti-inflammatory effects [79]. It has also shown significant antioxidant activity in vitro by scavenging nitric oxide and preventing DNA damage by scavenging hydroxyl radical (Figure 3) [80]. Antidiabetic and anti-inflammatory properties of SP [81] are based on its significant free-radical scavenging activities. SP contains compounds that fall under a broad spectrum of antioxidant agents, such as alkaloids, flavonoids [82], and phycocyanin [28,83,84,85,86]. Moreso, it provides trace minerals for the synthesis of antioxidant enzymes, demonstrated by the antidiabetic response in rats [87]. It has potential benefits in assisting with the reduction in chronic inflammatory conditions [88]. Spirulina incorporated into skin cream showed promising results as an anti-inflammatory and a wound-healing agent [89].
Spirulina against HIV-1 demonstrated its ability as an antiviral compound. Studies have demonstrated the ability of SP in the inhibition of HIV-1 replication in human T-cell lines, peripheral blood mononuclear cells (PBMC), and Langerhans cells (LC). The inhibition of the viral production by spirulina extract (between 0.3 and 1.2 μg/mL) was found to be approximately 50% in PBMCs [57]. More studies are needed to fully understand the mechanism behind the inhibition of HIV by SP.

3.3. Mechanism of Action

SP contains several vital antioxidant and anti-inflammatory compounds as mentioned above, such as chlorophyll, phycocyanin, and carotenoids (β-carotene). The antioxidant and anti-inflammatory properties of phycocyanin have been determined in numerous studies [28,83,84,85,86,90,91,92,93,94,95,96,97]. Phycocyanin is responsible for reducing oxidative stress and NADPH oxidase [28]. It scavenges free radicals, such as alkoxy, hydroxyl, and peroxyl radicals, and decreases nitrite production and inducible nitric oxide synthase (iNOS) expression. Phycocyanin also inhibits liver microsomal lipid peroxidation [28,83,84,85,86,90,91,92,93,94,95,96,97].
Phycocyanin has been proven to inhibit the formation of the pro-inflammatory cytokine TNF-α and cyclooxygenase-2 (COX-2) expression. Additionally, it decreases prostaglandin E(2) production [28,83,84,85,86]. Phycocyanin prevents the degradation of cytosolic IκB-α, which suppresses the activation of nuclear factor-κB (NF-κB) [28]. Furthermore, the inhibitory activity of phycocyanin is associated with the suppression of TNF-α formation in the macrophages [86]. In addition, phycocyanin exerts regulatory effects on mitogen-activated protein kinase (MAPK) activation pathways, such as the p38, c-Jun N-terminal kinase (JNK), and extracellular-signal-regulated kinase (ERK1/2) pathways [98,99,100]. The second compound of SP, carotenoids, β-carotene to be specific, is an antioxidant that has anti-carcinogenic, antioxidant, and anti-inflammatory activities [101,102,103]. As a membrane antioxidant, β-carotene protects against singlet oxygen-mediated lipid peroxidation [101]. Beta carotene inhibits the production of nitric oxide and prostaglandin E(2) and suppresses the expression of inducible nitric oxide synthase (iNOS), COX-2, TNF-α, and IL-1β. The suppression of inflammatory mediators by β-carotene results from its’ ability to inhibit NF-κB activation by preventing nuclear translocation of the NF-κB p65 subunit [102,104,105]. Studies have shown that β-carotene suppressed the transcription of inflammatory cytokines such as IL-1β, IL-6, and IL-12 in vitro [103]; this takes place in the macrophages. The third compound of spirulina, chlorophyll, can perform antioxidant and antimutagenic activities [59,60]. Spirulina’s mechanism of action is a concert of compounds, but it is not clear whether they all act simultaneously during demanding events.
SP promotes the activation and expression of heme oxygenase 1 (HO-1) and endothelial nitric oxide synthase (eNOS) [104,105]. HO-1 is suggested to play an important part in the adaptive reprogramming which could result in Nrf-2 activation, but this pathway is unclear, and more studies are required. Moreso, SP causes the activation of the Nrf2/HO-1 pathway [106]. Nrf-2 activation by SP results in the production and increased expression of antioxidant enzymes, such as superoxide dismutase (SOD) and catalase (CAT) [100].

4. Common Highly Active Antiretroviral Therapy (HAART) Combinations

HAART has several classes: nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs) [48,107,108,109,110,111,112,113,114], protease inhibitors (PIs) [114,115,116,117], integrase strand transfer inhibitors (INSTIs) [118,119,120,121,122,123], fusion inhibitors (FIs) [124], and chemokine receptor antagonists (CCR5 antagonists) [125,126,127]. HAART is a specifically selected combination of NRTI and NNRTI, PI, or INSTI drugs responsible for the inhibition of viral replication by multiple virus targets [113,128,129,130,131,132,133,134,135,136]. However, HAART can cause adverse drug reactions. Furthermore, 2′,3′-dideoxy-3′-thiacytidine (3TC), 2′,3′-dideoxy-5-fluoro-3′-thiacytidine (FTC), TDF (Tenofovir Disoproxil Fumarate), ZDV (Zidovudine), and d4T (Stavudine) are associated with mitochondrial toxicity and oxidative stress [137,138,139,140]. Additionally, NNRTIs are linked to toxic hepatitis, and PIs are implicated in insulin resistance and hyperlipidemia [114]. Chronic side effects linked to HAART include ROS-induced insulin resistance [141,142], lipodystrophy, gastrointestinal disorders [143], and cardiovascular disease [11,144]. Primarily, HAART is used for the treatment and prevention of HIV-1. These primary functions of HAART are achieved by attacking different components of the virus lifecycle which ensures inhibition regardless of the virus being resistant to one of the drugs [113,128,129,130,131,132,133,134,135,136]. A combination of two NRTIs (mostly FTC and TDF) and one NNRTI (e.g., EFV; Efavirenz) is a more favorable choice due to the convenience to dose, effectiveness, and less toxic effects compared to other drug combinations [114,145,146]. A combination of three NRTIs is less effective than two NRTIs with an NNRTI [147]. The d4T/ddI combination is associated with high toxicity and hence it is not often recommended [148]. The most popular NRTIs are cytidine analogs (XTC), FTC, 3TC, and TDF, which form part of the first-line therapy [149,150,151,152,153]. FTC and 3TC are similar in chemical structures with different pharmacokinetic and pharmacodynamic properties, and they have required deoxynucleosides for HIV DNA synthesis. They undergo phosphorylation through intracellular kinases to become FTC 50 -triphosphate (FTC-TP) and 3TC-TP; triphosphate metabolites with FTC-TP are more efficiently incorporated during HIV DNA synthesis than 3TC-TP [145,154,155,156,157]. Moreover, 3TC-TP has a shorter intracellular half-life compared to FTC-TP [155]. The TDF and FTC combination has a synergistic effect, increasing intracellular metabolites, and they are recommended for pre-exposure prophylaxis (PrEP) [155,158,159,160,161]. Contraindications (Table 1) on HAART treatment are antiretroviral medication-specific and can be overcome by a change in the HAART combination to suit the individuals own treatment [107,108,119,120,121,131,162,163,164,165] or the supplementation and treatment of the symptoms.

5. Mechanism of HAART-Induced Oxidative Stress, Inflammation, and Mitochondrial Dysfunction

The exact mechanism of HAART-induced oxidative stress has not been completely explored; however, studies have demonstrated the link between HAART use and oxidative stress. HAART is linked with lipid metabolism dysfunction through the induction of peripheral lipodystrophy. Lipodystrophy results from the impaired cytoplasmic retinoic-acid binding protein type 1 (CRABP1)-mediated cis-9-retinoic acid stimulation of peroxisome proliferator-activated receptor type gamma (PPAR-γ), leading to impaired differentiation and increased apoptosis of peripheral adipocytes. HIV-1 protease-inhibitors further inhibit the cytochrome P450 3A-mediated synthesis of cis-9-retinoic acid, one of the key activators of PPAR-γ [166]. Insulin resistance occurs following impaired fat storage and lipid release [141,142,166], which impacts the oxidant profile. The depletion of ATP production and mitochondrial dysfunction [8,9], and the depletion of mitochondrial DNA [167,168,169], are some of the ways HAART induces oxidative stress. HIV increases oxidative stress, and HAART increases lipid oxidation, which amplifies the ROS imbalance leading to increased oxidative stress complications [10,170].
Chronic exposure to HAART induces increased oxidative stress in endothelial cells and mononuclear cell recruitment, which leads to cardiovascular diseases in HIV patients on ARV therapy [12]. Inducing oxidative stress is common for protease and reverse transcriptase inhibitors [171]. HAART drugs induce oxidative stress in various ways, which include inhibiting DNA pol-γ activity and leading to mitochondrial dysfunction, and also through the depletion of mitochondrial DNA [167,168,169]. Studies have shown that patients on HAART have abnormally high levels of free oxygen radicals in sera compared to untreated HIV patients and HIV-uninfected participants [10,11,168,170,172,173].
Adverse drug reactions vary; TDF and lopinavir cause acute and chronic renal dysfunction [174,175,176,177,178]. TDF inhibits mitochondrial DNA-polymerase gamma, hence leading to the impaired function of energy-dependent transporters [179,180]. NRTIs are associated with the inhibition of mitochondrial DNA polymerase, lactic acidosis, subcutaneous lipoatrophy, peripheral neuropathy, and pancreatitis. The level of mitochondrial toxicity depends on the drugs; it is low with 3TC, FTC, and TDF, followed by ZDV, and higher with d4T. NNRTIs are associated with life-threatening skin reactions and toxic hepatitis. PIs are associated with insulin resistance and hyperlipidemia [7,114].

6. Combined and Synergistic Therapeutic Actions of HAART and SP

Studies have shown that the possible therapeutic effects of antioxidants may provide strategies in suppressing oxidative stress-induced comorbidities that emerge with the use of HAART therapy in HIV-infected individuals [12]. The combination of HIV and HAART has been associated with increased oxidative stress and lipid peroxidation. Furthermore, HIV or HAART induces ROS by inducing NADPH oxidase [181,182]. Interestingly, SP is a potent antioxidant [26,27] with anti-inflammatory activities [34], which makes it a potential supplement in the mitigation of oxidative stress induced by HAART adverse drug reactions. Moreover, SP can inhibit NADPH oxidase which is considered one of the main sources of ROS and free radicals in HIV-infected persons on HAART [34,181,182], resulting in reduced oxidative stress [28]. Moreover, β-carotene from SP protects against singlet oxygen-mediated lipid peroxidation [101]. Among HAART complications, TDF and lopinavir cause acute and chronic renal dysfunction [174,175,176,177,178]. Herein, phycocyanin from SP can normalize urinary and renal oxidative stress markers and inhibit NADPH-dependent superoxide production in renal mesangial cells [25], ameliorating renal dysfunction. Lately, SP has been an effective therapeutic approach to preventing diabetic nephropathy through the inhibition of oxidative stress [25]. These properties indicate SP as a potential agent to mitigate renal dysfunction caused by HAART. As mentioned above, NRTIs can inhibit mitochondrial DNA polymerase [179,180]. Studies in vitro showed that SP can enhance cell nucleus enzyme function, repair DNA synthesis [57], and enhance mitochondrial health [30,31,32,33,80]. Mitochondrial toxicity presented as peripheral neuropathy and lactic acidosis can be corrected by SP through providing trace minerals for the synthesis of antioxidant enzymes [87] and reducing chronic inflammatory conditions [88].
NNRTIs are associated with life-threatening skin reactions and toxic hepatitis [114], these conditions may be ameliorated by SP. Phycocyanin from SP can inhibit liver microsomal lipid peroxidation [28,83,84,85,86,90,91,92,93,94,95,96,97], and hence reducing toxic hepatitis. Moreso, SP incorporated into skin creams showed promising results as an anti-inflammatory and a wound-healing agent [89]; this can be beneficial in the mitigation of NNRTI-induced skin reactions. PI therapy induces insulin resistance and hyperlipidemia [7]. Additionally, HAART may be associated with a higher risk of myocardial infarction [114,183,184]. SP has a therapeutic effect against vascular diseases, cancer, diabetes, and neurodegenerative diseases [66]. In addition, the Spirulina family has also shown central neuroprotective effects in rodents [69] and may exert its neuroprotective activities through antioxidant and anti-inflammatory effects [79]. Therefore, SP is a recommended antioxidant to use as a supplement; the list of benefits is evident. It also has antiallergic effects [54], prevents cellular aging and infectious diseases, and promotes a strong immune system [57]. Herein, promotion of a strong immune system by SP can help increase CD4 cell counts, lower HIV viral loads, and slow down the progression to AIDS. Moreover, SP prevents FR-induced apoptotic cell death [65]; this may help decrease apoptosis of peripheral adipocytes induced by HAART. Chemically, SP is a recommended source of proteins, vitamins, and minerals [57], important nutrients for individuals on the HAART program. Finally, SP can assist HAART in the inhibition of HIV-1 replication because it has been shown to inhibit viral production in PBMCs.
There has been a number of clinical studies to investigate whether SP improves the quality of life in HIV-infected individuals. Marcel (2011) reported that insulin sensitivity in HIV patients improved more when a spirulina nutritional supplement was used instead of soybean [185]. Another study demonstrated for the first time that spirulina improves antioxidant capacity in people living with HIV [186]. Spirulina supplementation combined with a qualitative balanced diet showed potential to inhibit lipid abnormalities [187], significantly increase CD4 cells, and reduce the viral load in HIV-infected antiretroviral-naive patients [187,188,189]. However, there is limited information on the investigation of SP confirming the mechanism of antioxidant and anti-inflammatory effects and the impact on the quality of life in the HIV-positive population taking HAART. Thus far, it has been shown that supplementation with Spirulina platensis could improve the immune status of HIV patients on ART and decrease inflammatory and pro-oxidant levels [190]. The development of more clinical studies to confirm the SP protective effect in this population will answer many questions. The recommended concentrations of SP for daily supplementation varies, as studies have successfully used 19 g [185], 5 g [186], and 10 g [190,191].

7. Conclusions

HIV continues to be a major global cause of mortality. Besides the therapeutic benefits of HAART in HIV treatment, HAART has been linked to numerous adverse drug reactions which include oxidative stress, inflammation, and the disruption of mitochondrial function. SP as an antioxidant, anti-inflammatory, anticancer, and nutritional supplement possesses various corrective properties against attacks from viruses and bacteria. The corrective health properties of SP are largely attributed to antioxidant pigments found in SP. These pigments include chlorophyll, carotenoids, and phycocyanin which facilitate antioxidant, anti-inflammatory, and anticancer properties. The corrective properties of SP indicated in this review highlight its potential in the mitigation of HAART adverse drug reactions and MetS. The SP supplement potential is also supported by its ability to assist HAART in the inhibition of HIV-1. This review highlighted the corrective properties of potent antioxidant SP potential as a supplement for individuals on lifelong HAART experiencing MetS. Furthermore, this review highlights the need for more studies on SP and HAART synergy.

Author Contributions

Conceptualization, T.S., T.G., A.C.; writing—original draft preparation, T.S.; writing—review and editing, T.G. and A.C. All authors have read and agreed to the published version of the manuscript.

Funding

Grant sponsor: National Research Foundation Innovation Doctoral Scholarship; Grant number: 130023; Grant sponsor: University of KwaZulu Natal, College of Health Sciences Masters and Doctoral Research Scholarship.

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.

References

  1. WHO. HIV/AIDS. Available online: https://www.who.int/news-room/fact-sheets/detail/hiv-aids (accessed on 30 November 2021).
  2. UNAIDS. Joint United Nations Programme on HIV/AIDS (UNAIDS). Global Report: UNAIDS Report on the Global AIDS Epidemic 2021; Global HIV & AIDS Statistics: Fact Sheet; Joint United Nations Programme on HIV/AIDS: Geneva, Switzerland, 2021. [Google Scholar]
  3. UNAIDS. Global HIV & AIDS Statistics—2020 Fact. Available online: https://www.unaids.org/en/resources/fact-sheet (accessed on 11 September 2021).
  4. Release, S. Mid-Year Population Estimates 2021. Statistics South Africa. 2021. Available online: http://www.statssa.gov.za/publications/P0302/P03022021.pdf (accessed on 18 November 2021).
  5. UNAIDS. HIV and AIDS Estimates. Available online: https://www.unaids.org/en/regionscountries/countries/southafrica (accessed on 11 September 2021).
  6. National Institute on Drug Abuse (NIDA). What Is HAART? Available online: https://www.drugabuse.gov/publications/research-reports/hivaids/what-haart (accessed on 30 November 2021).
  7. Mohan, J.; Ghazi, T.; Chuturgoon, A.A. A Critical Review of the Biochemical Mechanisms and Epigenetic Modifications in HIV- and Antiretroviral-Induced Metabolic Syndrome. Int. J. Mol. Sci. 2021, 22, 12020. [Google Scholar] [CrossRef] [PubMed]
  8. Manda, K.R.; Banerjee, A.; Banks, W.A.; Ercal, N. Highly active antiretroviral therapy drug combination induces oxidative stress and mitochondrial dysfunction in immortalized human blood–brain barrier endothelial cells. Free Radic. Biol. Med. 2011, 50, 801–810. [Google Scholar] [CrossRef] [PubMed]
  9. Blas-Garcia, A.; Apostolova, N.; Esplugues, J.V. Oxidative stress and mitochondrial impairment after treatment with anti-HIV drugs: Clinical implications. Curr. Pharm. Des. 2011, 17, 4076–4086. [Google Scholar] [CrossRef] [PubMed]
  10. Ngondi, J.L.; Oben, J.; Forkah, D.M.; Etame, L.H.; Mbanya, D. The effect of different combination therapies on oxidative stress markers in HIV infected patients in Cameroon. AIDS Res. Ther. 2006, 3, 19. [Google Scholar] [CrossRef] [Green Version]
  11. Masiá, M.; Padilla, S.; Bernal, E.; Almenar, M.V.; Molina, J.; Hernández, I.; Graells, M.L.; Gutiérrez, F. Influence of antiretroviral therapy on oxidative stress and cardiovascular risk: A prospective cross-sectional study in HIV-infected patients. Clin. Ther. 2007, 29, 1448–1455. [Google Scholar] [CrossRef]
  12. Mondal, D.; Pradhan, L.; Ali, M.; Agrawal, K.C. HAART drugs induce oxidative stress in human endothelial cells and increase endothelial recruitment of mononuclear cells. Cardiovasc. Toxicol. 2004, 4, 287–302. [Google Scholar] [CrossRef]
  13. Finamore, A.; Palmery, M.; Bensehaila, S.; Peluso, I. Antioxidant, immunomodulating, and microbial-modulating activities of the sustainable and ecofriendly spirulina. Oxidative Med. Cell. Longev. 2017, 2017, 1–4. [Google Scholar] [CrossRef] [Green Version]
  14. Nyakudya, T.T.; Tshabalala, T.; Dangarembizi, R.; Erlwanger, K.H.; Ndhlala, A.R. The potential therapeutic value of medicinal plants in the management of metabolic disorders. Molecules 2020, 25, 2669. [Google Scholar] [CrossRef]
  15. Kim, D.S.; Choi, M.H.; Shin, H.J. Extracts of Moringa oleifera leaves from different cultivation regions show both antioxidant and antiobesity activities. J. Food Biochem. 2020, 44, e13282. [Google Scholar] [CrossRef]
  16. Pérez-Torres, I.; Ruiz-Ramírez, A.; Baños, G.; El-Hafidi, M. Hibiscus sabdariffa Linnaeus (Malvaceae), curcumin and resveratrol as alternative medicinal agents against metabolic syndrome. Cardiovasc. Hematol. Agents Med. Chem. 2013, 11, 25–37. [Google Scholar] [CrossRef]
  17. Mujawdiya, P.K.; Kapur, S. Mangiferin: A potential natural molecule for management of metabolic syndrome. Int. J. Pharm. Pharm. Sci. 2015, 7, 9–13. [Google Scholar]
  18. Kränkel, N.; Bahls, M.; van Craenenbroeck, E.M.; Adams, V.; Serratosa, L.; Solberg, E.E.; Hansen, D.; Dörr, M.; Kemps, H. Exercise training to reduce cardiovascular risk in patients with metabolic syndrome and type 2 diabetes mellitus: How does it work? Eur. J. Prev. Cardiol. 2019, 26, 701–708. [Google Scholar] [CrossRef] [PubMed]
  19. Opie, L.H. Metabolic syndrome. Circulation 2007, 115, e32–e35. [Google Scholar] [CrossRef] [PubMed]
  20. Marrone, G.; Guerriero, C.; Palazzetti, D.; Lido, P.; Marolla, A.; Di Daniele, F.; Noce, A. Vegan diet health benefits in metabolic syndrome. Nutrients 2021, 13, 817. [Google Scholar] [CrossRef] [PubMed]
  21. Bashandy, S.A.; El Awdan, S.A.; Ebaid, H.; Alhazza, I.M. Antioxidant potential of Spirulina platensis mitigates oxidative stress and reprotoxicity induced by sodium arsenite in male rats. Oxidative Med. Cell. Longev. 2016, 2016, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Estrada, J.P.; Bescós, P.B.; del Fresno, A.V. Antioxidant activity of different fractions of Spirulina platensis protean extract. Il Farm. 2001, 56, 497–500. [Google Scholar] [CrossRef]
  23. Iyer Uma, M.; Sophia, A.; Uliyar, V.M. Glycemic and lipemic responses of selected spirulina-supplemented rice-based recipes in normal subjects. Age Years 1999, 22, 1–8. [Google Scholar]
  24. Serban, M.C.; Sahebkar, A.; Dragan, S.; Stoichescu-Hogea, G.; Ursoniu, S.; Andrica, F.; Banach, M. A systematic review and meta-analysis of the impact of Spirulina supplementation on plasma lipid concentrations. Clin. Nutr. 2016, 35, 842–851. [Google Scholar] [CrossRef]
  25. Zheng, J.; Inoguchi, T.; Sasaki, S.; Maeda, Y.; McCarty, M.F.; Fujii, M.; Ikeda, N.; Kobayashi, K.; Sonoda, N.; Takayanagi, R. Phycocyanin and phycocyanobilin from Spirulina platensis protect against diabetic nephropathy by inhibiting oxidative stress. Am. J. Physiol. -Regul. Integr. Comp. Physiol. 2013, 304, R110–R120. [Google Scholar] [CrossRef] [Green Version]
  26. Hu, Z.; Liu, Z. Determination and purification of beta-carotene in Spirulina maximum. Chin. J. Chromatogr. 2001, 19, 85–87. [Google Scholar]
  27. Miranda, M.S.; Cintra, R.G.; Barros, S.; Mancini-Filho, J. Antioxidant activity of the microalga Spirulina maxima. Braz. J. Med. Biol. Res. 1998, 31, 1075–1079. [Google Scholar] [CrossRef] [PubMed]
  28. Riss, J.; Décordé, K.; Sutra, T.; Delage, M.; Baccou, J.C.; Jouy, N.; Brune, J.P.; Oréal, H.; Cristol, J.P.; Rouanet, J.M. Phycobiliprotein C-phycocyanin from Spirulina platensis is powerfully responsible for reducing oxidative stress and NADPH oxidase expression induced by an atherogenic diet in hamsters. J. Agric. Food Chem. 2007, 55, 7962–7967. [Google Scholar] [CrossRef] [PubMed]
  29. Abdelkhalek, N.K.; Ghazy, E.W.; Abdel-Daim, M.M. Pharmacodynamic interaction of Spirulina platensis and deltamethrin in freshwater fish Nile tilapia, Oreochromis niloticus: Impact on lipid peroxidation and oxidative stress. Environ. Sci. Pollut. Res. 2015, 22, 3023–3031. [Google Scholar] [CrossRef] [PubMed]
  30. Nawrocka, D.; Kornicka, K.; Śmieszek, A.; Marycz, K. Spirulina platensis improves mitochondrial function impaired by elevated oxidative stress in adipose-derived mesenchymal stromal cells (ASCs) and intestinal epithelial cells (IECs), and enhances insulin sensitivity in equine metabolic syndrome (EMS) horses. Mar. Drugs 2017, 15, 237. [Google Scholar] [CrossRef] [Green Version]
  31. Jadaun, P.; Yadav, D.; Bisen, P.S. Spirulina platensis prevents high glucose-induced oxidative stress mitochondrial damage mediated apoptosis in cardiomyoblasts. Cytotechnology 2018, 70, 523–536. [Google Scholar] [CrossRef] [PubMed]
  32. Sun, J.Y.; Hou, Y.J.; Fu, X.Y.; Fu, X.T.; Ma, J.K.; Yang, M.F.; Sun, B.L.; Fan, C.D.; Oh, J. Selenium-containing protein from selenium-enriched spirulina platensis attenuates cisplatin-induced apoptosis in MC3T3-E1 Mouse preosteoblast by inhibiting mitochondrial dysfunction and ROS-Mediated oxidative damage. Front. Physiol. 2019, 9, 1907. [Google Scholar] [CrossRef]
  33. Oriquat, G.A.; Ali, M.A.; Mahmoud, S.A.; Eid, R.M.; Hassan, R.; Kamel, M.A. Improving hepatic mitochondrial biogenesis as a postulated mechanism for the antidiabetic effect of Spirulina platensis in comparison with metformin. Appl. Physiol. Nutr. Metab. 2019, 44, 357–364. [Google Scholar] [CrossRef]
  34. Izadi, M.; Fazilati, M. Extraction and purification of phycocyanin from spirulina platensis and evaluating its antioxidant and anti-inflammatory activity. Asian J. Green Chem. 2018, 2, 364–379. [Google Scholar]
  35. Pelizer, L.H.; Danesi, E.D.G.; de O Rangel, C.; Sassano, C.E.; Carvalho, J.C.M.; Sato, S.; Moraes, I.O. Influence of inoculum age and concentration in Spirulina platensis cultivation. J. Food Eng. 2003, 56, 371–375. [Google Scholar] [CrossRef]
  36. Pina-Pérez, M.C.; Brück, W.M.; Brück, T.; Beyrer, M. Microalgae as healthy ingredients for functional foods. In The Role of Alternative and Innovative Food Ingredients and Products in Consumer Wellness; Elsevier: Amsterdam, The Netherlands, 2019; pp. 103–137. [Google Scholar]
  37. Uddin, A.J.; Mahbuba, S.; Rahul, S.; Ifaz, M.; Ahmad, H. Super food Spirulina (Spirulina platensis): Prospect and scopes in Bangladesh. Int. J. Bus. Soc. Sci. Res. 2018, 6, 51–55. [Google Scholar]
  38. Li Vecchi, V.; Maggi, P.; Rizzo, M.; Montalto, G. The metabolic syndrome and HIV infection. Curr. Pharm. Des. 2014, 20, 4975–5003. [Google Scholar] [CrossRef] [PubMed]
  39. Aounallah, M.; Dagenais-Lussier, X.; El-Far, M.; Mehraj, V.; Jenabian, M.A.; Routy, J.P.; van Grevenynghe, J. Current topics in HIV pathogenesis, part 2: Inflammation drives a Warburg-like effect on the metabolism of HIV-infected subjects. Cytokine Growth Factor Rev. 2016, 28, 1–10. [Google Scholar] [CrossRef] [PubMed]
  40. Palios, J.; Kadoglou, N.P.; Lampropoulos, S. The pathophysiology of HIV-/HAART-related metabolic syndrome leading to cardiovascular disorders: The emerging role of adipokines. Exp. Diabetes Res. 2011, 2012, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Barbaro, G.; Iacobellis, G. Metabolic syndrome associated with HIV and highly active antiretroviral therapy. Curr. Diabetes Rep. 2009, 9, 37–42. [Google Scholar] [CrossRef] [PubMed]
  42. Rogalska-Płońska, M.; Grzeszczuk, A.; Rogalski, P.; Łucejko, M.; Flisiak, R. Metabolic syndrome in HIV infected adults in Poland. Kardiol. Pol. Pol. Heart J. 2018, 76, 548–553. [Google Scholar] [CrossRef] [Green Version]
  43. Nguyen, K.A.; Peer, N.; Mills, E.J.; Kengne, A.P. A meta-analysis of the metabolic syndrome prevalence in the global HIV-infected population. PLoS ONE 2016, 11, e0150970. [Google Scholar] [CrossRef] [Green Version]
  44. Ataro, Z.; Ashenafi, W. Metabolic syndrome and associated factors among adult HIV positive people on antiretroviral therapy in Jugal hospital, Harar, Eastern Ethiopia. East Afr. J. Health Biomed. Sci. 2020, 4, 13–24. [Google Scholar]
  45. Aiuti, F.; Mezzaroma, I. Failure to reconstitute CD4+ T-cells despite suppression of HIV replication under HAART. AIDS Rev. 2006, 8, 88–97. [Google Scholar]
  46. Baker, R.G.; Hayden, M.S.; Ghosh, S. NF-κB, inflammation, and metabolic disease. Cell Metab. 2011, 13, 11–22. [Google Scholar] [CrossRef] [Green Version]
  47. Nasi, M.; De Biasi, S.; Gibellini, L.; Bianchini, E.; Pecorini, S.; Bacca, V.; Guaraldi, G.; Mussini, C.; Pinti, M.; Cossarizza, A. Ageing and inflammation in patients with HIV infection. Clin. Exp. Immunol. 2017, 187, 44–52. [Google Scholar] [CrossRef] [Green Version]
  48. Lucas, G.M.; Ross, M.J.; Stock, P.G.; Shlipak, M.G.; Wyatt, C.M.; Gupta, S.K.; Atta, M.G.; Wools-Kaloustian, K.K.; Pham, P.A.; Bruggeman, L.A.; et al. Clinical practice guideline for the management of chronic kidney disease in patients infected with HIV: 2014 update by the HIV Medicine Association of the Infectious Diseases Society of America. Clin. Infect. Dis. 2014, 59, e96–e138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Turner, S. Molecular systematics of oxygenic photosynthetic bacteria. Orig. Algae Plast. 1997, 11, 13–52. [Google Scholar]
  50. Whitton, B.A. Diversity, ecology, and taxonomy of the cyanobacteria. In Photosynthetic Prokaryotes; Springer: Berlin/Heidelberg, Germany, 1992; pp. 1–51. [Google Scholar]
  51. Vonshak, A. Spirulina Platensis Arthrospira: Physiology, Cell-Biology and Biotechnology; CRC Press: Boca Raton, FL, USA, 1997. [Google Scholar]
  52. Gershwin, M.E.; Belay, A. Spirulina in Human Nutrition and Health; CRC Press: Boca Raton, FL, USA, 2007. [Google Scholar]
  53. Khan, Z.; Bhadouria, P.; Bisen, P. Nutritional and therapeutic potential of Spirulina. Curr. Pharm. Biotechnol. 2005, 6, 373–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Karkos, P.D.; Leong, S.C.; Karkos, C.D.; Sivaji, N.; Assimakopoulos, D.A. Spirulina in clinical practice: Evidence-based human applications. Evid.-Based Complementary Altern. Med. 2011, 2011, 1–3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ciferri, O. Spirulina, the edible microorganism. Microbiol. Rev. 1983, 47, 551. [Google Scholar] [CrossRef]
  56. Kamata, K.; Piao, Z.; Suzuki, S.; Fujimori, T.; Tajiri, W.; Nagai, K.; Iyoda, T.; Yamada, A.; Hayakawa, T.; Ishiwara, M.; et al. Spirulina-templated metal microcoils with controlled helical structures for THz electromagnetic responses. Sci. Rep. 2014, 4, 1–7. [Google Scholar] [CrossRef] [Green Version]
  57. Ali, S.K.; Saleh, A.M. Spirulina-an overview. Int. J. Pharm. Pharm. Sci. 2012, 4, 9–15. [Google Scholar]
  58. Henrikson, R. Earth Food Spirulina; Ronore Enterprises, Inc.: Laguna Beach, CA, USA, 1989; p. 187. [Google Scholar]
  59. Ferruzzi, M.G.; Blakeslee, J. Digestion, absorption, and cancer preventative activity of dietary chlorophyll derivatives. Nutr. Res. 2007, 27, 1–12. [Google Scholar] [CrossRef]
  60. Hosikian, A.; Lim, S.; Halim, R.; Danquah, M.K. Chlorophyll extraction from microalgae: A review on the process engineering aspects. Int. J. Chem. Eng. 2010, 2010, 1–10. [Google Scholar] [CrossRef] [Green Version]
  61. Hoseini, S.M.; Khosravi-Darani, K.; Mozafari, M.R. Nutritional and medical applications of spirulina microalgae. Mini Rev. Med. Chem. 2013, 13, 1231–1237. [Google Scholar] [CrossRef]
  62. Asghari, A.; Fazilati, M.; Latifi, A.M.; Salavati, H.; Choopani, A. A review on antioxidant properties of Spirulina. J. Appl. Biotechnol. Rep. 2016, 3, 345–351. [Google Scholar]
  63. Wu, Q.; Liu, L.; Miron, A.; Klímová, B.; Wan, D.; Kuča, K. The antioxidant, immunomodulatory, and anti-inflammatory activities of Spirulina: An overview. Arch. Toxicol. 2016, 90, 1817–1840. [Google Scholar] [CrossRef] [PubMed]
  64. Deng, R.; Chow, T.J. Hypolipidemic, antioxidant, and antiinflammatory activities of microalgae Spirulina. Cardiovasc. Ther. 2010, 28, e33–e45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Chu, W.L.; Lim, Y.W.; Radhakrishnan, A.K.; Lim, P.E. Protective effect of aqueous extract from Spirulina platensis against cell death induced by free radicals. BMC Complementary Altern. Med. 2010, 10, 1–8. [Google Scholar] [CrossRef] [Green Version]
  66. McCarty, M.F. ‘‘Iatrogenic Gilbert syndrome’’–A strategy for reducing vascular and cancer risk by increasing plasma unconjugated bilirubin. Med. Hypotheses 2007, 69, 974–994. [Google Scholar] [CrossRef]
  67. Balachandran, P.; Pugh, N.D.; Ma, G.; Pasco, D.S. Toll-like receptor 2-dependent activation of monocytes by Spirulina polysaccharide and its immune enhancing action in mice. Int. Immunopharmacol. 2006, 6, 1808–1814. [Google Scholar] [CrossRef]
  68. Mao, T.; Water, J.V.d.; Gershwin, M.E. Effects of a Spirulina-based dietary supplement on cytokine production from allergic rhinitis patients. J. Med. Food 2005, 8, 27–30. [Google Scholar] [CrossRef] [Green Version]
  69. Chamorro, G.; Pérez-Albiter, M.; Serrano-García, N.; Mares-Sámano, J.J.; Rojas, P. Spirulina maxima pretreatment partially protects against 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine neurotoxicity. Nutr. Neurosci. 2006, 9, 207–212. [Google Scholar] [CrossRef]
  70. Hernández-Corona, A.; Nieves, I.; Meckes, M.; Chamorro, G.; Barron, B.L. Antiviral activity of Spirulina maxima against herpes simplex virus type 2. Antivir. Res. 2002, 56, 279–285. [Google Scholar] [CrossRef]
  71. Luescher-Mattli, M. Algae, a possible source for new drugs in the treatment of HIV and other viral diseases. Curr. Med. Chem. -Anti-Infect. Agents 2003, 2, 219–225. [Google Scholar] [CrossRef]
  72. Hayashi, T.; Hayashi, K.; Maeda, M.; Kojima, I. Calcium spirulan, an inhibitor of enveloped virus replication, from a blue-green alga Spirulina platensis. J. Nat. Prod. 1996, 59, 83–87. [Google Scholar] [CrossRef] [PubMed]
  73. Ayehunie, S.; Belay, A.; Baba, T.W.; Ruprecht, R.M. Inhibition of HIV-1 replication by an aqueous extract of Spirulina platensis (Arthrospira platensis). J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 1998, 18, 7–12. [Google Scholar] [CrossRef] [PubMed]
  74. Choudhary, M.; Jetley, U.K.; Khan, M.A.; Zutshi, S.; Fatma, T. Effect of heavy metal stress on proline, malondialdehyde, and superoxide dismutase activity in the cyanobacterium Spirulina platensis-S5. Ecotoxicol. Environ. Saf. 2007, 66, 204–209. [Google Scholar] [CrossRef] [PubMed]
  75. Jiang, F.; Roberts, S.J.; Datla, S.R.; Dusting, G.J. NO modulates NADPH oxidase function via heme oxygenase-1 in human endothelial cells. Hypertension 2006, 48, 950–957. [Google Scholar] [CrossRef] [Green Version]
  76. Bhat, V.B.; Madyastha, K. C-phycocyanin: A potent peroxyl radical scavenger in vivo and in vitro. Biochem. Biophys. Res. Commun. 2000, 275, 20–25. [Google Scholar] [CrossRef]
  77. Ashaolu, T.J.; Samborska, K.; Lee, C.C.; Tomas, M.; Capanoglu, E.; Tarhan, Ö.; Taze, B.; Jafari, S.M. Phycocyanin, a super functional ingredient from algae; properties, purification characterization, and applications. Int. J. Biol. Macromol. 2021, 193, 2320–2331. [Google Scholar] [CrossRef]
  78. Grover, P.; Bhatnagar, A.; Kumari, N.; Bhatt, A.N.; Nishad, D.K.; Purkayastha, J. C-Phycocyanin-a novel protein from Spirulina platensis-In vivo toxicity, antioxidant and immunomodulatory studies. Saudi J. Biol. Sci. 2021, 28, 1853–1859. [Google Scholar] [CrossRef]
  79. Lima, F.A.V.; Joventino, I.P.; Joventino, F.P.; de Almeida, A.C.; Neves, K.R.T.; do Carmo, M.R.; Leal, L.K.A.M.; de Andrade, G.M.; de Barros Viana, G.S. Neuroprotective activities of Spirulina platensis in the 6-OHDA model of Parkinson’s disease are related to its anti-inflammatory effects. Neurochem. Res. 2017, 42, 3390–3400. [Google Scholar] [CrossRef]
  80. Kamble, S.P.; Gaikar, R.B.; Padalia, R.B.; Shinde, K.D. Extraction and purification of C-phycocyanin from dry Spirulina powder and evaluating its antioxidant, anticoagulation and prevention of DNA damage activity. J. Appl. Pharm. Sci. 2013, 3, 149. [Google Scholar]
  81. Joventino, I.P.; Alves, H.G.; Neves, L.C.; Pinheiro-Joventino, F.; Leal, L.K.A.; Neves, S.A.; Ferreira, F.V.; Brito, G.A.C.; Viana, G.B. The microalga Spirulina platensis presents anti-inflammatory action as well as hypoglycemic and hypolipidemic properties in diabetic rats. J. Complementary Integr. Med. 2012, 9, 1–19. [Google Scholar] [CrossRef]
  82. Anbarasan, V.; Kumar, V.K.; Kumar, P.S.; Venkatachalam, T. In vitro evaluation of antioxidant activity of blue green algae Spirulina platensis. Int. J. Pharm. Sci. Res. 2011, 2, 2616. [Google Scholar]
  83. Romay, C.; Delgado, R.; Remirez, D.; Gonzalez, R.; Rojas, A. Effects of phycocyanin extract on tumor necrosis factor-α and nitrite levels in serum of mice treated with endotoxin. Arzneimittelforschung 2001, 51, 733–736. [Google Scholar] [CrossRef] [PubMed]
  84. Remirez, D.; Fernández, V.; Tapia, G.; González, R.; Videla, L.A. Influence of C-phycocyanin on hepatocellular parameters related to liver oxidative stress and Kupffer cell functioning. Inflamm. Res. 2002, 51, 351–356. [Google Scholar] [CrossRef] [PubMed]
  85. Patel, A.; Mishra, S.; Ghosh, P. Antioxidant potential of C-phycocyanin isolated from cyanobacterial species Lyngbya, Phormidium and Spirulina spp. Indian J. Biochem. Biophys. 2006, 43, 25–31. [Google Scholar] [PubMed]
  86. Cherng, S.C.; Cheng, S.N.; Tarn, A.; Chou, T.C. Anti-inflammatory activity of c-phycocyanin in lipopolysaccharide-stimulated RAW 264.7 macrophages. Life Sci. 2007, 81, 1431–1435. [Google Scholar] [CrossRef] [PubMed]
  87. Nasirian, F.; Dadkhah, M.; Moradi-Kor, N.; Obeidavi, Z. Effects of Spirulina platensis microalgae on antioxidant and anti-inflammatory factors in diabetic rats. Diabetes Metab. Syndr. Obes. Targets Ther. 2018, 11, 375. [Google Scholar] [CrossRef] [Green Version]
  88. Jensen, G.S.; Attridge, V.L.; Beaman, J.L.; Guthrie, J.; Ehmann, A.; Benson, K.F. Antioxidant and anti-inflammatory properties of an aqueous cyanophyta extract derived from arthrospira platensis: Contribution to bioactivities by the non-phycocyanin aqueous fraction. J. Med. Food 2015, 18, 535–541. [Google Scholar] [CrossRef] [Green Version]
  89. Gunes, S.; Tamburaci, S.; Dalay, M.C.; Deliloglu Gurhan, I. In vitro evaluation of Spirulina platensis extract incorporated skin cream with its wound healing and antioxidant activities. Pharm. Biol. 2017, 55, 1824–1832. [Google Scholar] [CrossRef] [Green Version]
  90. Romay, C.H.; Armesto, J.; Remirez, D.; González, R.; Ledon, N.; Garcia, I. Antioxidant and anti-inflammatory properties of C-phycocyanin from blue-green algae. Inflamm. Res. 1998, 47, 36–41. [Google Scholar] [CrossRef]
  91. Romay, C.; Ledon, N.; Gonzalez, R. Further studies on anti-inflammatory activity of phycocyanin in some animal models of inflammation. Inflamm. Res. 1998, 47, 334–338. [Google Scholar] [CrossRef]
  92. Gonzalez, R.; Rodriguez, S.; Romay, C.; González, A.; Armesto, J.; Remirez, D.; Merino, N. Anti-inflammatory activity of phycocyanin extract in acetic acid-induced colitis in rats. Pharmacol. Res. 1999, 39, 55–59. [Google Scholar] [CrossRef] [PubMed]
  93. Remirez, D.; Ledon, N.; González, R. Role of histamine in the inhibitory effects of phycocyanin in experimental models of allergic inflammatory response. Mediat. Inflamm. 2002, 11, 81–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Romay, C.H.; Gonzalez, R.; Ledon, N.; Remirez, D.; Rimbau, V. C-phycocyanin: A biliprotein with antioxidant, anti-inflammatory and neuroprotective effects. Curr. Protein Pept. Sci. 2003, 4, 207–216. [Google Scholar] [CrossRef]
  95. Khan, M.; Varadharaj, S.; Shobha, J.C.; Naidu, M.U.; Parinandi, N.L.; Kutala, V.K.; Kuppusamy, P. C-phycocyanin ameliorates doxorubicin-induced oxidative stress and apoptosis in adult rat cardiomyocytes. J. Cardiovasc. Pharmacol. 2006, 47, 9–20. [Google Scholar] [CrossRef] [PubMed]
  96. Shih, C.M.; Cheng, S.N.; Wong, C.S.; Kuo, Y.L.; Chou, T.C. Antiinflammatory and antihyperalgesic activity of C-phycocyanin. Anesth. Analg. 2009, 108, 1303–1310. [Google Scholar] [CrossRef]
  97. Manconia, M.; Pendás, J.; Ledón, N.; Moreira, T.; Sinico, C.; Saso, L.; Fadda, A.M. Phycocyanin liposomes for topical anti-inflammatory activity: In-vitro in-vivo studies. J. Pharm. Pharmacol. 2009, 61, 423–430. [Google Scholar] [CrossRef]
  98. Khan, M.; Varadharaj, S.; Ganesan, L.P.; Shobha, J.C.; Naidu, M.U.; Parinandi, N.L.; Tridandapani, S.; Kutala, V.K.; Kuppusamy, P. C-phycocyanin protects against ischemia-reperfusion injury of heart through involvement of p38 MAPK and ERK signaling. Am. J. Physiol. -Heart Circ. Physiol. 2006, 290, H2136–H2145. [Google Scholar] [CrossRef] [Green Version]
  99. Li, X.L.; Xu, G.; Chen, T.; Wong, Y.S.; Zhao, H.L.; Fan, R.R.; Gu, X.M.; Tong, P.C.; Chan, J.C. Phycocyanin protects INS-1E pancreatic beta cells against human islet amyloid polypeptide-induced apoptosis through attenuating oxidative stress and modulating JNK and p38 mitogen-activated protein kinase pathways. Int. J. Biochem. Cell Biol. 2009, 41, 1526–1535. [Google Scholar] [CrossRef]
  100. Sadek, K.M.; Lebda, M.A.; Nasr, S.M.; Shoukry, M. Spirulina platensis prevents hyperglycemia in rats by modulating gluconeogenesis and apoptosis via modification of oxidative stress and MAPK-pathways. Biomed. Pharmacother. 2017, 92, 1085–1094. [Google Scholar] [CrossRef]
  101. Schafer, F.Q.; Wang, H.P.; Kelley, E.E.; Cueno, K.L.; Buettner, S.M.A.G. Comparing β-carotene, vitamin E and nitric oxide as membrane antioxidants. Biol. Chem. 2002, 383, 671–681. [Google Scholar] [CrossRef]
  102. Bai, S.K.; Lee, S.J.; Na, H.J.; Ha, K.S.; Han, J.A.; Lee, H.; Kwon, Y.G.; Chung, C.K.; Kim, Y.M. β-Carotene inhibits inflammatory gene expression in lipopolysaccharide-stimulated macrophages by suppressing redox-based NF-κB activation. Exp. Mol. Med. 2005, 37, 323–334. [Google Scholar] [CrossRef] [PubMed]
  103. Katsuura, S.; Imamura, T.; Bando, N.; Yamanishi, R. β-Carotene and β-cryptoxanthin but not lutein evoke redox and immune changes in RAW264 murine macrophages. Mol. Nutr. Food Res. 2009, 53, 1396–1405. [Google Scholar] [CrossRef] [PubMed]
  104. Strasky, Z.; Zemankova, L.; Nemeckova, I.; Rathouska, J.; Wong, R.J.; Muchova, L.; Subhanova, I.; Vanikova, J.; Vanova, K.; Vitek, L.; et al. Spirulina platensis and phycocyanobilin activate atheroprotective heme oxygenase-1: A possible implication for atherogenesis. Food Funct. 2013, 4, 1586–1594. [Google Scholar] [CrossRef] [PubMed]
  105. Jensen, G.S.; Drapeau, C.; Lenninger, M.; Benson, K.F. Clinical safety of a high dose of Phycocyanin-enriched aqueous extract from Arthrospira (Spirulina) platensis: Results from a randomized, double-blind, placebo-controlled study with a focus on anticoagulant activity and platelet activation. J. Med. Food 2016, 19, 645–653. [Google Scholar] [CrossRef] [Green Version]
  106. Piovan, A.; Battaglia, J.; Filippini, R.; Dalla Costa, V.; Facci, L.; Argentini, C.; Pagetta, A.; Giusti, P.; Zusso, M. Pre-and Early Post-treatment With Arthrospira platensis (Spirulina) Extract Impedes Lipopolysaccharide-triggered Neuroinflammation in Microglia. Front. Pharmacol. 2021, 12, 1–10. [Google Scholar] [CrossRef]
  107. Eggleton, J.S.; Nagalli, S. Highly Active Antiretroviral Therapy (HAART); StatPearls Publishing: Treasure Island, FL, USA, 2020. [Google Scholar]
  108. Chang, J.L.; Tsai, A.C.; Musinguzi, N.; Haberer, J.E.; Boum, Y.; Muzoora, C.; Bwana, M.; Martin, J.N.; Hunt, P.W.; Bangsberg, D.R.; et al. Depression and suicidal ideation among HIV-infected adults receiving efavirenz versus nevirapine in Uganda: A prospective cohort study. Ann. Intern. Med. 2018, 169, 146–155. [Google Scholar] [CrossRef]
  109. Lyseng-Williamson, K.A.; Reynolds, N.A.; Plosker, G.L. Tenofovir disoproxil fumarate. Drugs 2005, 65, 413–432. [Google Scholar] [CrossRef]
  110. Chen, Y.F.; Stampley, J.E.; Irving, B.A.; Dugas, T.R. Chronic nucleoside reverse transcriptase inhibitors disrupt mitochondrial homeostasis and promote premature endothelial senescence. Toxicol. Sci. 2019, 172, 445–456. [Google Scholar] [CrossRef]
  111. Martin, A.M.; Nolan, D.; Gaudieri, S.; Almeida, C.A.; Nolan, R.; James, I.; Carvalho, F.; Phillips, E.; Christiansen, F.T.; Purcell, A.W.; et al. Predisposition to abacavir hypersensitivity conferred by HLA-B * 5701 and a haplotypic Hsp70-Hom variant. Proc. Natl. Acad. Sci. USA 2004, 101, 4180–4185. [Google Scholar] [CrossRef] [Green Version]
  112. Kakuda, T. Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin. Ther. 2000, 22, 685–708. [Google Scholar] [CrossRef]
  113. Günthard, H.F.; Saag, M.S.; Benson, C.A.; Del Rio, C.; Eron, J.J.; Gallant, J.E.; Hoy, J.F.; Mugavero, M.J.; Sax, P.E.; Thompson, M.A.; et al. Antiretroviral drugs for treatment and prevention of HIV infection in adults: 2018 recommendations of the International Antiviral Society–USA Panel. JAMA 2018, 320, 379–396. [Google Scholar]
  114. Yeni, P. Update on HAART in HIV. J. Hepatol. 2006, 44, S100–S103. [Google Scholar] [CrossRef] [PubMed]
  115. Eastone, J.A.; Decker, C.F. New-onset diabetes mellitus associated with use of protease inhibitor. Ann. Intern. Med. 1997, 127, 948. [Google Scholar] [CrossRef] [PubMed]
  116. Soliman, E.Z.; Lundgren, J.; Roediger, M.P.; Duprez, D.A.; Temesgen, Z.; Bickel, M.; Shlay, J.C.; Somboonwit, C.; Reiss, P.; Stein, J.; et al. Boosted protease inhibitors and the electrocardiographic measures of QT and PR durations. AIDS 2011, 25, 367–377. [Google Scholar] [CrossRef] [Green Version]
  117. Landovitz, R.J.; Ribaudo, H.J.; Ofotokun, I.; Na, L.H.; Godfrey, C.; Kuritzkes, D.R.; Sagar, M.; Brown, T.T.; Cohn, S.E.; McComsey, G.A.; et al. Efficacy and tolerability of 3 nonnucleoside reverse transcriptase inhibitor–sparing antiretroviral regimens for treatment-naive volunteers infected with HIV-1: A randomized, controlled equivalence trial. Ann. Intern. Med. 2014, 161, 461–471. [Google Scholar]
  118. Gutiérrez, F.; Fulladosa, X.; Barril, G.; Domingo, P. Renal tubular transporter-mediated interactions of HIV drugs: Implications for patient management. AIDS Rev. 2014, 16, 199–212. [Google Scholar]
  119. Hoffmann, C.; Welz, T.; Sabranski, M.; Kolb, M.; Wolf, E.; Stellbrink, H.J.; Wyen, C. Higher rates of neuropsychiatric adverse events leading to dolutegravir discontinuation in women and older patients. HIV Med. 2017, 18, 56–63. [Google Scholar] [CrossRef]
  120. Fettiplace, A.; Stainsby, C.; Winston, A.; Givens, N.; Puccini, S.; Vannappagari, V.; Hsu, R.; Fusco, J.; Quercia, R.; Aboud, M.; et al. Psychiatric Symptoms in Patients Receiving Dolutegravir. JAIDS J. Acquir. Immune Defic. Syndr. 2017, 74, 423–431. [Google Scholar] [CrossRef] [Green Version]
  121. Zash, R.; Makhema, J.; Shapiro, R.L. Neural-Tube Defects with Dolutegravir Treatment from the Time of Conception. N. Engl. J. Med. 2018, 379, 979–981. [Google Scholar] [CrossRef]
  122. Schafer, J.J.; E Squires, K. Integrase Inhibitors: A Novel Class of Antiretroviral Agents. Ann. Pharmacother. 2010, 44, 145–156. [Google Scholar] [CrossRef]
  123. Hazuda, D.J.; Felock, P.; Witmer, M.; Wolfe, A.; Stillmock, K.; Grobler, J.A.; Espeseth, A.; Gabryelski, L.; Schleif, W.; Blau, C.; et al. Inhibitors of Strand Transfer That Prevent Integration and Inhibit HIV-1 Replication in Cells. Science 2000, 287, 646–650. [Google Scholar] [CrossRef] [PubMed]
  124. Hardy, H.; Skolnik, P.R. Enfuvirtide, a New Fusion Inhibitor for Therapy of Human Immunodeficiency Virus Infection. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2004, 24, 198–211. [Google Scholar] [CrossRef] [PubMed]
  125. Dorr, P.; Westby, M.; Dobbs, S.; Griffin, P.; Irvine, B.; Macartney, M.; Mori, J.; Rickett, G.; Smith-Burchnell, C.; Napier, C.; et al. Maraviroc (UK-427,857), a Potent, Orally Bioavailable, and Selective Small-Molecule Inhibitor of Chemokine Receptor CCR5 with Broad-Spectrum Anti-Human Immunodeficiency Virus Type 1 Activity. Antimicrob. Agents Chemother. 2005, 49, 4721–4732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Miao, M.; De Clercq, E.; Li, G. Clinical significance of chemokine receptor antagonists. Expert Opin. Drug Metab. Toxicol. 2020, 16, 11–30. [Google Scholar] [CrossRef] [PubMed]
  127. De Luca, A.; Pezzotti, P.; Boucher, C.; Döring, M.; Incardona, F.; Kaiser, R.; Lengauer, T.; Pfeifer, N.; Schülter, E.; Vandamme, A.-M.; et al. Clinical use, efficacy, and durability of maraviroc for antiretroviral therapy in routine care: A European survey. PLoS ONE 2019, 14, e0225381. [Google Scholar] [CrossRef]
  128. Shafer, R.; Vuitton, D. Highly active antiretroviral therapy (HAART) for the treatment of infection with human immunodeficiency virus type 1. Biomed. Pharmacother. 1999, 53, 73–86. [Google Scholar] [CrossRef]
  129. Kitahata, M.M.; Koepsell, T.D.; Deyo, R.A.; Maxwell, C.L.; Dodge, W.T.; Wagner, E.H. Physicians’ experience with the acquired immunodeficiency syndrome as a factor in patients’ survival. N. Engl. J. Med. 1996, 334, 701–707. [Google Scholar] [CrossRef]
  130. Cunningham, W.E.; Tisnado, D.M.; Lui, H.H.; Nakazono, T.T.; Carlisle, D.M. The effect of hospital experience on mortality among patients hospitalized with acquired immunodeficiency syndrome in California. Am. J. Med. 1999, 107, 137–143. [Google Scholar] [CrossRef]
  131. Rackal, J.M.; Tynan, A.-M.; Handford, C.D.; Rzeznikiewiz, D.; Agha, A.; Glazier, R. Provider training and experience for people living with HIV/AIDS. Cochrane Database Syst. Rev. 2011, 6, CD003938. [Google Scholar] [CrossRef] [Green Version]
  132. Ford, N.; Migone, C.; Calmy, A.; Kerschberger, B.; Kanters, S.; Nsanzimana, S.; Mills, E.J.; Meintjes, G.; Vitoria, M.; Doherty, M.; et al. Benefits and risks of rapid initiation of antiretroviral therapy. AIDS 2018, 32, 17–23. [Google Scholar] [CrossRef] [Green Version]
  133. Group, I.S.S. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N. Engl. J. Med. 2015, 373, 795–807. [Google Scholar]
  134. Thompson, M.A.; Aberg, J.A.; Hoy, J.F.; Telenti, A.; Benson, C.; Cahn, P.; Eron, J.J.; Günthard, H.F.; Hammer, S.M.; Reiss, P.; et al. Antiretroviral treatment of adult HIV infection: 2012 recommendations of the International Antiviral Society–USA panel. Jama 2012, 308, 387–402. [Google Scholar] [CrossRef] [PubMed]
  135. Sax, P.E.; Tierney, C.; Collier, A.C.; Fischl, M.A.; Mollan, K.; Peeples, L.; Godfrey, C.; Jahed, N.C.; Myers, L.; Katzenstein, D.; et al. Abacavir–lamivudine versus tenofovir–emtricitabine for initial HIV-1 therapy. N. Engl. J. Med. 2009, 361, 2230–2240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Post, F.A.; Moyle, G.J.; Stellbrink, H.J.; Domingo, P.; Podzamczer, D.; Fisher, M.; Norden, A.G.; Cavassini, M.; Rieger, A.; Khuong-Josses, M.A.; et al. Randomized comparison of renal effects, efficacy, and safety with once-daily abacavir/lamivudine versus tenofovir/emtricitabine, administered with efavirenz, in antiretroviral-naive, HIV-1–infected adults: 48-week results from the ASSERT study. JAIDS J. Acquir. Immune Defic. Syndr. 2010, 55, 49–57. [Google Scholar] [CrossRef]
  137. Glover, M.; Hebert, V.Y.; Nichols, K.; Xue, S.Y.; Thibeaux, T.M.; Zavecz, J.A.; Dugas, T.R. Overexpression of mitochondrial antioxidant manganese superoxide dismutase (MnSOD) provides protection against AZT- or 3TC-induced endothelial dysfunction. Antivir. Res. 2014, 111, 136–142. [Google Scholar] [CrossRef] [Green Version]
  138. Brown, L.; Jin, J.; Ferrell, D.; Sadic, E.; Obregon, D.; Smith, A.J.; Tan, J.; Giunta, B. Efavirenz Promotes β-Secretase Expression and Increased Aβ1-40,42 via Oxidative Stress and Reduced Microglial Phagocytosis: Implications for HIV Associated Neurocognitive Disorders (HAND). PLoS ONE 2014, 9, e95500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Canale, D.; De Bragança, A.C.; Gonçalves, J.G.; Shimizu, M.H.M.; Sanches, T.R.; Andrade, L.; Volpini, R.A.; Seguro, A.C. Vitamin D Deficiency Aggravates Nephrotoxicity, Hypertension and Dyslipidemia Caused by Tenofovir: Role of Oxidative Stress and Renin-Angiotensin System. PLoS ONE 2014, 9, e103055. [Google Scholar] [CrossRef]
  140. Venhoff, N.; Setzer, B.; Melkaoui, K.; A Walker, U. Mitochondrial Toxicity of Tenofovir, Emtricitabine and Abacavir Alone and in Combination with Additional Nucleoside Reverse Transcriptase Inhibitors. Antivir. Ther. 2007, 12, 1075–1086. [Google Scholar] [CrossRef]
  141. Bonomini, F.; Rodella, L.F.; Rezzani, R. Metabolic Syndrome, Aging and Involvement of Oxidative Stress. Aging Dis. 2015, 6, 109–120. [Google Scholar] [CrossRef] [Green Version]
  142. Dos Santos, W.; Paes, P.; dos Santos, A.; Machado, D.; Navarro, A.; Frenandes, A. Impact of progressive resistance training in Brazilian HIV patients with lipodystrophy. J. AIDS Clin. Res. 2013, 4, 2. [Google Scholar] [CrossRef]
  143. Sharma, B. Anti-HIV-1 drug toxicity and management strategies. Neurobehav. HIV Med. 2011, 3, 27–40. [Google Scholar] [CrossRef] [Green Version]
  144. Csányi, G. Oxidative Stress in Cardiovascular Disease; Multidisciplinary Digital Publishing Institute: Basel, Switzerland, 2014. [Google Scholar]
  145. McColl, D.J.; Margot, N.; Chen, S.-S.; Harris, J.; Borroto-Esoda, K.; Miller, M.D. Reduced Emergence of the M184V/I Resistance Mutation When Antiretroviral-Naïve Subjects Use Emtricitabine Versus Lamivudine in Regimens Composed of Two NRTIs Plus the NNRTI Efavirenz. HIV Clin. Trials 2011, 12, 61–70. [Google Scholar] [CrossRef] [PubMed]
  146. Hong, S.Y.; Jonas, A.; DeKlerk, M.; Shiningavamwe, A.; Desta, T.; Badi, A.; Morris, L.; Hunt, G.M.; Ledwaba, J.; Sheehan, H.B.; et al. Population-Based Surveillance of HIV Drug Resistance Emerging on Treatment and Associated Factors at Sentinel Antiretroviral Therapy Sites in Namibia. JAIDS J. Acquir. Immune Defic. Syndr. 2015, 68, 463–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Gulick, R.M.; Ribaudo, H.J.; Shikuma, C.M.; Lustgarten, S.; Squires, K.E.; Meyer, W.A., III; Acosta, E.P.; Schackman, B.R.; Pilcher, C.D.; Murphy, R.L.; et al. Triple-nucleoside regimens versus efavirenz-containing regimens for the initial treatment of HIV-1 infection. N. Engl. J. Med. 2004, 350, 1850–1861. [Google Scholar] [CrossRef] [PubMed]
  148. Robbins, G.K.; De Gruttola, V.; Shafer, R.W.; Smeaton, L.M.; Snyder, S.W.; Pettinelli, C.; Dubé, M.P.; Fischl, M.A.; Pollard, R.B.; Delapenha, R.; et al. Comparison of sequential three-drug regimens as initial therapy for HIV-1 infection. N. Engl. J. Med. 2003, 349, 2293–2303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Rakhmanina, N.Y.; la Porte, C.J. Therapeutic drug monitoring of antiretroviral drugs in the management of human immunodeficiency virus infection. Ther. Drug Monit. Newer Drugs Biomark. 2012, 373, 374–389. [Google Scholar]
  150. EACS. European AIDS Clinical Society Guidelines. Amsterdam, The Netherlands, Version 6. 2011. Available online: https://www.eacsociety.org/media/2011_eacsguidelines-v6.0-english_oct.pdf (accessed on 6 December 2021).
  151. Gilks, C.F.; Crowley, S.; Ekpini, R.; Gove, S.; Perriens, J.; Souteyrand, Y.; Sutherland, D.; Vitoria, M.; Guerma, T.; De Cock, K. The WHO public-health approach to antiretroviral treatment against HIV in resource-limited settings. Lancet 2006, 368, 505–510. [Google Scholar] [CrossRef]
  152. Kahana, S.Y.; Rohan, J.; Allison, S.; Frazier, T.W.; Drotar, D. A Meta-Analysis of Adherence to Antiretroviral Therapy and Virologic Responses in HIV-Infected Children, Adolescents, and Young Adults. AIDS Behav. 2012, 17, 41–60. [Google Scholar] [CrossRef]
  153. Sliwa, K.; Hilfiker-Kleiner, D.; Petrie, M.C.; Mebazaa, A.; Pieske, B.; Buchmann, E.; Regitz-Zagrosek, V.; Schaufelberger, M.; Tavazzi, L.; van Veldhuisen, D.J.; et al. Current state of knowledge on aetiology, diagnosis, management, and therapy of peripartum cardiomyopathy: A position statement from the Heart Failure Association of the European Society of Cardiology Working Group on peripartum cardiomyopathy. Eur. J. Hear. Fail. 2010, 12, 767–778. [Google Scholar] [CrossRef] [Green Version]
  154. Schinazi, R.F.; Lloyd, R.M.; Nguyen, M.H.; Cannon, D.L.; McMillan, A.; Ilksoy, N.; Chu, C.K.; Liotta, D.C.; Bazmi, H.Z.; Mellors, J.W. Characterization of human immunodeficiency viruses resistant to oxathiolane-cytosine nucleosides. Antimicrob. Agents Chemother. 1993, 37, 875–881. [Google Scholar] [CrossRef] [Green Version]
  155. Maserati, R.; De Silvestri, A.; Uglietti, A.; Colao, G.; Di Biagio, A.; Bruzzone, B.; Di Pietro, M.; Re, M.C.; Tinelli, C.; Zazzi, M. Emerging mutations at virological failure of HAART combinations containing tenofovir and lamivudine or emtricitabine. AIDS 2010, 24, 1013–1018. [Google Scholar] [CrossRef] [PubMed]
  156. Schinazi, R.F.; McMillan, A.; Cannon, D.; Mathis, R.; Lloyd, R.M.; Peck, A.; Sommadossi, J.P.; Clair, M.S.; Wilson, J.; A Furman, P. Selective inhibition of human immunodeficiency viruses by racemates and enantiomers of cis-5-fluoro-1-[2-(hydroxymethyl)-1,3-oxathiolan-5-yl]cytosine. Antimicrob. Agents Chemother. 1992, 36, 2423–2431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Margot, N.A.; Enejosa, J.; Cheng, A.K.; Miller, M.D.; McColl, D.J. Development of HIV-1 drug resistance through 144 weeks in antiretroviral-naive subjects on emtricitabine, tenofovir disoproxil fumarate, and efavirenz compared with lamivudine/zidovudine and efavirenz in study GS-01-934. JAIDS J. Acquir. Immune Defic. Syndr. 2009, 52, 209–221. [Google Scholar] [CrossRef] [PubMed]
  158. Mugwanya, K.K.; John-Stewart, G.; Baeten, J. Safety of oral tenofovir disoproxil fumarate-based HIV pre-exposure prophylaxis use in lactating HIV-uninfected women. Expert Opin. Drug Saf. 2017, 16, 867–871. [Google Scholar] [CrossRef] [PubMed]
  159. Spinner, C.D.; Boesecke, C.; Zink, A.; Jessen, H.; Stellbrink, H.J.; Rockstroh, J.K.; Esser, S. HIV pre-exposure prophylaxis (PrEP): A review of current knowledge of oral systemic HIV PrEP in humans. Infection 2016, 44, 151–158. [Google Scholar] [CrossRef] [PubMed]
  160. Fonner, V.A.; Dalglish, S.L.; Kennedy, C.E.; Baggaley, R.; O’Reilly, K.R.; Koechlin, F.M.; Rodolph, M.; Hodges-Mameletzis, I.; Grant, R. Effectiveness and safety of oral HIV preexposure prophylaxis for all populations. AIDS 2016, 30, 1973–1983. [Google Scholar] [CrossRef] [Green Version]
  161. Borroto-Esoda, K.; E Vela, J.; Myrick, F.; Ray, A.S.; Miller, M.D. In Vitro Evaluation of the Anti-HIV Activity and Metabolic Interactions of Tenofovir and Emtricitabine. Antivir. Ther. 2005, 11, 377–384. [Google Scholar] [CrossRef]
  162. De Ruiter, A.; Taylor, G.P.; Clayden, P.; Dhar, J.; Gandhi, K.; Gilleece, Y.; Harding, K.; Hay, P.; Kennedy, J.; Low-Beer, N.; et al. British HIV Association guidelines for the management of HIV infection in pregnant women 2012 (2014 interim review). HIV Med. 2014, 15, 1–77. [Google Scholar]
  163. Law, W.P.; Duncombe, C.J.; Mahanontharit, A.; Boyd, M.; Ruxrungtham, K.; Lange, J.M.A.; Phanuphak, P.; Cooper, D.A.; Dore, G.J. Impact of viral hepatitis co-infection on response to antiretroviral therapy and HIV disease progression in the HIV-NAT cohort. AIDS 2004, 18, 1169–1177. [Google Scholar] [CrossRef] [Green Version]
  164. Zdanowicz, M.M. The Pharmacology of HIV Drug Resistance. Am. J. Pharm. Educ. 2006, 70, 1–8. [Google Scholar] [CrossRef]
  165. Hare, C.B.; Mellors, J.; Krambrink, A.; Su, Z.; Skiest, D.; Margolis, D.M.; Patel, S.S.; Barnas, D.; Frenkel, L.; Coombs, R.W.; et al. Detection of nonnucleoside reverse-transcriptase inhibitor-resistant HIV-1 after discontinuation of virologically suppressive antiretroviral therapy. Clin. Infect. Dis. 2008, 47, 421–424. [Google Scholar] [CrossRef] [PubMed]
  166. Carr, A.; Samaras, K.; Chisholm, D.J.; A Cooper, D. Pathogenesis of HIV-1-protease inhibitor-associated peripheral lipodystrophy, hyperlipidaemia, and insulin resistance. Lancet 1998, 351, 1881–1883. [Google Scholar] [CrossRef]
  167. Del Valle, L.G.; Hernández, R.G.; Ávila, J.P. Oxidative stress associated to disease progression and toxicity during antiretroviral therapy in human immunodeficiency virus infection. J. Virol. Microbiol. 2013, 13, 15. [Google Scholar]
  168. Awodele, O.; O Olayemi, S.; A Nwite, J.; A Adeyemo, T. Investigation of the levels of oxidative stress parameters in HIV and HIV-TB co-infected patients. J. Infect. Dev. Ctries. 2011, 6, 79–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Pinti, M.; Nasi, M.; Gibellini, L.; Roat, E.; De Biasi, S.; Bertoncelli, L.; Cossarizza, A. The Role of Mitochondria in HIV Infection and Its Treatment. J. Exp. Clin. Med. 2010, 2, 145–155. [Google Scholar] [CrossRef]
  170. Mandas, A.; Iorio, E.L.; Congiu, M.G.; Balestrieri, C.; Mereu, A.; Cau, D.; Dessì, S.; Curreli, N. Oxidative Imbalance in HIV-1 Infected Patients Treated with Antiretroviral Therapy. J. Biomed. Biotechnol. 2009, 2009, 1–7. [Google Scholar] [CrossRef] [PubMed]
  171. Apostolova, N.; Gomez-Sucerquia, L.; Moran, A.; Alvarez, A.; Blas-Garcia, A.; Esplugues, J. Enhanced oxidative stress and increased mitochondrial mass during Efavirenz-induced apoptosis in human hepatic cells. J. Cereb. Blood Flow Metab. 2010, 160, 2069–2084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Estrada, V.; Monge, S.; Garre, G.; Sobrino, P.; Masiá, M.; Berenguer, J.; Portilla, J.; Viladés, C.; Martinez, E.; Blanco, J.; et al. Relationship between plasma bilirubin level and oxidative stress markers in HIV-infected patients on atazanavir-vs. efavirenz-based antiretroviral therapy. HIV Med. 2016, 17, 653–661. [Google Scholar] [CrossRef] [Green Version]
  173. Elias, A.; Ijeoma, O.; Edikpo, N.J.; Oputiri, D.; Geoffrey, O.-B.P. Tenofovir Renal Toxicity: Evaluation of Cohorts and Clinical Studies—Part One. Pharmacol. Pharm. 2013, 4, 651–662. [Google Scholar] [CrossRef]
  174. Fernandez-Fernandez, B.; Montoya-Ferrer, A.; Sanz, A.B.; Sanchez-Niño, M.D.; Izquierdo, M.C.; Poveda, J.; Sainz-Prestel, V.; Ortiz-Martin, N.; Parra-Rodriguez, A.; Selgas, R.; et al. Tenofovir Nephrotoxicity: 2011 Update. AIDS Res. Treat. 2011, 2011, 1–11. [Google Scholar] [CrossRef]
  175. Shafi, T.; Choi, M.; Racusen, L.; Spacek, L.; Berry, C.; Atta, M.; Fine, D. Ritonavir-induced acute kidney injury: Kidney biopsy findings and review of literature. Clin. Nephrol. 2011, 75, 60–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Boffito, M. From concept to care: Pharmacokinetic boosting of protease inhibitors. PRN Noteb. 2004, 9, 15–18. [Google Scholar]
  177. Patel, A.K.; Ranjan, R.R.; Patel, A.R.; Patel, J.K.; Patel, K.K. Tenofovir-associated renal dysfunction in clinical practice: An observational cohort from western India. Indian J. Sex. Transm. Dis. AIDS 2010, 31, 30–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Cao, Y.; Han, Y.; Xie, J.; Cui, Q.; Zhang, L.; Li, Y.; Li, Y.; Song, X.; Zhu, T.; Li, T. Impact of a tenofovir disoproxil fumarate plus ritonavir-boosted protease inhibitor-based regimen on renal function in HIV-infected individuals: A prospective, multicenter study. BMC Infect. Dis. 2013, 13, 301–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Grigsby, I.F.; Pham, L.; Mansky, L.M.; Gopalakrishnan, R.; Mansky, K.C. Tenofovir-associated bone density loss. Ther. Clin. Risk Manag. 2010, 6, 41. [Google Scholar]
  180. Moss, D.M.; Neary, M.; Owen, A. The role of drug transporters in the kidney: Lessons from tenofovir. Front. Pharmacol. 2014, 5, 248. [Google Scholar] [CrossRef] [Green Version]
  181. Yeligar, S.M.; Guidot, D.M.; Brown, L.A.S.; Cribbs, S.K. HIV Infection Induces NADPH Oxidase Expression in Alveolar Macrophages of Otherwise Healthy Subjects. In C55 HIV-Associated Lung Diseases and Infections; American Thoracic Society: New York, NY, USA, 2015; p. A4720. [Google Scholar]
  182. Wang, X.; Liao, D.; Lin, P.H.; Yao, Q.; Chen, C. Highly active antiretroviral therapy drugs inhibit in vitro cholesterol efflux from human macrophage-derived foam cells. Lab. Investig. 2009, 89, 1355–1363. [Google Scholar] [CrossRef] [Green Version]
  183. So-Armah, K.; Freiberg, M.S. HIV and Cardiovascular Disease: Update on Clinical Events, Special Populations, and Novel Biomarkers. Curr. HIV/AIDS Rep. 2018, 15, 233–244. [Google Scholar] [CrossRef]
  184. Brouwer, E.S.; Napravnik, S.; Eron, J.J.; Stalzer, B.; Floris-Moore, M.; Simpson, R.J.; Stürmer, T. Effects of Combination Antiretroviral Therapies on the Risk of Myocardial Infarction Among HIV Patients. Epidemiology 2014, 25, 406–417. [Google Scholar] [CrossRef] [Green Version]
  185. Marcel, A.K.; Ekali, L.G.; Eugene, S.; Arnold, O.E.; Sandrine, E.D.; Von der Weid, D.; Gbaguidi, E.; Ngogang, J.; Mbanya, J.C. The effect of Spirulina platensis versus soybean on insulin resistance in HIV-infected patients: A randomized pilot study. Nutrients 2011, 3, 712–724. [Google Scholar] [CrossRef] [Green Version]
  186. Winter, F.S.; Emakam, F.; Kfutwah, A.; Hermann, J.; Azabji-Kenfack, M.; Krawinkel, M.B. The Effect of Arthrospira platensis Capsules on CD4 T-Cells and Antioxidative Capacity in a Randomized Pilot Study of Adult Women Infected with Human Immunodeficiency Virus Not under HAART in Yaoundé, Cameroon. Nutrients 2014, 6, 2973–2986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Ngo-Matip, M.-E.; Pieme, C.A.; Azabji-Kenfack, M.; Biapa, P.C.N.; Germaine, N.; Heike, E.; Moukette, B.M.; Emmanuel, K.; Philippe, S.; Mbofung, C.M.; et al. Effects of Spirulina platensis supplementation on lipid profile in HIV–infected antiretroviral naïve patients in Yaounde-Cameroon: A randomized trial study. Lipids Health Dis. 2014, 13, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Ngo-Matip, M.E.; Pieme, C.A.; Azabji-Kenfack, M.; Moukette, B.M.; Korosky, E.; Stefanini, P.; Ngogang, J.Y.; Mbofung, C.M. Impact of daily supplementation of Spirulina platensis on the immune system of naïve HIV-1 patients in Cameroon: A 12-months single blind, randomized, multicenter trial. Nutr. J. 2015, 14, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Koskey, S.; Okoth, F.; Waihenya, R. Monitoring of CD4+ T-cell counts in HIV infected patients on Arthrospira platensis supplement in Kisumu, Kenya. Afr. J. Health Sci. 2013, 24, 1–8. [Google Scholar]
  190. Moor, V.J.A.; Pieme, C.A.; Nkeck, J.R.; Nya, P.C.B.; Mondinde, G.I.; Amazia, F.; Kouanfack, C.; Assoumou, M.C.O.; Ngogang, J. Effects of Spirulina platensis on the Immune Status, Inflammatory and Oxidative Markers of HIV Patients on Antiretroviral Therapy in Cameroon. Acta Sci. Pharm. Sci. 2021, 5, 50–57. [Google Scholar] [CrossRef]
  191. Antonelli, M.; Donelli, D. Effects of Spirulina on CD4+ T-Lymphocyte Count in Patients with HIV Infection: A Literature Review. Biol. Life Sci. Forum 2022, 12, 6. [Google Scholar] [CrossRef]
Figure 1. Diagrammatic representation of Spirulina reduction of oxidative stress via several pathways. Spirulina inhibits NADPH oxidase, reduces ROS, blocks FR-induced apoptosis, and promotes mitochondrial health (Created with BioRender.com, accessed on 17 November 2021).
Figure 1. Diagrammatic representation of Spirulina reduction of oxidative stress via several pathways. Spirulina inhibits NADPH oxidase, reduces ROS, blocks FR-induced apoptosis, and promotes mitochondrial health (Created with BioRender.com, accessed on 17 November 2021).
Nutrients 14 03076 g001
Figure 2. The 2D chemical structures of phycocyanin, phycocyanobilin, bilirubin, and biliverdin (prepared by author using maestro 11.2).
Figure 2. The 2D chemical structures of phycocyanin, phycocyanobilin, bilirubin, and biliverdin (prepared by author using maestro 11.2).
Nutrients 14 03076 g002
Figure 3. Antioxidant and anti-inflammatory effects of SP (created with BioRender.com, accessed on 17 November 2021).
Figure 3. Antioxidant and anti-inflammatory effects of SP (created with BioRender.com, accessed on 17 November 2021).
Nutrients 14 03076 g003
Table 1. HAART drugs mechanism and their adverse effects.
Table 1. HAART drugs mechanism and their adverse effects.
HAART MechanismExampleAdverse Effect
Nucleoside/Nucleotide reverse transcriptase inhibitors (NRTIs)NRTIs require intracellular phosphorylation via host enzymes before they can inhibit viral replication. These agents are nucleoside or nucleotide analogs with an absent hydroxyl at the 3′ end that are incorporated into the growing viral DNA strand. They competitively bind to reverse transcriptase and cause premature DNA chain termination as they inhibit 3′ to 5′ phosphodiester bond formation.abacavir, didanosine, lamivudine, stavudine, tenofovir, emtricitabine, atazanavir, and zidovudineMitochondrial toxicity, bone marrow suppression, anemia, and lipodystrophy.
NRTIs inhibit mitochondrial DNA polymerase.
Tenofovir may cause kidney injury or decreased bone mineral density or osteoporosis.
Abacavir is associated with a CD8-mediated hypersensitivity reaction in patients with the HLA-B*5701 mutation.
Didanosine is associated with a high risk of pancreatitis and hepatomegaly.
Non-nucleoside reverse transcriptase inhibitors (NNRTIs) NNRTIs bind to HIV reverse transcriptase at an allosteric, hydrophobic site. These agents cause a stereochemical change within reverse transcriptase, thus inhibiting nucleoside binding and inhibition of DNA polymerase.delavirdine, efavirenz, nevirapine, rilpivirineTemporary rashes but may progress to Stevens–Johnson’s syndrome.
Hepatitis may progress to liver failure.
Efavirenz may cause teratogenicity
Risk of neural tube defects.
NNRTIs (mostly rilpivirine) may result in QT prolongation.
Numerous interactions with hepatic cytochrome P450 enzymes.
Efavirenz is linked to various psychiatric and CNS effects, including, but not limited to: vivid dreams, delusions, sleep disturbances, dizziness, headaches, increased suicidality, psychosis-like behavior, and mania.
Protease inhibitors (PIs)PIs competitively inhibit the proteolytic cleavage of the gag/pol polyproteins in HIV-infected cells. These agents result in immature, noninfectious virions. PIs are administered with boosting agents such as ritonavir or cobicistat to patients that are failing their initial HAART combination. atazanavir, darunavir, indinavirHepatotoxicity, insulin resistance, hyperglycemia, hyperlipidemia, lipodystrophy, and PR interval prolongation.
Other PIs are inefficient and have a high resistance and increased risk of nephrolithiasis, hence indinavir and saquinavir are no longer used.
Integrase strand transfer inhibitors (INSTIs)Integrase inhibitors bind viral integrase and prevent viral DNA from being incorporated into the host cell chromosome.dolutegravir, elvitegravir, raltegravirSome patients may experience dizziness, sleep disturbances, or depression.
Raltegravir and dolutegravir can cause rhabdomyolysis and myopathy.
Dolutegravir can block the secretion of creatinine and occasionally cause a decrease in the GFR. It can also have interactions with several medications, including those that inhibit/induce CYP3A4 enzymes, metformin, rifampin, and antiepileptics.
Fusion inhibitors (FIs) Fusion inhibitors bind to the envelope glycoprotein gp41 and prevent viral fusion to the CD4 T-cells.enfuvirtideEnfuvirtide is generally well tolerated though some patients may experience injection site reactions.
Chemokine receptor antagonists (CCR5 antagonists) CCR5 antagonists selectively and reversibly block entry into the CD4 T-cells by preventing interaction between CD4 cells and the gp120 subunit of the viral envelope glycoprotein.maravirocSome patients may experience dizziness or skin rashes.
There is a risk of hepatotoxicity with allergic features, high risk of hepatic dysfunction. Drug–drug interactions should be a consideration if patients are taking concurrent CYP3A4 inhibitors or inducers.
Abbreviations: HLA—human leucocyte antigen; CNS—central nervous system; GFR—glomerular filtration rate.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sibiya, T.; Ghazi, T.; Chuturgoon, A. The Potential of Spirulina platensis to Ameliorate the Adverse Effects of Highly Active Antiretroviral Therapy (HAART). Nutrients 2022, 14, 3076. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14153076

AMA Style

Sibiya T, Ghazi T, Chuturgoon A. The Potential of Spirulina platensis to Ameliorate the Adverse Effects of Highly Active Antiretroviral Therapy (HAART). Nutrients. 2022; 14(15):3076. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14153076

Chicago/Turabian Style

Sibiya, Thabani, Terisha Ghazi, and Anil Chuturgoon. 2022. "The Potential of Spirulina platensis to Ameliorate the Adverse Effects of Highly Active Antiretroviral Therapy (HAART)" Nutrients 14, no. 15: 3076. https://0-doi-org.brum.beds.ac.uk/10.3390/nu14153076

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