Open access peer-reviewed chapter

Challenges in Platelet Functions in HIV/AIDS Management

Written By

Gordon Ogweno

Submitted: 02 June 2022 Reviewed: 07 June 2022 Published: 22 July 2022

DOI: 10.5772/intechopen.105731

From the Edited Volume

Future Opportunities and Tools for Emerging Challenges for HIV/AIDS Control

Edited by Samuel Okware

Chapter metrics overview

263 Chapter Downloads

View Full Metrics

Abstract

The interest in platelet functions in HIV/AIDS is due to the high incidence of microvascular thrombosis in these individuals. A lot of laboratory data have been generated regarding platelet functions in this population. The tests demonstrate platelet hyperactivity but decreased aggregation, though results are inconsistent depending on the study design. Antiretroviral treatments currently in use display complex interactions. Many studies on platelet functions in these patients have been for research purposes, but none have found utility in guiding drug treatment of thrombosis.

Keywords

  • HIV
  • AIDS
  • platelet functions
  • light transmission aggregometry
  • flow cytometry
  • microparticles
  • combined antiretroviral
  • antiplatelets

1. Introduction

There is increasing focus on platelet functions in people living with HIV/AIDS. This is because of the high incidence of cardiovascular events in these individuals that is 10 times higher than general population [1] independent of traditional risk factors such as age, hyperlipidemia, and ethnic/racial differences. Acquired platelet dysfunctions are often observed in association with HIV/AIDS. Of the available tests for platelet functions [2, 3], none fully captures the complexity involved in this population group.

The results of the functional assays are modified by the viral count, CD4/CD8 ratio, and immunological response and whether or not on antiretroviral treatment. The effects of combined antiretroviral therapy (cART) on platelet functions are complex. Despite achieving viral suppression, these drugs have been demonstrated to have independent effects on platelet functions.

Advertisement

2. Platelet count and indices in HIV/AIDS

Complete blood count and microscopic examination of formed elements are often the first investigations in suspected hemostatic disorders in clinical situations. Platelet count and morphological changes have impact on bleeding or thrombosis.

2.1 HIV-associated thrombocytopenia

Globally, the prevalence of HIV-associated thrombocytopenia is 4–40% [4] though there are geographical, racial as well as ethnic differences from the same locality [5] and stage of disease. Indeed, thrombocytopenia has been considered as a marker of disease progression and improvement [6]. Whereas platelet counts improve with initiation of combined antiretroviral therapy (cART) viral suppression [7], beneficial effect does not apply to zidovudin (AZT) [8].

Despite thrombocytopenia, very low rates of clinical hemorrhage have been reported, estimated at only 3.2% among HIV thrombocytopenic patients [9] even with platelet count as low as 50 × 109/L [10] casting doubt on the clinical relevance of the laboratory results. As a result of lack of clear correlation between HIV-associated thrombocytopenia and clinical significance, some authors have questioned benefit of treatments purely directed toward improvement of platelet count [11].

2.2 HIV-associated thrombocytosis

The prevalence of HIV-associated thrombocytosis, defined as platelet count of more than 400× 109/L [12], is low but depends on the population studied and concurrent medications. Reported prevalence of thrombocytosis in pediatric group who were also HIV-positive cART naive was found at 6% [13], though could be higher at 14% (more than thrombocytopenia at 7% in same cohort) for children on co-trimoxazole prophylaxis [14]. Whether these findings were independent or dependent on co-administered drugs remains undetermined.

Thrombocytosis is an emerging toxic complication accounting for 9% on stable cART depending on the regimen [7] up from 5.8% in treatment-naïve individuals [7]. It remains undetermined the relationship between HIV-associated thrombocytosis and accelerated thrombosis.

2.3 Platelet ultrastructure in HIV/AIDS

Despite the thrombocytopenia being associated with HIV, peripheral blood film smears of platelets are either unremarkable or hypogranular, which are of different sizes appearing as fragments [15].

Ultrastructure of platelets from HIV individuals, apart from showing normal features of hyperactivated aggregates having membrane pseudopodia/filopodia formation, in addition have shriveled aggregates with irregular and torn membrane surfaces, membrane blebbing and shedding of vesicles [16, 17]. The most distinctive features are alteration of granular structure though data are limited.

Advertisement

3. Tests based on platelet aggregation

3.1 Light transmission aggregometry (LTA)

Most studies on platelet aggregation in HIV have used single or fewer than the recommended panel of agonists with conflicting results [18]. Application of escalating agonist concentrations has uncovered dose-response patterns [19]. In this study, while epinephrine demonstrated greater potency indicating hyperresponsiveness, responses with collagen, TRAP, and ADP showed lesser maximum aggregation indicating lesser efficacy and hyporesponsiveness. The agonist dose-response curve is, however, modified by cART viral suppression, especially abacavir-containing regimens [20] depending on agonist [21]. It must be remembered that although cART is a commonly mentioned modifier, the effects of fever associated with HIV are neither reported nor analyzed in these studies. Hyperthermic conditions such as fever are associated with reduced platelet aggregation [22].

3.2 Whole blood platelet aggregometry-multiple electrode aggregometry (MEA) and impedance aggregometry

A study comparing whole blood platelet aggregation using MEA found hyporeactivity in both HIV-treated and untreated individuals [23], similar to findings by impedance aggregometry [24]. It is worth noting that co-infection with HBV (6 vs. 4%) and HCV (0 vs. 2%) and low CRP levels [23] could have obscured the overall response. Co-infection with other viruses modulates platelet responses in HIV [25].

3.3 Thromboelastography (TEG)/ Thromboelastometry (ROTEM)

Few studies have been performed using thromboelastography (TEG) in HIV individuals. Of the few studies done, MA amplitude was low despite higher normal fibrinogen levels in both cART-treated [21] and untreated HIV subjects [23]. These study results of hypocoagulability are not in keeping with other tests, probably reflecting lack of sensitivity of TEG as a platelet function assay.

Advertisement

4. Platelet activation

Activated platelets are characterized by surface expression of activation-specific molecules such as P-selectin or CD62P, active GPIIbIIIa (PAC-1), phosphatidyleserine (PS) externalization; platelet-leukocyte aggregates (PLA); platelet microparticle formation (PMP), in addition to granule secretion such as platelet factor 4(PF4), β-thromboglobulin, and intracellular calcium flux [26].

4.1 Flow cytometry for membrane surface glycoprotein expression

A number of studies have documented platelet hyperactivity in HIV characterized by increased plasma membrane surface expression of CD62P, PAC-1, PS, CD63, [27], but paradoxically decreased GPIbα [28]. The levels positively correlate with viral loads but not CD4 count [29].

Although activation markers are higher in HIV sero-positive individuals who are cART naïve compared to healthy controls [30], with cART treatment levels decrease but do not normalize to pre-treatment levels [20, 31]. The persistent levels are related to inflammatory markers in virally suppressed individuals [32].

4.2 Intracellular signal transduction test—VASP

There is evidence of altered signal transduction affecting protein synthesis, degranulation, and activation functioning in HIV platelets. Experimental data show that HIV platelets had upregulation of ABCC4 (ATP-binding cassette subfamily 4), increase in cAMP, decrease in vasodilator-stimulated phosphoprotein (VASP), which correlated with increased membrane expression of CD62P and integrin αIIbβ3 (GPIIbIIIa) [33]. It must be noted that VASP is only sensitive to PY12 inhibitors, and not much data are available from HIV patients.

Advertisement

5. Platelet secretion

5.1 Alpha granules

People living with HIV have increased secretion of alpha granule contents such as RANTES, sP-selectin, and sCD40L [34], despite viral suppression [33]. The persistence of these chemokines, especially anomalous secretion of RANTES, despite cART treatment [28] remains unexplained to date.

5.2 Dense granules

HIV platelets have low basal dense granule content and diminished secretion response as evidenced by low mepacrine uptake and release [33]. Although platelet mepacrine uptake and release have been considered among dense granule assays, it is not as specific as serotonin and lummiaggregometry for ATP [35, 36]. Despite this knowledge, the measurements of platelet serotonin and ATP remain largely undescribed in people living with HIV.

5.3 Concept of “platelet exhaustion” in HIV

Although HIV-associated platelets display increased baseline expression of surface activation markers compared to healthy controls [32], there is evidence of refractoriness to further agonist stimulation. This behavior has been referred to as “platelet exhaustion” in many publications [25, 28, 32, 37, 38].

Platelet “exhaustion” as a concept was postulated in references to previous observations, before HIV era, where activated platelets continued to circulate [39, 40] and were shown to be activated [41] but with decreased aggregation [42, 43]. They were considered refractory to further agonist stimulation [44] owing to acquired storage pool granule depletion [45, 46].

In HIV, stimulation with increased agonist concentration leads to lesser response at each corresponding dose [21]. Specifically, decreased thrombin dose-response curve for granule content and secretions for P-selectin, PFA/CXCL4,TXA and RANTES in HIV platelets less than healthy controls [32]. The decreased P-selectin and PAC-1 secretory responses correspond to impaired c-AMP, ABCC4 and VASP signal transduction mechanisms [33]. Furthermore, HIV platelets display decreased mepacrine uptake and release [33], and wheat germ agglutinin staining (WGA) [32] indicating reduction of dense and alpha granule contents respectively.

Despite many studies mentioning “platelet exhaustion” in HIV, however the results in support are neither consistent for all agonists nor confirmed by other tests. In patients who are cART naïve, stimulation with AA, ADP or collagen, the dose-response curves for CD62P are higher than the uninfected controls [30]. None of the LTA aggregation tests have been accompanied by corresponding Lummiaggregometry test which could have better characterized platelet ATP dense granule secretion [47, 48]. Platelet lumiaggregometry testing remains largely un-described in HIV. Furthermore, the studies are on people who are already infected by HIV, but platelet responses prior to HIV infection remains unknown.

From the foregoing, evidence in support for “platelet exhaustion” in HIV is suggestive but inconclusive. Although decreased dose-response to thrombin has been described, however response to epinephrine was enhanced in some studies. The maintained response to epinephrine casts doubt on granule exhaustion, since true storage pool disorder do not respond to epinephrine [49] or variable [50]. Indeed HIV platelets maintain both alpha and dense granule secretions to collagen and ADP agonists stimulation [51]. Perhaps a better term to use could be “anergy,” refractory or “tired” platelets.

Advertisement

6. Platelet adhesion

HIV platelets have enhanced adherence to fibrinogen-coated surfaces [32, 33]. However, testing by this method is technically difficult and not available in clinical situations.

Although platelet PFA-100/200 testing is always recorded as aggregation in most studies, in actual fact it is marker of adhesion [2, 52]. The few tests of PFA-100 in HIV compared those on cART treatment with untreated [31], or in addition to [53] all of which showed shorter closure time in treatment-naïve individuals. The short closure times were neither normalized with aspirin nor with cART. The results are strongly indicative of influence of vWF as a third dimension in platelet function testing [54, 55].

Advertisement

7. vWF-ADAMTS-13 axis in HIV/AIDS

People living with HIV (PLWHIV) despite having very low platelet counts do not have issues of bleeding [56, 57, 58]. Instead, HIV-associated thrombotic complications [59] are an emerging issue of concern [60]. Although congenital thrombotic thrombocytopenic purpura (TTP) is very rare, acquired TTP is on the increase and associated with HIV estimated to be 15–40 times than the HIV negative in the general population [61]. It has been reported that HIV is responsible for 80% of TTP cases [62].

TTP is characterized by reduced or absent ADAMTS-13 and elevated vWF antigen as well as activity [63] especially the Unusually Ultralarge vWF multimers [64]. Elevated vWF Ag and high-molecular-weight vWF multimers [65] with reduced ADAMTS-13 have been detected in acute and chronic HIV [66, 67] and those with confirmed thrombosis [68]. Unusually, ultralarge vWF multimers that have increased adhesion to platelet GPIbα-V-IX receptors [69] compensates for hemostasis in the presence of the low platelet count in HIV.

Advertisement

8. Platelet microparticles

It has been demonstrated that blood from HIV individuals have abundant circulating platelet microparticles [70], and this is despite viral suppression [71, 72]. The levels were associated with increased cellular ROS, caspases, eNOS [72], and mitochondrial membrane depolarization [73] indicative of apoptosis [74] . Further, co-existence of platelet microparticles with increased LPS and platelet P-selectin and TF [29] are strong indicators that they are products of platelet activation.

Advertisement

9. Mechanisms of platelet activation in HIV

9.1 Direct effect of HIV

Recently, in mice, HIV particles were shown to be endocytosed by platelets by binding to TLR-7&9 leading to increased secretion of alpha (PFA-4) and dense granules (serotonin), and membrane expression of P-selectin [75]. Additionally, HIV interacts directly with platelets CLEC-2 and DC-SIGN receptors [76] via its trans-activating factor (Tat) [77]. The consequence is increased intracellular calcium flux, translocation of P-selectin (CD62P) from the alpha granules to the membrane surface, secretion of chemokine CD154, and release of platelet microparticles [77]. The process is by enhancing platelet NOX-2 oxidative stress [78].

9.2 Gut microbiol translocation

HIV preferentially infects CD4-T lymphocytes present in the gut leading to reduction in number and function [79]. The consequence is loss of gut epithelial immune protection and disruption of gut epithelial barrier allowing luminal indigenous intestinal bacteria to translocate out of the mucosa and into circulation [80]. Once in circulation, bacterial products such as lipopolysaccharides (LPS) interact with platelet toll-like receptors 4 (TLR4) [81]. The microbial products induce signal transduction mechanisms that eventually lead to facilitating platelet membrane receptor expression [82, 83]. The phenomenon of gut microbial translocation has been used to explain enhanced platelet reactivity despite therapy with antiplatelets such as ticagrelor in myocardial infarction [84]. However, some studies have disputed the role of LPS in platelet activation instead of reporting attenuation of receptor expression and aggregation in the presence of agonists [85] contradicting earlier findings. The paradoxical result may be due to the absence or presence of other factors such as soluble CD14 that prime TLR4 sensing of LPS [86], extent of TLR expression [87] or the different LPS isoforms [88], and experimental conditions [89] as well as clinical condition [89].

9.3 Immune complexes, cytokines, and inflammatory markers

9.3.1 Cytokines

HIV infection is associated with elaboration of cytokines from inflammatory cells, and these have been shown to induce platelet activation [90, 91] The platelet activation is not limited to interleukins only, since tumor necrosis factor in blood leads to dose- and time-dependent increase in platelet expression of GPIIbIIIa, PS, and mitochondrial dysfunction [92]. The role of TNF-α in platelet activation and apoptosis are well supported by empirical evidence [93].

9.3.2 Immune complexes

Platelets express FcRIIA (CD32a) or simply FcR receptor that recognizes the constant region of IgG in immune complexes [94]. The consequence of platelet-immune complex binding leads to platelet activation [95], aggregation and release of contents from alpha and dense granules [94], and microparticle formation [96]. The platelet activation from immune complexes is dependent on membrane GP IIbIIIa [97]. However, the immune complex-induced platelet aggregation is dependent on dose and charge [98].

Cross-reactive antibodies between HIV epitopes and platelet receptors have been described [99, 100].

9.3.3 Neutrophil extracellular traps (NETS)

When neutrophils encounter viruses such as HIV, they respond by releasing reactive oxygen species and net-like structures called neutrophil extracellular traps [101, 102]. The NETs, composed of DNA, histones, myeloperoxidase, citrinulated histones, and elastases, are the potent inducers of platelet aggregation and activation [103, 104, 105].

9.3.4 Platelet-leukocyte complexes

There is often cross-talk between platelets and leukocytes associated with bidirectional priming and activation of each other [106, 107]. These two cells interact through platelets such as P-selecti-PSGL-1, GPIb-vWF-CD18, integrin IIaIIIb-fibrinogen-MAC-1 neutrophil linkages that lead to the formation of platelet-leukocyte aggregates (PLA) [108] linked by P-selectin-PSGL. These PLA conjugates have been found in HIV patients involving T-cells associated with CD42b and CD62P [109]. Elevated PLA together with other immune markers is positively correlated with increased platelet CD36, CD62P, and platelet aggregation but inversely with CD4 count [110].

9.3.5 vWF-GPIbα in platelet activation in HIV

There is evidence of endothelial damage [111] and increased vWF levels in HIV patients [66, 67, 68, 112, 113]. Apart from the high vWF Ag levels, of significant is the persistently high functionally active Ultralarge vWF multimers (ULvWFM) in HIV individuals [65] that causes adhesion even at low platelet counts [114]. Correspondingly, as HIV disease progresses, platelet expression of the integrin GPIbα decreases paradoxically unlike the other surface receptors indicating consumption [28].

9.4 Platelet apoptosis

There are similarities in markers of platelet activation and apoptosis [115]. In both processes, there is phosphatidyleserine (PS) exposure on the membrane [116] and microparticles [117]. However, specific features of platelet apoptosis include mitochondrial membrane leakage characterized by changes in membrane depolarization (Δψm) and increase in cytosolic caspases 3&8, [118, 119]. Indeed, features of platelet apoptosis and activation have been demonstrated in HIV patients [25, 32, 38]. It should be noted that the few studies demonstrating occurrence of full spectra of apoptosis in HIV individuals were confounded by cART viral suppression [32] and dengue co-infection [25] and therefore, whether results were specific to HIV in itself largely remains undetermined.

Some of the consequences of platelet apoptosis include thrombocytopenia [120, 121]. This is because, apart from the fact that apoptotic platelet eventually disintegrates [74], the surface exposure of PS acts as “eat me” signal for engulfment by the macrophages thus removing the altered cells from circulation shortening survival [122, 123, 124].

Advertisement

10. Antiretrovirals and platelet functions in HIV

Despite the success attained by cART in viral suppression and recovery of platelet counts [125, 126], their effects on platelet function remain variable. In general, platelet surface markers such as CD62P, PAC-1 and CD40L, soluble sCD62P, sCD40L as well as platelet-secreted chemokines such as RANTES persist despite cART viral suppression [27] with some variations between the individual drugs and study designs.

Platelet signal transduction and secretory effects are enhanced by HIV, but these effects are accentuated by cART. This was demonstrated by Pastori et al.’s [78] study in which levels of sCD40L, platelet sNOX-dp, and 8-iso-PGF2-α were elevated, the effects of PIs greater than NNRTI. The mechanism appears to be induction of oxidative stress, ROS, and arachidonic pathways that synergistically augment AA platelet activation. cART causes mitochondrial toxicities [127] via ROS release and inner membrane depolarization that eventually lead to apoptosis that persists even with viral suppression [32].

Abacavir is unique among cART [51] since it is a guanosine analogue and induces platelet activation via its effects on NO-cGMP signal transduction pathway [20]. In vitro, incubation of platelets with abacavir inhibits cAMP pathway and dose-dependently increases surface expression of P-selectin, an observation that is augmented by ADP agonist [128]. Compared with TDF and TAF, abacavir enhances platelet aggregation and increases agonist-induced platelet activation in vivo (CD62P, PAC-1) [31]. It has been shown that it induces both alpha and dense platelet granule secretions thereby increasing membrane CD62P [128] levels as well as increasing PAC-1 [129]. It also alters metabolic enzymes that lead to increase in PAF from leukocytes [130]. This likely explains its association with cardiovascular events where enhanced platelet hyperactivity plays a central role [131, 132].

Despite other studies reporting levels of platelets MP remaining unchanged [29] or increased [71] after initiating antiretrovirals, one study found MP TF levels decreased with cART treatment [133]. The difference could be attributed to monocyte phenotypes [134] and level of activation and attendant TF expression with cART [135]. This is because platelets undergo decryption [136] and transfer TF to monocytes using microparticles as vehicles [137, 138].

The effects of cART on platelets are complicated by other factors such as TNF-α, a known platelet activator and apoptosis inducer. Although TNF levels are often elevated in HIV infection, levels persist despite cART [139] even if used over 24-month period [34]. Whereas cART treatment decreases circulating bacterial LPS levels in HIV patients, platelet reactivity is increased instead [23] suggesting intrinsic effects of the drugs independent of bacterial translocation.

11. Antiplatelets in HIV/AIDS

People living with HIV/AIDS are at increased risk of cardiovascular events [140, 141], especially coronary heart disease [142, 143] and ischemic stroke [144, 145], than the general population. The increased risk is due to HIV infection alone and accentuated by cART [146, 147].

Although there is evidence of enhanced platelet activation in association with HIV [27], studies of antiplatelet therapy in these patients have yielded inconsistent results, perhaps owing to drug interactions [148]. It should be noted that the studies so far done were on patients concurrently taking cART.

In a study of HIV-1 infected patients who had been on 6-month cART, it was found that 325 mg of oral aspirin-attenuated platelet aggregation to agonists, activation markers [37]. In the same study, although levels of urinary thromboxane were decreased in both HIV-positive cART untreated and treated, it was least responsive to aspirin. Furthermore, despite aspirin administration, suppression of platelet hyperactivity did not decline to baseline levels indicating the contributory effects of cART. Apart from the small sample size and short duration of therapy, other limitations of this pilot study are that it evaluated only one antiplatelet drug, and it did not perform subgroup analysis among the different cART drugs (NNRTI, PI, Raltegravir, and abacavir) as well as the racial and ethnic differences.

Although aspirin and R406 (thromboxane analogue) but not ticagrelor inhibits platelet engulfment, they do not inhibit CD62P expression or PMA complex formation [149]. Other studies have confirmed the suboptimal effects of aspirin on platelets agonist (collagen and epinephrine)-induced aggregation, surface expression of CD62P, CD40L, and PAC-1 from individuals with HIV taking ABC [53]. This study identified subjects taking abacavir-containing cART as poor responders. While cART is currently standard of care in the treatment of HIV, there are no data on effects of antiplatelets in PLWH before adoption of practice.

Clopidogrel reduces thrombogenicity and platelet hyperreactivity better than aspirin in PLWH on cART [21]. The question whether dual antiplatelet therapy compared to single agent may have a better reduction in platelet hyperreactivity in HIV concurrently taking cART was evaluated in the EVERE2ST-HIV [18]. This study evaluated the extent of platelet inhibition patients with acute coronary patients on dual antiplatelet therapy undergoing PCI utilizing various platelet function assays [18]. The findings were that P2Y12 inhibitors (clopidogrel, prasugrel, and ticagralor) and aspirin were all associated with residual platelet reactivity on light transmission aggregometry (LTA), VerifyNow, and VASP assays. Furthermore, HIV infection was an independent risk factor for the high on antiplatelet reactivity that was increased by combined antiretroviral therapy (cART). Of the cART, protease inhibitors had greater effects than the NNRTIs. The residual platelet reactivity in PLWHIV despite viral suppression and dual antiplatelet therapy can probably be accounted by the active immune mechanisms and drug interactions [148].

Overall, few studies have evaluated the effects of antiplatelets in persons living with HIV. The available studies suffer from small sample sizes and have not been performed in populations not taking cART. Furthermore, the different classes of antiplatelets have not been evaluated. Of the studies done so far, the results do demonstrate neither efficacy nor improved outcomes with either aspirin or clopidogrel.

12. Conclusion

Infection with HIV is associated with reduced platelet count; extent of thrombocytopenia inversely correlates with viral load and disease progression. Despite thrombocytopenia, cardiovascular events are on the increase. There is associated platelet hyperactivity, as evidenced by increased surface expression of CD62P, CD40L, platelet microparticles, and platelet leukocyte aggregates. There is enhanced secretion of chemokines such as RANTES. Combined antiretroviral drugs independently and synergistically with HIV enhance platelet hyperactivity that persists despite viral suppression. Data on the effects of antiplatelets in this population can at best be described as clinical equipoise.

Other declarations

Autor’s ORCID identifier: 0000-0001-6466-172X.

References

  1. 1. Ahonkhai AA, Gebo KA, Steiff MB, Moore RD, Segal JB. Venous thromboembolism in patients with HIV/AIDS. A case-control study. Journal of Acquired Immune Deficiency Syndromes. 2008;48(3):310-314. DOI: 10.1097/QAI.0b013e318163bd70
  2. 2. Lordkipanidzé M. Platelet function tests. Seminars in Thrombosis and Hemostasis. 2016;42(3):258-267. DOI: 10.1055/s-0035-1564834
  3. 3. Mansouritorghabeh H, De Laat B, Roest M. Current methods of measuring platelet activity : Pros and cons. Blood Coagulation & Fibrinolysis. 2020;31:426-433. DOI: 10.1097/MBC.0000000000000941
  4. 4. Getawa S, Aynalem M, Bayleyegn B, Adane T. The global prevalence of thrombocytopenia among HIV-infected adults: A systematic review and meta-analysis. International Journal of Infectious Diseases. 2021;105:495-504. DOI: 10.1016/j.ijid.2021.02.118
  5. 5. Sloand EM, Klein HG, Banks SM, Vareldzis B, Merritt S, Pierce P. Epidemiology of thrombocytopenia in HIV infection. European Journal of Haematology. 1992;48(3):168-172. DOI: 10.1111/j.1600-0609.1992.tb00591.x
  6. 6. Passos AM, Treitinger A, Spada C. An overview of the mechanisms of HIV-related thrombocytopenia. Acta Haematologica. 2010;124(1):13-18. DOI: 10.1159/000313782
  7. 7. Li B et al. Manifestations and related risk factors of thrombocyte abnormalities in HIV-positive patients before and after the initiation of art. Infection and Drug Resistance. 2021;14:4809-4819. DOI: 10.2147/IDR.S334046
  8. 8. Marchionatti A, Parisi MM. Anemia and thrombocytopenia in people living with HIV/AIDS: A narrative literature review. International Health. 2021;13(2):98-109. DOI: 10.1093/inthealth/ihaa036
  9. 9. Ongondi M, Amayo EO, Lule GN, Rajab JA. Thrombocytopenia in HAART NAIVE HIV infected patients attending the Comprehensive Care Clinic at Kenyatta National Hospital. East African Medical Journal. 2016;93(9):406-408
  10. 10. Dominguez A, Gamallo G, Garcia R, Lopez-Pastor A, Peña JM, Vazquez JJ. Pathophysiology of HIV related thrombocytopenia: An analysis of 41 patients. Journal of Clinical Pathology. 1994;47(11):999-1003. DOI: 10.1136/jcp.47.11.999
  11. 11. Miguez-Burbano MJ, Jackson J, Hadrigan S. Thrombocytopenia in HIV disease: Clinical relevance, physiopathology and management. Current Medicinal Chemistry. Cardiovascular and Hematological Agents. 2005;3(4):365-376. DOI: 10.2174/156801605774322364
  12. 12. Mathur A, Samaranayake S, Storrar NPF, Vickers MA. Investigating thrombocytosis. BMJ. 2019;366(July):1-6. DOI: 10.1136/bmj.l4183
  13. 13. Ellaurie M. Thrombocytosis in pediatric HIV infection. Clinical Pediatrics (Phila). 2004;43(7):627-629. DOI: 10.1177/000992280404300707
  14. 14. Mateveke-Kuona P, Bwakura MF, Dzangare J, Pazvakavambwa I. Haematological features in children less than 12 years on co-trimoxazole prophylaxis seen in opportunistic infection clinics at Harare and Parirenyatwa Teaching Hospitals. The Central African Journal of Medicine. 2010;56(9/12):51-56
  15. 15. Bamberg R, Johnson J. Segmented neutrophil size and platelet morphology in HIV/AIDS patients. Clinical Laboratory Science. 2002;15(1):18-22
  16. 16. Pretorius E, Smit E, Oberholzer HM, Steyn E, Briedenhann S, Franz RC. Investigating the ultrastructure of platelets of HIV patients treated with the immuno-regulator, Canova. Histology and Histopathology. 2009;24:399-405
  17. 17. Jackson BS, Nunes Goncalves J, Pretorius E. Comparison of pathological clotting using haematological, functional and morphological investigations in HIV-positive and HIV-negative patients with deep vein thrombosis. Retrovirology. 2020;17(1):1-13. DOI: 10.1186/s12977-020-00523-3
  18. 18. Hauguel-Moreau M et al. Platelet reactivity in human immunodeficiency virus infected patients on dual antiplatelet therapy for an acute coronary syndrome: The EVERE2ST-HIV study. European Heart Journal. 2017;38(21):1676-1686. DOI: 10.1093/eurheartj/ehw583
  19. 19. Satchell CS et al. Platelet function and HIV : A case – Control study. AIDS. 2010;24:649-657. DOI: 10.1097/QAD.0b013e328336098c
  20. 20. Satchell CS et al. Increased platelet reactivity in HIV-1 – Infected patients receiving abacavir-containing antiretroviral therapy. JID. 2011;204:1202-1210. DOI: 10.1093/infdis/jir509
  21. 21. Brien MPO et al. Targeting thrombogenicity and inflammation in chronic HIV infection. Science Advances. 2019;5:eaav5463
  22. 22. Etulain J et al. Hyperthermia inhibits platelet hemostatic functions and selectively regulates the release of alpha-granule proteins. Journal of Thrombosis and Haemostasis. 2011;9(8):1562-1571. DOI: 10.1111/j.1538-7836.2011.04394.x
  23. 23. Haugaard AK, Lund TT, Birch C, Trøseid M, Ullum H, Gerstoft J. Discrepant coagulation profile in HIV infection : Elevated D-dimer but impaired platelet aggregation and clot initiation. AIDS. 2013;27:2749-2758. DOI: 10.1097/01.aids.0000432462.21723.ed
  24. 24. Muñoz RP et al. Whole blood platelet aggregometry in HIV-infected patients on treatment with abacavir *. OJIM. 2012;2012:62-66. DOI: 10.4236/ojim.2012.22013
  25. 25. Hottz ED, Quirino-teixeira AC, Valls-de-souza R, Zimmerman GA, Bozza FA, Bozza PT. Platelet function in HIV plus dengue coinfection associates with reduced inflammation and milder dengue illness. Scientific Reports. 2019;9(1):1-13. DOI: 10.1038/s41598-019-43275-7
  26. 26. Kannan M, Ahmad F, Saxena R. Platelet activation markers in evaluation of thrombotic risk factors in various clinical settings. Blood Reviews. 2019;37:100583. DOI: 10.1016/j.blre.2019.05.007
  27. 27. Nkambule BB et al. Platelet activation in adult HIV-infected patients on antiretroviral therapy: A systematic review and meta-analysis. BMC Medicine. 2020;18:357. DOI: 10.1186/s12916-020-01801-9
  28. 28. Holme PA, Muller F, Solum NO, Brosstad F, Land Q , Aukrust PAL. Enhanced activation of platelets with abnormal release of RANTES in human immunodeficiency virus type 1 infection. The FASEB Journal. 1998;12:79-89
  29. 29. Mayne E et al. Increased platelet and microparticle activation in HIV infection : Upregulation of P-selectin and tissue factor expression. Journal of Acquired Immune Deficiency Syndromes. 2012;59(4):340-346
  30. 30. Nkambule BB, Davison GM, Ipp H. The evaluation of platelet function in HIV infected, asymptomatic treatment-naïve individuals using flow cytometry. Thrombosis Research. 2015;135(6):1131-1139. DOI: 10.1016/j.thromres.2015.01.031
  31. 31. Francisci D, Falcinelli E, Belfiori B, Petito E, Guglielmini G, Malincarne L. In vivo platelet activation and platelet hyperreactivity in abacavir- treated HIV-infected patients. Thrombosis and Haemostasis. 2013;110(8):349-357. DOI: 10.1160/TH12-07-0504
  32. 32. Mesquita EC et al. Persistent platelet activation and apoptosis in virologically suppressed HIV-infected individuals. Scientific Reports. 2018;8(1):1-10. DOI: 10.1038/s41598-018-33403-0
  33. 33. Marcantoni E et al. Platelet transcriptome profiling in as a mediator of platelet activity. JACC: Basic to Translational Science. 2018;3:9-22. DOI: 10.1016/j.jacbts.2017.10.005
  34. 34. Landrø L, Ueland T, Otterdal K, Frøland SS, Aukrust P. Persistently raised plasma levels of platelet-derived inflammatory mediators in HIV-infected patients during highly active anti-retroviral therapy. Journal of Thrombosis and Haemostasis. 2011;9(5):1075-1077. DOI: 10.1111/j.1538-7836.2011.04242.x
  35. 35. Mumford AD et al. A review of platelet secretion assays for the diagnosis of inherited platelet secretion disorders. Thrombosis and Haemostasis. 2015;114(1):14-25. DOI: 10.1160/TH14-11-0999
  36. 36. Pai M et al. Diagnostic usefulness of a lumi-aggregometer adenosine triphosphate release assay for the assessment of platelet function disorders. American Journal of Clinical Pathology. 2011;136(3):350-358. DOI: 10.1309/AJCP9IPR1TFLUAGM
  37. 37. O’Brien M et al. Aspirin attenuates platelet activation and immune activation in HIV-1-infected subjects on antiretroviral therapy: A pilot study. Journal of Acquired Immune Deficiency Syndromes. 2013;63(3):280-288. DOI: 10.1097/QAI.0b013e31828a292c
  38. 38. Gama WM et al. Increased levels of reactive oxygen species in platelets and platelet-derived microparticles and the risk of respiratory failure in HIV/AIDS patients. Memórias do Instituto Oswaldo Cruz. 2020;115:e200082. DOI: 10.1590/0074-02760200082
  39. 39. O’Brien JR. "Exhausted " platelets continue to circulate. The Lancet. 1978;312(8103):1316-1317. DOI: 10.1016/s0140-6736(78)92087-1
  40. 40. Pareti FI, Capitanio A, Mannucci L, Ponticelli C, Mannucci PM. Acquired dysfunction due to the circulation of ‘exhausted’ platelets. The American Journal of Medicine. 1980;69(2):235-240. DOI: 10.1016/0002-9343(80)90383-6
  41. 41. Boneu B, Bugat R, Boneu A, Eche N, Sie P, Combes P-F. Exhausted platelets in patients with malignant solid tumors without evidence of active consumption coagulation. European Journal of Cancer & Clinical Oncology. 1984;20(7):890-903
  42. 42. Evans RJ, Gordon JL. Refractoriness in blood platelets: Effect of prior exposure to aggregating agents on subsequent aggregation responses. British Journal of Pharmacology. 1974;51(1):123
  43. 43. Fong BJSC, Kaplan BS. Ipairment of platelet aggregation in Hemolytic uremic syndrome: Evidence for platelet ‘exhaustion’. Blood. 1982;60(3):564-571
  44. 44. O’Brien JR, Etherrington M, Jameson S. Refractory state of platelet aggregation with major operations. The Lancet. 1971;2(7727):741-743. DOI: 10.1016/s0140-6736(71)92107-6
  45. 45. Pareti F, Capitanio A, Mannucci P. Acquired storage pool disease in platelets during disseminated intravascular coagulation. Blood. 1976;48(4):511-515. DOI: 10.1182/blood.v48.4.511.511
  46. 46. Zahavi J, Marder VJ. Acquired ‘storage pool disease’ of platelets associated with circulating antiplatelet antibodies. The American Journal of Medicine. 1974;56(6):883-890. DOI: 10.1016/0002-9343(74)90819-5
  47. 47. Hughes CE. How to perform aggregometry and lumi-aggregometry in mouse platelets. Platelets. 2018;29(7):638-643. DOI: 10.1080/09537104.2018.1478074
  48. 48. Jurk K, Shiravand Y. Platelet phenotyping and function testing in thrombocytopenia. Journal of Clinical Medicine. 2021;10(5):1114. DOI: 10.3390/jcm10051114
  49. 49. Fritsma GA. Platelet function testing: Aggregometry and lumiaggregometry. Clinical Laboratory Science. 2007;20(1):32-37. DOI: 10.29074/ascls.20.1.32
  50. 50. Weiss HJ, Lages B. The response of platelets to epinephrine in storage pool deficiency - evidence pertaining to the role of adenosine diphosphate in mediating primary and secondary aggregation. Blood. 1988;72(5):1717-1725. DOI: 10.1182/blood.v72.5.1717.1717
  51. 51. Taylor KA et al. Pharmacological impact of antiretroviral therapy on platelet function to investigate human immunodeficiency virus - associated cardiovascular risk. British Journal of Pharmacology. 2019;176:879-889. DOI: 10.1111/bph.14589
  52. 52. Paniccia R, Priora R, Liotta AA, Abbate R. Platelet function tests: A comparative review. Vascular Health and Risk Management. 2015;11:133-148. DOI: 10.2147/VHRM.S44469
  53. 53. Falcinelli E et al. Effect of aspirin treatment on abacavir-associated platelet hyperreactivity in HIV-infected patients. International Journal of Cardiology. 2018;263:118-124. DOI: 10.1016/j.ijcard.2018.04.052
  54. 54. Castaman G et al. The impact of bleeding history, von Willebrand factor and PFA – 100 â on the diagnosis of type 1 von Willebrand disease : Results from the European study MCMDM-1VWD. British Journal of Haematology. 2010;151:245-251. DOI: 10.1111/j.1365-2141.2010.08333.x
  55. 55. Gianetti J, Parri MS, Della Pina F, Marchi F, Koni E, De Caterina A, et al. Von willebrand factor antigen predicts response to double dose of aspirin and clopidogrel by PFA-100 in patients undergoing primary angioplasty for ST elevation myocardial infarction. The Scientific World Journal. 2013:313492. DOI: 10.1155/2013/313492
  56. 56. Franzetti M et al. Changes in the incidence of severe thrombocytopenia and its predisposing conditions in HIV-infected patients since the introduction of highly active antiretroviral therapy. Journal of Acquired Immune Deficiency Syndromes. 2014;67(5):493-498. DOI: 10.1097/QAI.0000000000000347
  57. 57. Nascimento FG, Tanaka PY. Thrombocytopenia in HIV-infected patients. Indian Journal of Hematology and Blood Transfusion. 2012;28(2):109-111. DOI: 10.1007/s12288-011-0124-9
  58. 58. Vannappagari V, Nkhoma ET, Atashili J, Laurent SS, Zhao H. Prevalence, severity, and duration of thrombocytopenia among HIV patients in the era of highly active antiretroviral therapy. Platelets. 2011;22(8):611-618. DOI: 10.3109/09537104.2011.582526
  59. 59. Ahmed S, Siddiqui RK, Siddiqui AK, Zaidi SA, Cervia J. HIV associated thrombotic microangiopathy. Postgraduate Medical Journal. 2002;78:520-525
  60. 60. Dentali F, Nicolini E, Ageno W. Venous and arterial thrombosis associated with HIV infection. Seminars in Thrombosis and Hemostasis. 2012;38(5):524-529. DOI: 10.1055/s-0032-1306434
  61. 61. Meiring M, Webb M, Goedhals D, Louw V. HIV-associated thrombotic thrombocytopenic purpura - what we know so far. European Oncology & Haematology. 2012;8(2):89-91. DOI: 10.17925/eoh.2012.08.02.89
  62. 62. Gunther K, Garizio D, Dhlamini B. The pathogenesis of HIV-related thrombotic thrombocytopaenic purpura – Is it different? ISBT Science Series. 2006;1(1):246-250. DOI: 10.1111/j.1751-2824.2006.00041.x
  63. 63. De La, Rubia J, Contreras E, Del Río-Garma J. Thrombotic thrombocytopenic purpura. Medicina Clínica (Barcelona). 2011;136(12):534-540. DOI: 10.1016/j.medcli.2010.02.011
  64. 64. Tsai H-M. Deficiency of ADAMTS13 causes thrombotic thrombocytopenic Purpura. Arteriosclerosis, Thrombosis, and Vascular Biology. 2003;23:388-397. DOI: 10.1161/01.ATV.0000058401.34021.D4
  65. 65. Aukrust P et al. Persistently elevated levels of von Willebrand factor antigen in HIV infection. Downregulation during highly active antiretroviral therapy. Thrombosis and Haemostasis. 2000;84:183-187
  66. 66. Graham SM et al. Von willebrand factor adhesive activity and ADAMTS13 protease activity in HIV-1-infected men. International Journal of Medical Sciences. 2019;16(2):276-284. DOI: 10.7150/ijms.28110
  67. 67. Graham SM et al. Elevated plasma von Willebrand factor levels are associated with subsequent ischemic stroke in persons with treated HIV infection. Open Forum Infectious Diseases. 2021;1:1-9. DOI: 10.1093/ofid/ofab521
  68. 68. van den, Dries LWJ et al. von Willebrand factor is elevated in HIV patients with a history of thrombosis. Frontiers in Microbiology. 2015;6:1-8. DOI: 10.3389/fmicb.2015.00180
  69. 69. Varga-Szabo D, Pleines I, Nieswandt B. Cell adhesion mechanisms in platelets. Arteriosclerosis, Thrombosis, and Vascular Biology. 2008;28(3):403-413. DOI: 10.1161/ATVBAHA.107.150474
  70. 70. Kelly C et al. Circulating microparticles are increased amongst people presenting with HIV and advanced immune suppression in Malawi and correlate closely with arterial stiffness: A nested case control study. Wellcome Open Research. 2021;6:264. DOI: 10.12688/wellcomeopenres.17044.1
  71. 71. Corrales-Medina VF et al. Increased levels of platelet microparticles in HIV-infected patients with good response to highly active antiretroviral therapy. Journal of Acquired Immune Deficiency Syndromes. 2010;54(2):217-218
  72. 72. Hijmans JG et al. Circulating microparticles are elevated in treated HIV-1 infection and are deleterious to endothelial cell function. Journal of the American Heart Association. 2019;8:e011134. DOI: 10.1161/JAHA.118.011134
  73. 73. van der, Heijden WA et al. Long-term treated HIV infection is associated with platelet mitochondrial dysfunction. Scientific Reports. 2021;11(1):1-12. DOI: 10.1038/s41598-021-85775-5
  74. 74. Gyulkhandanyan AV, Mutlu A, Freedman J, Leytin V. Markers of platelet apoptosis: Methodology and applications. Journal of Thrombosis and Thrombolysis. 2012;33(4):397-411. DOI: 10.1007/s11239-012-0688-8
  75. 75. Banerjee M et al. Platelets endocytose viral particles and are activated via TLR (toll-like receptor) signalling. Arteriosclerosis, Thrombosis, and Vascular Biology. 2020;40:1635-1650. DOI: 10.1161/ATVBAHA.120.314180
  76. 76. Chaipan C et al. DC-SIGN and CLEC-2 mediate human immunodeficiency virus type 1 capture by platelets. Journal of Virology. 2006;80(18):8951-8960. DOI: 10.1128/jvi.00136-06
  77. 77. Wang J, Zhang W, Nardi MA, Li Z. HIV-1 Tat-induced platelet activation and release of CD154 contribute to HIV-1-associated autoimmune thrombocytopenia. Journal of Thrombosis and Haemostasis. 2011;9(3):562-573. DOI: 10.1111/j.1538-7836.2010.04168.x
  78. 78. Pastori D et al. HIV-1 induces in vivo platelet activation by enhancing platelet NOX2 activity. The Journal of Infection. 2015;70(6):651-658. DOI: 10.1016/j.jinf.2015.01.005
  79. 79. Hsue PY. Mechanisms of cardiovascular disease in the setting of HIV infection. The Canadian Journal of Cardiology. 2019;35(3):238-248. DOI: 10.1016/j.cjca.2018.12.024
  80. 80. Shan L, Siliciano RF. Unraveling the relationship between microbial translocation and systemic immune activation in HIV infection. The Journal of Clinical Investigation. 2014;124(6):2368-2371. DOI: 10.1172/JCI75799
  81. 81. Lopes Pires ME, Clarke SR, Marcondes S, Gibbins JM. Lipopolysaccharide potentiates platelet responses via toll-like receptor 4-stimulated Akt-Erk-PLA2 signalling. PLoS One. 2017;12(11):1-22. DOI: 10.1371/journal.pone.0186981
  82. 82. Nocella C et al. Lipopolysaccharide induces platelet activation in HIV patients: The role of different viral load patterns. HIV Medicine. 2021;22(6):434-444. DOI: 10.1111/hiv.13059
  83. 83. Zhang G et al. Lipopolysaccharide stimulates platelet secretion and potentiates platelet aggregation via TLR4/MyD88 and the cGMP-dependent protein kinase pathway. Journal of Immunology. 2009;182(12):7997-8004. DOI: 10.4049/jimmunol.0802884
  84. 84. Zhang X et al. Gut microbiota induces high platelet response in patients with ST segment elevation myocardial infarction after ticagrelor treatment. eLife. 2022;11:e70240. DOI: 10.7554/eLife.70240
  85. 85. Martyanov AA et al. Effects of bacterial lipopolysaccharides on platelet function: Inhibition of weak platelet activation. Scientific Reports. 2020;10(1):1-10. DOI: 10.1038/s41598-020-69173-x
  86. 86. Damien P et al. LPS stimulation of purified human platelets is partly dependent on plasma soluble CD14 to secrete their main secreted product, soluble-CD40-ligand. BMC Immunology. 2015;16(1):1-7. DOI: 10.1186/s12865-015-0067-2
  87. 87. Aslam R et al. Platelet toll-like receptor expression modulates lipopolysaccharide-induced thrombocytopenia and tumor necrosis factor-α production in vivo. Blood. 2006;107(2):637-641. DOI: 10.1182/blood-2005-06-2202
  88. 88. Berthet J et al. Human platelets can discriminate between various bacterial LPS isoforms via TLR4 signaling and differential cytokine secretion. Clinical Immunology. 2012;145(3):189-200. DOI: 10.1016/j.clim.2012.09.004
  89. 89. Jonathan S, Rajendran V, Dash P, Ketan P, Darius W. Effect of ultrapure lipopolysaccharides derived from diverse bacterial species on the modulation of platelet activation. Scientific Reports. 2019;9:18258. DOI: 10.1038/s41598-019-54617-w
  90. 90. Lumadue JA, Lanzkron SM, Kennedy SD, Kuhl DT, Mt BS, Kickler TS. Cytokine induction of platelet activation. American Journal of Clinical Pathology. 1996;106:795-798
  91. 91. Burstein SA et al. Cytokine-induced alteration of platelet and hemostatic function. Stem Cells. 1996;14(Suppl 1):154-162
  92. 92. Davizon-Castillo P et al. TNF-a–driven inflammation and mitochondrial dysfunction define the platelet hyperreactivity of aging. Blood. 2019;134(9):727-740. DOI: 10.1182/blood.2019000200
  93. 93. Çevİk Ö, Adigüzel Z, Baykal AT, Şener A. Tumor necrosis factor-alpha induced caspase-3 activation-related iNOS gene expression in ADP-activated platelets. Turkish Journal of Biology. 2017;41:31-40. DOI: 10.3906/biy-1509-64
  94. 94. Arman M, Krauel K. Human platelet IgG Fc receptor Fc c RIIA in immunity and thrombosis. Journal of Thrombosis and Haemostasis. 2015;13:893-908. DOI: 10.1111/jth.12905
  95. 95. Goette NP, Glembotsky AC, Lev PR, Grodzielski M. Platelet apoptosis in adult immune thrombocytopenia : Insights into the mechanism of damage triggered by auto- antibodies. PLoS One. 2016;11(8):e0160563. DOI: 10.1371/journal.pone.0160563
  96. 96. Larson A, Egberg N, Lindahl L. Platelet activation and binding of complement components to platelets induced by immune complexes. Platelets. 1994;5:149-155
  97. 97. Zhi H, Dai J, Liu J, Zhu J, Newman DK, Gao C. Platelet activation and thrombus formation over IgG immune complexes requires integrin α IIb β 3 and Lyn kinase. PLoS One. 2015;10(8):e0135738. DOI: 10.1371/journal.pone.0135738
  98. 98. Schattner BM et al. Activation of human platelets by immune complexes prepared with cationized human IgG. Blood. 1993;82(10):3045-3051
  99. 99. Li Z, Nardi MA, Karpatkin S. Role of molecular mimicry to HIV-1 peptides in HIV-1-related immunologic thrombocytopenia. Blood. 2005;106(2):572-576. DOI: 10.1182/blood-2005-01-0243
  100. 100. Zhang W, Nardi MA, Borkowsky W, Li Z, Karpatkin S. Role of molecular mimicry of hepatitis C virus protein with platelet GPIIIa in hepatitis C-related immunologic thrombocytopenia. Blood. 2009;113(17):4086-4093. DOI: 10.1182/blood-2008-09-181073
  101. 101. Saitoh T et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host & Microbe. 2012;12(1):109-116. DOI: 10.1016/j.chom.2012.05.015
  102. 102. Barr FD, Ochsenbauer C, Wira CR, Rodriguez-Garcia M. Neutrophil extracellular traps prevent HIV infection in the female genital tract. Mucosal Immunology. 2018;11(5):1420-1428. DOI: 10.1038/s41385-018-0045-0
  103. 103. Fuchs TA et al. Extracellular DNA traps promote thrombosis. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(36):15880-15885. DOI: 10.1073/pnas.1005743107
  104. 104. Elaskalani O, Abdol Razak NB, Metharom P. Neutrophil extracellular traps induce aggregation of washed human platelets independently of extracellular DNA and histones. Cell Communication and Signalling. 2018;16(1):1-15. DOI: 10.1186/s12964-018-0235-0
  105. 105. Zhou P et al. Interactions between neutrophil extracellular traps and activated platelets enhance procoagulant activity in acute stroke patients with ICA occlusion. eBioMedicine. 2020;53:102671. DOI: 10.1016/j.ebiom.2020.102671
  106. 106. Zarbock A, Polanowska-Grabowska RK, Ley K. Platelet-neutrophil-interactions: Linking hemostasis and inflammation. Blood Reviews. 2007;21(2):99-111. DOI: 10.1016/j.blre.2006.06.001
  107. 107. Stark K. Platelet-neutrophil crosstalk and netosis. HemaSphere. 2019;3(S2):89-91. DOI: 10.1097/HS9.0000000000000231
  108. 108. Carestia A, Kaufman T, Schattner M. Platelets: New bricks in the building of neutrophil extracellular traps. Frontiers in Immunology. 2016;7:271. DOI: 10.3389/fimmu.2016.00271
  109. 109. Green SA et al. Activated platelet – T-cell conjugates in peripheral blood of patients with HIV infection : Coupling coagulation / inflammation and T cells. AIDS. 2015;29:1297-1308. DOI: 10.1097/QAD.0000000000000701
  110. 110. Nkambule BB, Davison G, Ipp H. Platelet leukocyte aggregates and markers of platelet aggregation, immune activation and disease progression in HIV infected treatment naive asymptomatic individuals. Journal of Thrombosis and Thrombolysis. 2015;40(4):458-467. DOI: 10.1007/s11239-015-1212-8
  111. 111. Francisci D et al. HIV type 1 infection, and not short-term HAART , induces endothelial dysfunction. AIDS. 2009;23:589-596. DOI: 10.1097/QAD.0b013e328325a87c
  112. 112. Jong E, Louw S, Van Gorp ECM, Meijers JCM, Ten Cate H, Jacobson BF. The effect of initiating combined antiretroviral therapy on endothelial cell activation and coagulation markers in South African HIV-infected individuals. Thrombosis and Haemostasis. 2010;104(6):1228-1234. DOI: 10.1160/TH10-04-0233
  113. 113. Van Den Dries LWJ, Gruters RA, Van Der Borden SBCH. von Willebrand factor is elevated in HIV patients with a history of thrombosis. Frontiers in Microbiology. 2015;6:180. DOI: 10.3389/fmicb.2015.00180
  114. 114. Reininger AJ. The function of ultra-large von Willebrand factor multimers in high shear flow controlled by ADAMTS13. Hämostaseologie. 2015;35:225-233
  115. 115. Mutlu A, Gyulkhandanyan AV, Freedman J, Leytin V. Concurrent and separate inside-out transition of platelet apoptosis and activation markers to the platelet surface. British Journal of Haematology. 2013;163(3):377-384. DOI: 10.1111/bjh.12529
  116. 116. Reddy EC, Rand ML. Procoagulant phosphatidylserine-exposing platelets in vitro and in vivo. Frontiers in Cardiovascular Medicine. 2020;7:1-11. DOI: 10.3389/fcvm.2020.00015
  117. 117. Böing AN, Hau CM, Sturk A, Nieuwland R. Platelet microparticles contain active caspase 3. Platelets. 2008;19(2):96-103. DOI: 10.1080/09537100701777295
  118. 118. Gyulkhandanyan AV et al. Mitochondrial inner membrane depolarization as a marker of platelet apoptosis : Disclosure of nonapoptotic membrane depolarization. Clinical and Applied Thrombosis/Hemostasis. 2017;23(2):139-147. DOI: 10.1177/1076029616665924
  119. 119. Leytin V, Allen DJ, Mutlu A, Gyulkhandanyan AV, Mykhaylov S, Freedman J. Mitochondrial control of platelet apoptosis: Effect of cyclosporin a, an inhibitor of the mitochondrial permeability transition pore. Laboratory Investigation. 2009;89(4):374-384. DOI: 10.1038/labinvest.2009.13
  120. 120. Thushara RM, Hemshekhar M, Kemparaju K, Rangappa KS, Girish KS. Biologicals, platelet apoptosis and human diseases : An outlook. Critical Reviews in Oncology/Hematology. 2015;93(3):149-158. DOI: 10.1016/j.critrevonc.2014.11.002
  121. 121. Von Gunten S, Wehrli M, Simon H. Cell death in immune thrombocytopenia_ novel insights and perspectives. Seminars in Hematology. 2013;50(1):S109-S115. DOI: 10.1053/j.seminhematol.2013.03.016
  122. 122. Rand ML, Wang H, Bang KWA, Poon KSV, Packhams MA, Freedman J. Procoagulant surface exposure and apoptosis in rabbit platelets : Association with shortened survival and steady-state senescence. Journal of Thrombosis and Haemostasis. 2004;2:651-659
  123. 123. Lebois M, Josefsson EC, Lebois M, Josefsson EC. Regulation of platelet lifespan by apoptosis. Platelets;27(6):497-504. DOI: 10.3109/09537104.2016.1161739
  124. 124. Dasgupta SK et al. Platelet senescence and phosphatidylserine exposure. Transfusion. 2010;50(10):2167-2175. DOI: 10.1111/j.1537-2995.2010.02676.x.Platelet
  125. 125. Carbonara S et al. Response of severe HIV-associated thrombocytopenia to highly active antiretroviral therapy including protease inhibitors. The Journal of Infection. 2001;42(4):251-256. DOI: 10.1053/jinf.2001.0833
  126. 126. Servais J et al. HIV-associated hematologic disorders are correlated with plasma viral load and improve under highly active antiretroviral therapy. JAIDSs. 2001;28:221-225
  127. 127. Lewis W. Mitochondrial toxicity of antiviral drugs. Nature Medicine. 1995;1(5):417-422
  128. 128. Baum PD, Sullam PM, Stoddart CA, McCune JM. Abacavir increases platelet reactivity via competitive inhibition of soluble guanylyl cyclase. AIDS. 2011;25(18):2243-2248. DOI: 10.1097/QAD.0b013e32834d3cc3
  129. 129. Khawaja AA et al. HIV antivirals affect endothelial activation and endothelial-platelet crosstalk. Circulation Research. 2020:1365-1380. DOI: 10.1161/CIRCRESAHA.119.316477
  130. 130. Chini M et al. Effects of highly active antiretroviral therapy on platelet activating factor metabolism in naïve HIV-infected patients: II study of the abacavir/lamivudine/efavirenz haart regimen. International Journal of Immunopathology and Pharmacology. 2012;25(1):247-258. DOI: 10.1177/039463201202500127
  131. 131. Jaschinski NJ, Greenberg L, Beesgaard B, et al. Recent abacavir use and incident cardiovascular disease in contemporary treated people living with HIV (PLWH) within the RESPOND cohort consortium. In: 18th European AIDS Conference. London, EACS 2021, October 27-30, 2021. Abstract BPD1/3
  132. 132. Dorjee K, Baxi SM, Reingold AL, Hubbard A. Risk of cardiovascular events from current, recent, and cumulative exposure to abacavir among persons living with HIV who were receiving antiretroviral therapy in the United States: A cohort study. BMC Infectious Diseases. 2017;17(1):1-12. DOI: 10.1186/s12879-017-2808-8
  133. 133. Baker JV, Hullsiek KH, Bradford RL, Prosser R, Tracy RP, Key NS. Circulating levels of tissue factor microparticle procoagulant activity are reduced with antiretroviral therapy and are associated with persistent inflammation and coagulation activation among HIV-positive patients. Journal of Acquired Immune Deficiency Syndromes. 2013;63(3):367-371. DOI: 10.1097/QAI.0b013e3182910121
  134. 134. Funderburg NT et al. Shared monocyte subset phenotypes in HIV-1 infection and in uninfected subjects with acute coronary syndrome. Blood. 2012;120(23):4599-4608. DOI: 10.1182/blood-2012-05-433946
  135. 135. Funderburg NT et al. Increased tissue factor expression on circulating monocytes in chronic HIV infection: Relationship to in vivo coagulation and immune activation. Blood. 2010;115(2):161-167. DOI: 10.1182/blood-2009-03-210179
  136. 136. Østerud B. The role of platelets in decrypting monocyte tissue factor. Seminars in Hematology. 2001;38(Suppl 12):2-5. DOI: 10.1053/shem.2001.29508
  137. 137. Scholz T, Temmler U, Krause S, Heptinstall S, Lösche W. Transfer of tissue factor from platelets to monocytes: Role of platelet-derived microvesicles and CD62P. Thrombosis and Haemostasis. 2002;88(6):1033-1038. DOI: 10.1055/s-0037-1613351
  138. 138. Lösche W, Scholz T, Temmler U, Oberle V, Claus RA. Platelet-derived microvesicles transfer tissue factor to monocytes but not to neutrophils. Platelets. 2004;15(2):109-115. DOI: 10.1080/09537100310001649885
  139. 139. De Pablo-Bernal RS et al. TNF-α levels in HIV-infected patients after long-term suppressive cART persist as high as in elderly, HIV-uninfected subjects. The Journal of Antimicrobial Chemotherapy. 2014;69(11):3041-3046. DOI: 10.1093/jac/dku263
  140. 140. Laurence J, Elhadad S, Ahamed J. HIV-associated cardiovascular disease: Importance of platelet activation and cardiac fibrosis in the setting of specific antiretroviral therapies. Open Heart. 2018;5(2):1-13. DOI: 10.1136/openhrt-2018-000823
  141. 141. Feinstein MJ et al. Characteristics, prevention, and Management of Cardiovascular Disease in people living with HIV: A scientific statement from the American Heart Association. Circulation. 2019;140(2):e98-e124. DOI: 10.1161/CIR.0000000000000695
  142. 142. Currier JS et al. Coronary heart disease in HIV-infected individuals. JAIDS. 2003;33:506-512
  143. 143. Boccara F et al. HIV and coronary heart disease: Time for a better understanding. Journal of the American College of Cardiology. 2013;61(5):511-523. DOI: 10.1016/j.jacc.2012.06.063
  144. 144. Benjamin LA et al. HIV infection and stroke : Current perspectives and future directions. Lancet Neurology. 2012;11(10):878-890. DOI: 10.1016/S1474-4422(12)70205-3
  145. 145. Lin HL, Muo CH, Lin CY, Chen HJ, Chen PC. Incidence of stroke in patients with HIV infection: A population-based study in Taiwan. PLoS One. 2019;14(5):1-14. DOI: 10.1371/journal.pone.0217147
  146. 146. Currier JS et al. Epidemiological evidence for cardiovascular disease in HIV-infected patients and relationship to highly active antiretroviral therapy. Circulation. 2008;118(2):e29-e35. DOI: 10.1161/CIRCULATIONAHA.107.189624
  147. 147. Obel N et al. Ischemic heart disease in HIV-infected and HIV-uninfected individuals: A population-based cohort study. Clinical Infectious Diseases. 2007;44(12):1625-1631. DOI: 10.1086/518285
  148. 148. Archer O et al. Patients living with HIV and coronary disease: Are we using appropriate anti platelets as part of dual antiplatelet therapy? Cardiology & Vascular Research. 2021;5(1):1-5. DOI: 10.33425/2639-8486.1094
  149. 149. Simpson SR, Singh MV, Dewhurst S, Schifitto G, Maggirwar SB. Platelets function as an acute viral reservoir during HIV-1 infection by harboring virus and T-cell complex formation. Blood Advances. 2020;4(18):4512-4521. DOI: 10.1182/bloodadvances.2020002420

Written By

Gordon Ogweno

Submitted: 02 June 2022 Reviewed: 07 June 2022 Published: 22 July 2022