Skip to main content

The role of HIV Tat protein in HIV-related cardiovascular diseases

Abstract

The human immunodeficiency virus (HIV) is a major global public health issue. HIV-related cardiovascular disease remains a leading cause of morbidity and mortality in HIV positive patients. HIV Tat is a regulatory protein encoded by tat gene of HIV-1, which not only promotes the transcription of HIV, but it is also involved in the pathogenesis of HIV-related complications. This review is aimed at summarizing the current understanding of Tat in HIV-related cardiovascular diseases.

Background

The human immunodeficiency virus (HIV) is a major global public health issue. It has claimed more than 35 million lives so far. In 2016, 1 million people died from HIV-related causes globally [1]. With the advent of highly active antiretroviral therapy (HAART), the life expectancy of HIV patients was extensively increased and HIV infection has become a chronic disease [2, 3]. Presently, alleviation of the complications induced by long term HIV infection remains an unresolved problem. Among all HIV-related complications, cardiovascular disease remains a leading cause of mortality in HIV positive patients [4]. Cardiovascular involvement in HIV-infection was first described in 1983 by Autran et al. who noted myocardial Kaposi’s sarcoma at autopsy [5]. Thereafter, additional research revealed the association between HIV infection and cardiovascular diseases. HIV infection increased the risk of cardiovascular disease, which is independent of antiretroviral therapies [6, 7]. Though some antiretroviral drugs, such as zidovudine, have a deleterious effect on myocardium, HAART has dramatically decreased the incidence of cardiomyopathy and mortality of HIV infected patients [8, 9]. Therefore, over the past few decades, scientific research has increasingly focused on unraveling the mechanism and role of HIV-infection in the pathogenesis of cardiovascular diseases

The characteristics of HIV Tat

The RNA genome of HIV consists of at least nine genes, including gag, pol, env, tat, rev, nef, vif, vpr, and vpu. Tat stands for “trans-activator of transcription”, which is a small nuclear protein encoded by the tat gene in HIV-1. It consists of 86–101 amino acids depending on the subtype. During the process of HIV replication, Tat serves as a trans-activator that drastically enhances the efficiency of viral transcription [10, 11]. Tat is encoded by two exons, highly conserved regions (1–72 amino acids) and sub-type dependent regions (73–86 to 101 amino acids). The first region contains five functional domains which are: the acidic/proline rich, the cysteine-rich/ZnF, the core, the basic and the glutamine-rich domains. The first three regions constitute a minimal activation domain. The basic region contains nuclear localization signal and RNA-binding domain. The cysteine-rich/ZnF region represents the protein transduction domain [12]. The structure of Tat gives it the properties of both transcription promotion and membrane transduction.

HIV Tat and cardiovascular diseases

HIV Tat and cardiomyopathy

HIV infection is recognized as an important cause of dilated cardiomyopathy. Cohen et al. reported the connection between HIV infection and dilated cardiomyopathy for the first time in 1986 [13]. HIV-related cardiomyopathy with reduced systolic function occurred much more frequently before the advent of HAART therapy or in developing countries in which HAART is not widely available [8, 14, 15]. After HAART with excellent virologic control, most patients are minimally symptomatic or have diastolic dysfunction [8, 16,17,18].

HIV-transgenic mice (Tg26) is a well-described murine line of NL4-3 Δgag/pol that expresses HIV-related proteins and develops HIV-related complications in the hemizygotes [19]. Under basal conditions, WT and Tg26 mice have normal systolic and diastolic function. Cardiac contractile dysfunction in the isolated work-performing heart from Tg26 mice was originally reported by Lewis et al. which was worsened by antiretroviral treatment with zidovudine [20]. Similarly, Cheung et al. found that Tg26 mice exhibits normal cardiac contractile function under basal conditions, but with less tolerance on surgical stress and the down regulation of Bcl2-associated athanogene 3 (BAG3) induced contractile abnormalities [21]. Though they observed different phenotype in HIV transgenic mice, there were similarities that transgenic expression of HIV facilitates cardiac dysfunction.

Further study proved that Tat protein plays an important role in HIV-related cardiac dysfunction. Targeted myocardial HIV Tat transgenic mice showed a depression in myocardial systolic and diastolic functions [22]. The occurrence of cardiac dysfunction was proved to be related with mitochondrial ultrastructural damage. Profound cardiac mitochondrial damage has been associated with the death of homozygote +/+ Tat mice within 14 days, in contrast to +/− Tat high and +/− Tat low mice which developed, matured and survived up to 2 years [23].

Latest research reveals that Tat has profound impacts on cardiomyocytes. Transfection of adenoviral-Tat impaired the uptake of mitochondrial Ca2+ ([Ca2+]m) and the electrophysiological activity of cardiomyocytes, which also aggravated hypoxia/re-oxygenation-induced cardiomyocyte injury through interference with the initiation of autophagy and the clearance of autophagic proteins [24]. It is worth mentioning that methamphetamine, a cardiotoxic stimulant, is abused epidemically and is frequently associated with acquisition of HIV-1 infection.

A study addressed the individual and combined effects of HIV-1 and methamphetamine on cardiac dysfunction using a transgenic mouse model of HIV infection. The results demonstrated that Tat and methamphetamine increased calcium uptake and promoted mitochondrial dysfunction in cardiomyocytes, this altered the DNA methylation and expression of CACNA1C, that encodes the alpha 1c (Cav1.2) subunit of the L-type calcium channel [25].

HIV and arrhythmia

Corrected QT (QTc) prolongation is predictive of cardiovascular mortality. Frequent occurrence of QT interval prolongation have been found in HIV-positive patients [26, 27]. The application of HIV protease inhibitors (PIs) are suspected to be responsible for HIV-related long QT syndrome (LQTs) through the blockade of cardiac rapidly activating delayed rectifier K+ current (IKr) channels encoded by human ether-a-go-go-related gene (hERG) [28]. A study proved that a low CD4+ cell count is associated with LQTs, specifically in this study, the HAART was not associated with QTc prolongation in HIV-positive patients [29].

In HIV transgenic mice, cardiac repolarization is prolonged compared with wild-type littermates, which is at least partially attributed to the reduction of transient outward K+ current (Ito). Thus, it appears that HIV itself contributes to the delayed repolarization observed in the transgenic mice [30]. Subsequent studies explored the effect and the underlying mechanisms of Tat in cardiomyocytes. It was also observed that Tat protein prolonged QTc interval in guinea pig. Correspondingly, isolated cardiomyocytes form Tat-treated guinea pig exhibited prolonged action potential duration at 90% of repolarization (APD90) and reduced IKr [31]. Furthermore, incubation of Tat protein also significantly decreased IKr in hERG stably expressed HEK293 cells through the inhibition of hERG protein expression but with no effect on the hERG mRNA expression [31]. Similar results were achieved in human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs). Tat transfection in heterologous expression systems led to a decrease in IKr and IKs through sequestering membrane phosphatidylinositol-(4,5)-bisphosphate (PIP2), with no significant alteration in ICa-L [32].

In addition, Tat could affect both cardiomyocytes and neurons thus inducing bradycardia. Transgenic mice with myocardial overexpression of Tat exhibits a relative bradycardia phenotype, which is associated with reduced susceptibility of adrenergic responsiveness [22]. Further study proposes that Tat also excites cardiac parasympathetic neurons of nucleus ambiguus and produces a sustained bradycardia. Tat-induced Ca2+ elevation in neurons was largely attributed to the promotion of lysosomal Ca2+ mobilization, Ca2+ release via inositol 1,4,5-trisphosphate-sensitive endoplasmic reticulum pools, and Ca2+ influx via transient receptor potential vanilloid type 2 (TRPV2) channels [33].

HIV Tat and atherosclerosis

HIV-seropositive patients are at higher risk for atherosclerosis than HIV-seronegative persons. This has been variably attributed to antiretroviral treatment, advanced immunodeficiency, and HIV-associated inflammation [34, 35]. Priscilla Y et al. proposed that increased atherosclerosis with HIV infection occurs in the absence of antiretroviral therapy, detectable viremia, or overt immunodeficiency [36]. Francisci D et al. also proposed that HIV infection induces alterations of markers of endothelial function, which is independent of antiretroviral treatment [37]. Therefore, persistent HIV-associated inflammation and endothelial dysfunction might be potential causes for accelerated atherosclerosis in HIV infection.

Carotid intima-media thickness (IMT), an indicator of generalized atherosclerosis, is higher in HIV patients than in age-matched control subjects, which is associated with classic coronary risk factors and with low CD4+ T cell count [38]. Beyond traditional cardiovascular disease risk factors, low CD4+ T-cell count is the most robust risk factor for increased subclinical carotid atherosclerosis in HIV-infected patients [39]. In animal models, HIV transgenic mice exhibit endothelial dysfunction with increased intima-media thickness and arterial stiffness. Furthermore, the arteries from HIV transgenic mice show decreased elastin content, increased cathepsin K and cathepsin S activity and mechanical residual stress. Thus, HIV transgenic mice exhibition of pre-clinical markers of vascular remodeling are consistent with early on-set atherosclerosis [40]. Although there is no direct evidence that Tat causes atherosclerosis, circumstantial evidence implicates a relationship between them. Tat significantly decreased endothelium-dependent vasorelaxation and endothelial nitric oxide synthase (eNOS) production in porcine coronary arteries [41].

The effect of Tat on inflammation and immune activation

Chronic immune activation and inflammation are risk factors of cardiovascular diseases. HIV-infection is also associated with increase in inflammation factors and immune activation. The inflammatory and coagulation biomarkers IL-6, hsCRP and D-dimer expression levels are associated with an increased risk of cardiovascular disease in HIV-infected individuals [42]. The immune hyperactivation of HIV-infected T cells is mediated by Tat [43]. Low concentrations (0.1–1 nM) of Tat protein increase the anti-apoptotic ability of CD4+ T lymphoblastoid Jurkat cells through stimulation of the catalytic activity of phosphatidylinositol 3-kinase (PI3K)/Akt pathway [44]. Although HAART suppresses HIV replication, it is often unable to restore immune homeostasis [45]. Thus, immunization with Tat acts in synergy with HAART to help in restoring immune homeostasis [46].

The inflammatory vascular environment is critical for the initiation and development of atherosclerosis. Tat induces vascular inflammation through the activation of nuclear factor-kappa B (NF-κB) in human vascular endothelial cells (HUVECs) and leads to the upregulation of inflammatory mediators, including IL-1β, MCP-1, VCAM-1 and E-selectin, which could be attenuated by estrogen [47]. Further study found that Tat stimulated upregulation of intercellular cell adhesion molecules specifically ICAM-1 in HUVECs was through the suppression of miR-221/-222 in a NF-κB-dependent pathway [48]. Tat-related impairment of the survival and differentiation of mesenchymal stem cells might play an important role in vessel damage and formation of the atherosclerotic lesions observed in HIV-infected patients and this could be considered an additional important mechanism involved in promoting vascular damage and atherosclerosis in the course of HIV disease [49]. In addition, Tat could mediate the induction of adhesion molecules and also function as an exogenous cytokine in the activation of human endothelial cells, which upregulates E-selectin expression, enhances the secretion of IL-6, and synergizes with TNF in mediating these effects [50].

HIV Tat and pulmonary arterial hypertension

HIV infected patients have a greater incidence of pulmonary arterial hypertension (PAH) compared to the general population. Specifically, in comparison with the incidence of idiopathic PAH in the general population (1–2 per million), HIV-infected patients have a 2500-fold increased risk of developing PAH [51]. The development of HIV-related PAH reduces the probability of survival by half as compared with HIV-infected individuals without HIV-related PAH [52]. Depending on gender and ethnic differences, the proportion of mortality due to pulmonary heart disease and diseases of pulmonary circulation in HIV-infected adults ranges from 4.3% to 17.5% of overall cardiovascular disease mortality in a cohort of HIV-infected adults in the United States [53]. The advent of HAART ameliorates the outcome of patients with HIV-related PAH [52, 54].

HIV infection and intravenous drug abuse have been identified as common independent risk factors of PAH. For example, morphine potentiates HIV Tat protein-mediated apoptosis and proliferation of pulmonary endothelial cells, which may have led to strikingly more pronounced pulmonary vascular remodeling than exposed alone [55]. The processes of PAH progression include the transformation of endothelial cells from initial apoptosis to apoptosis-resistant hyper-proliferation. A recent study found a potential link between autophagy and HIV-mediated enhancement of proliferation of apoptosis-resistant pulmonary endothelial cells. The initial exposure of pulmonary endothelial cells to morphine and Tat results in independent activation of both autophagy and apoptosis. However, chronic increase in autophagy reduces the incidence of oxidative stress and apoptosis that allows the cells to adapt to stress leading to cell survival and uncontrolled proliferation of pulmonary endothelial cells. Therefore, morphine in combination with Tat results in the induction of autophagy in pulmonary endothelial cells that may lead to an increase in severity of angio-proliferative remodeling of the pulmonary vasculature [56].

Tat transgenic mice

Transgenic technology has made it possible to investigate the pathogenic mechanism of HIV in mice. HIV transgenic mice develops HIV-infection related complications. Some HIV encoded proteins, including Tat, Nef and Vpr, are important determinants for the pathogenicity of HIV [57, 58]. Tat is one of the extensively investigated proteins, which is sufficient to induce some HIV-related complications in the absence of HIV infection. To clarify the specific role of Tat, a model of transgenic mice expressing Tat protein was established. Tat expression induces dermal lesions resembling Kaposi’s sarcoma and facilitates the generation and progression of tumors/cancers of different histotypes in transgenic mice [59,60,61,62]. In Tat-transgenic mice, Tat expression is sufficient to cause neuropathologies similar to most of those noted in the brain of HIV-infected patients [63]. Tat is an immunological manipulator, which increases the production of pro-inflammatory cytokines and induces lymphoid hyperplasia [64, 65]. With an α-myosin heavy chain promoter, Tat was selectively expressed in mice cardiac myocyte. Targeted myocardial transgenic expression of HIV Tat in mice results in relative bradycardia and contractile dysfunction, this model has helped to provide a better understanding of the pathophysiological mechanisms of Tat in HIV-related cardiomyopathy [22, 23].

Conclusions

All these findings confirmed that Tat protein not only promotes the transcription of HIV but also participates in the pathogenesis of HIV-associated cardiovascular complications (Fig. 1). It is a particularly good target for prevention, treatment of HIV and development of antiviral drugs. Tat-specific antibodies appear to be an excellent method to prevent HIV acquisition and spreading. It has been proven to prevent and/or control infection with pathogenic simian/human immunodeficiency virus (SHIV) in nonhuman primates [66, 67].

Fig. 1
figure 1

HIV Tat and HIV-related cardiovascular diseases

In recent years, a randomized phase II exploratory clinical trial found that Tat immunization is safe, well tolerated and could induce anti-Tat antibodies in most patients. It promoted a durable and significant restoration of immune homeostasis and induced a significant reduction of blood proviral DNA in patients on effective combination anti-retroviral therapy regimen. Thus, Tat immunization represents a promising pathogenesis-driven intervention to intensify HAART efficacy [68]. HIV-1 Oyi was a strain cloned from a seropositive patient [69]. Tat Oyi vaccine can generate neutralizing antibodies against Tat variants regardless of their mutations, thereby establishing a rationale for testing Tat Oyi as a potential therapeutic vaccine in human trials [70]. The Tat Oyi vaccine in association with combination anti-retroviral therapy may provide an efficient means of controlling the HIV-infected cell reservoir [71]. Furthermore, as a cell penetrating peptide, Tat has been developed as a drug delivery system in the treatment of cancer [72]. However, Tat and anti-cancer drugs both have cardiovascular toxicity. Whether they have synergistic injury effect on cardiovascular system remains unknown.

Besides the conventional therapy involved in circumventing the effect of TAT in HIV- related cardiovascular diseases, there are also natural herbal extracts reported to possess anti-HIV properties. For instance, Curcumin is a natural herb extract, which has anti-HIV activity. A recent study revealed that curcumin causes inhibition and degradation of Tat which may be one of the major mechanisms behind its anti-HIV activity [73]. Also, Curcumin has been shown to possess anti-inflammatory and ameliorative properties, thereby playing a protective role in cardiovascular disease [74]. These findings suggest that harnessing the potential efficacy of natural herbs might be a source of new drugs for the treatment of HIV infection and related cardiovascular complications, however further studies could still be carried out on Curcumin in relation to HIV-related cardiovascular diseases.

This review has been able to show the effects of Tat on HIV-related cardiovascular diseases. Further investigation is still needed to elucidate the molecular mechanism of Tat and HIV-related cardiovascular disorders which may suggest novel insights and therapeutic targets.

Abbreviations

Akt:

protein kinase B

APD90 :

action potential duration at 90% of repolarization

BAG3:

Bcl2-associated athanogene 3

[Ca2+]m :

mitochondrial Ca2+

eNOS:

endothelial nitric oxide synthase

HAART:

highly active antiretroviral therapy

hERG:

human ether-a-go-go-related gene

hiPSC-CMs:

human induced pluripotent stem cell derived cardiomyocytes

HIV:

human immunodeficiency virus

HUVECs:

human vascular endothelial cells

IKr :

rapidly activating delayed rectifier potassium current

Ito :

outward potassium current

LQTs:

long QT syndrome

NF-κB:

nuclear factor-kappa B

PIs:

Protease inhibitors

PIP2 :

phosphatidylinositol-(4,5)-bisphosphate

PI3K:

phosphatidylinositol 3-kinase

QTc:

corrected QT

Tat:

trans-activator of transcription

TRPV2:

transient receptor potential vanilloid type 2

References

  1. World Health Organization. Fact sheet on HIV/AIDS. http://www.who.int/mediacentre/factsheets/fs360/en/. Accessed 30 July 2017.

  2. Mocroft A, Ledergerber B, Katlama C, Kirk O, Reiss P, d’Arminio Monforte A, Knysz B, Dietrich M, Phillips AN, Lundgren JD, Euro Ssg. Decline in the AIDS and death rates in the EuroSIDA study: an observational study. Lancet. 2003;362:22–9.

    Article  CAS  PubMed  Google Scholar 

  3. Palella FJ Jr, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, Aschman DJ, Holmberg SD. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med. 1998;338:853–60.

    Article  PubMed  Google Scholar 

  4. Hemkens LG, Bucher HC. HIV infection and cardiovascular disease. Eur Heart J. 2014;35:1373–81.

    Article  CAS  PubMed  Google Scholar 

  5. Autran B, Gorin I, Leibowitch M, Laroche L, Escande JP, Hewitt J, Marche C. AIDS in a Haitian woman with cardiac Kaposi’s sarcoma and Whipple’s disease. Lancet. 1983;1:767–8.

    Article  CAS  PubMed  Google Scholar 

  6. d’Amati G, di Gioia CR, Gallo P. Pathological findings of HIV-associated cardiovascular disease. Ann NY Acad Sci. 2001;946:23–45.

    Article  PubMed  Google Scholar 

  7. Prendergast BD. HIV and cardiovascular medicine. Heart. 2003;89:793–800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Remick J, Georgiopoulou V, Marti C, Ofotokun I, Kalogeropoulos A, Lewis W, Butler J. Heart failure in patients with human immunodeficiency virus infection: epidemiology, pathophysiology, treatment, and future research. Circulation. 2014;129:1781–9.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Patel K, Van Dyke RB, Mittleman MA, Colan SD, Oleske JM, Seage GR 3rd, International Maternal Pediatric Adolescent ACTCST. The impact of HAART on cardiomyopathy among children and adolescents perinatally infected with HIV-1. AIDS. 2012;26:2027–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Frankel AD, Young JA. HIV-1: fifteen proteins and an RNA. Annu Rev Biochem. 1998;67:1–25.

    Article  CAS  PubMed  Google Scholar 

  11. Debaisieux S, Rayne F, Yezid H, Beaumelle B. The ins and outs of HIV-1 Tat. Traffic. 2012;13:355–63.

    Article  CAS  PubMed  Google Scholar 

  12. Tahirov TH, Babayeva ND, Varzavand K, Cooper JJ, Sedore SC, Price DH. Crystal structure of HIV-1 Tat complexed with human P-TEFb. Nature. 2010;465:747–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cohen IS, Anderson DW, Virmani R, Reen BM, Macher AM, Sennesh J, DiLorenzo P, Redfield RR. Congestive cardiomyopathy in association with the acquired immunodeficiency syndrome. N Engl J Med. 1986;315:628–30.

    Article  CAS  PubMed  Google Scholar 

  14. Herskowitz A, Vlahov D, Willoughby S, Chaisson RE, Schulman SP, Neumann DA, Baughman KL. Prevalence and incidence of left ventricular dysfunction in patients with human immunodeficiency virus infection. Am J Cardiol. 1993;71:955–8.

    Article  CAS  PubMed  Google Scholar 

  15. Barbaro G, Barbarini G, Di Lorenzo G. Early impairment of systolic and diastolic function in asymptomatic HIV-positive patients: a multicenter echocardiographic and echo-Doppler study. The Gruppo Italiano Per lo Studio Cardiologico dei Pazienti Affetti da AIDS. AIDS Res Hum Retroviruses. 1996;12:1559–63.

    Article  CAS  PubMed  Google Scholar 

  16. Twagirumukiza M, Nkeramihigo E, Seminega B, Gasakure E, Boccara F, Barbaro G. Prevalence of dilated cardiomyopathy in HIV-infected African patients not receiving HAART: a multicenter, observational, prospective, cohort study in Rwanda. Curr HIV Res. 2007;5:129–37.

    Article  CAS  PubMed  Google Scholar 

  17. Reinsch N, Neuhaus K, Esser S, Potthoff A, Hower M, Brockmeyer NH, Erbel R, Neumann T, German Competence Network for Heart F, German Competence Network for HA. Prevalence of cardiac diastolic dysfunction in HIV-infected patients: results of the HIV-HEART study. HIV Clin Trials. 2010;11:156–62.

    Article  PubMed  Google Scholar 

  18. Reinsch N, Kahlert P, Esser S, Sundermeyer A, Neuhaus K, Brockmeyer N, Potthoff A, Erbel R, Buck T, Neumann T. Echocardiographic findings and abnormalities in HIV-infected patients: results from a large, prospective, multicenter HIV-heart study. Am J Cardiovasc Dis. 2011;1:176–84.

    PubMed  PubMed Central  Google Scholar 

  19. De SK, Devadas K, Notkins AL. Elevated levels of tumor necrosis factor alpha (TNF-alpha) in human immunodeficiency virus type 1-transgenic mice: prevention of death by antibody to TNF-alpha. J Virol. 2002;76:11710–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lewis W, Grupp IL, Grupp G, Hoit B, Morris R, Samarel AM, Bruggeman L, Klotman P. Cardiac dysfunction occurs in the HIV-1 transgenic mouse treated with zidovudine. Lab Invest. 2000;80:187–97.

    Article  CAS  PubMed  Google Scholar 

  21. Cheung JY, Gordon J, Wang J, Song J, Zhang XQ, Tilley DG, Gao E, Koch WJ, Rabinowitz J, Klotman PE, et al. Cardiac dysfunction in HIV-1 transgenic mouse: role of stress and BAG3. Clin Transl Sci. 2015;8:305–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Fang Q, Kan H, Lewis W, Chen F, Sharma P, Finkel MS. Dilated cardiomyopathy in transgenic mice expressing HIV Tat. Cardiovasc Toxicol. 2009;9:39–45.

    Article  CAS  PubMed  Google Scholar 

  23. Raidel SM, Haase C, Jansen NR, Russ RB, Sutliff RL, Velsor LW, Day BJ, Hoit BD, Samarel AM, Lewis W. Targeted myocardial transgenic expression of HIV Tat causes cardiomyopathy and mitochondrial damage. Am J Physiol Heart Circ Physiol. 2002;282:H1672–8.

    Article  CAS  PubMed  Google Scholar 

  24. Tahrir FG, Shanmughapriya S, Ahooyi TM, Knezevic T, Gupta MK, Kontos CD, McClung JM, Madesh M, Gordon J, Feldman AM, et al. Dysregulation of mitochondrial bioenergetics and quality control by HIV-1 Tat in cardiomyocytes. J Cell Physiol. 2018;233:748–58.

    Article  CAS  PubMed  Google Scholar 

  25. Koczor CA, Fields E, Jedrzejczak MJ, Jiao Z, Ludaway T, Russ R, Shang J, Torres RA, Lewis W. Methamphetamine and HIV-Tat alter murine cardiac DNA methylation and gene expression. Toxicol Appl Pharmacol. 2015;288:409–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Reinsch N, Buhr C, Krings P, Kaelsch H, Neuhaus K, Wieneke H, Erbel R, Neumann T, German Heart Failure N. Prevalence and risk factors of prolonged QTc interval in HIV-infected patients: results of the HIV-HEART study. HIV Clin Trials. 2009;10:261–8.

    Article  PubMed  Google Scholar 

  27. Ogunmola OJ, Oladosu YO, Olamoyegun MA. QTc interval prolongation in HIV-negative versus HIV-positive subjects with or without antiretroviral drugs. Ann Afr Med. 2015;14:169–76.

    Article  PubMed  Google Scholar 

  28. Anson BD, Weaver JG, Ackerman MJ, Akinsete O, Henry K, January CT, Badley AD. Blockade of HERG channels by HIV protease inhibitors. Lancet. 2005;365:682–6.

    Article  CAS  PubMed  Google Scholar 

  29. Gili S, Mancone M, Ballocca F, Grosso Marra W, Calcagno A, D’Ettorre G, Cannillo M, D’Ascenzo F, Orofino G, Marruncheddu L, et al. Prevalence and predictors of long corrected QT interval in HIV-positive patients: a multicenter study. J Cardiovasc Med (Hagerstown). 2017;18:539–44.

    Article  Google Scholar 

  30. Brouillette J, Grandy SA, Jolicoeur P, Fiset C. Cardiac repolarization is prolonged in CD4C/HIV transgenic mice. J Mol Cell Cardiol. 2007;43:159–67.

    Article  CAS  PubMed  Google Scholar 

  31. Bai YL, Liu HB, Sun B, Zhang Y, Li Q, Hu CW, Zhu JX, Gong DM, Teng X, Zhang Q, et al. HIV Tat protein inhibits hERG K+ channels: a potential mechanism of HIV infection induced LQTs. J Mol Cell Cardiol. 2011;51:876–80.

    Article  CAS  PubMed  Google Scholar 

  32. Es-Salah-Lamoureux Z, Jouni M, Malak OA, Belbachir N, Al Sayed ZR, Gandon-Renard M, Lamirault G, Gauthier C, Baro I, Charpentier F, et al. HIV-Tat induces a decrease in IKr and IKsvia reduction in phosphatidylinositol-(4,5)-bisphosphate availability. J Mol Cell Cardiol. 2016;99:1–13.

    Article  CAS  PubMed  Google Scholar 

  33. Brailoiu E, Deliu E, Sporici RA, Benamar K, Brailoiu GC. HIV-1-Tat excites cardiac parasympathetic neurons of nucleus ambiguus and triggers prolonged bradycardia in conscious rats. Am J Physiol Regul Integr Comp Physiol. 2014;306:R814–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Siedner MJ, Kim JH, Nakku RS, Bibangambah P, Hemphill L, Triant VA, Haberer JE, Martin JN, Mocello AR, Boum Y 2nd, et al. Persistent immune activation and carotid atherosclerosis in HIV-infected ugandans receiving antiretroviral therapy. J Infect Dis. 2016;213:370–8.

    Article  CAS  PubMed  Google Scholar 

  35. Coll B, Parra S, Alonso-Villaverde C, Aragones G, Montero M, Camps J, Joven J, Masana L. The role of immunity and inflammation in the progression of atherosclerosis in patients with HIV infection. Stroke. 2007;38:2477–84.

    Article  PubMed  Google Scholar 

  36. Hsue PY, Hunt PW, Schnell A, Kalapus SC, Hoh R, Ganz P, Martin JN, Deeks SG. Role of viral replication, antiretroviral therapy, and immunodeficiency in HIV-associated atherosclerosis. AIDS. 2009;23:1059–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Francisci D, Giannini S, Baldelli F, Leone M, Belfiori B, Guglielmini G, Malincarne L, Gresele P. HIV type 1 infection, and not short-term HAART, induces endothelial dysfunction. AIDS. 2009;23:589–96.

    Article  PubMed  Google Scholar 

  38. Hsue PY, Lo JC, Franklin A, Bolger AF, Martin JN, Deeks SG, Waters DD. Progression of atherosclerosis as assessed by carotid intima-media thickness in patients with HIV infection. Circulation. 2004;109:1603–8.

    Article  PubMed  Google Scholar 

  39. Kaplan RC, Kingsley LA, Gange SJ, Benning L, Jacobson LP, Lazar J, Anastos K, Tien PC, Sharrett AR, Hodis HN. Low CD4+ T-cell count as a major atherosclerosis risk factor in HIV-infected women and men. AIDS. 2008;22:1615–24.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Hansen L, Parker I, Sutliff RL, Platt MO, Gleason RL Jr. Endothelial dysfunction, arterial stiffening, and intima-media thickening in large arteries from HIV-1 transgenic mice. Ann Biomed Eng. 2013;41:682–93.

    Article  PubMed  Google Scholar 

  41. Paladugu R, Fu W, Conklin BS, Lin PH, Lumsden AB, Yao Q, Chen C. Hiv Tat protein causes endothelial dysfunction in porcine coronary arteries. J Vasc Surg. 2003;38:549–55 (discussion 555-6).

    Article  PubMed  Google Scholar 

  42. Duprez DA, Neuhaus J, Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, Lane HC, Ledergerber B, Lundgren J, et al. Inflammation, coagulation and cardiovascular disease in HIV-infected individuals. PLoS ONE. 2012;7:e44454.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ott M, Emiliani S, Van Lint C, Herbein G, Lovett J, Chirmule N, McCloskey T, Pahwa S, Verdin E. Immune hyperactivation of HIV-1-infected T cells mediated by Tat and the CD28 pathway. Science. 1997;275:1481–5.

    Article  CAS  PubMed  Google Scholar 

  44. Borgatti P, Zauli G, Colamussi ML, Gibellini D, Previati M, Cantley LL, Capitani S. Extracellular HIV-1 Tat protein activates phosphatidylinositol 3- and Akt/PKB kinases in CD4+ T lymphoblastoid Jurkat cells. Eur J Immunol. 1997;27:2805–11.

    Article  CAS  PubMed  Google Scholar 

  45. Kelley CF, Kitchen CM, Hunt PW, Rodriguez B, Hecht FM, Kitahata M, Crane HM, Willig J, Mugavero M, Saag M, et al. Incomplete peripheral CD4+ cell count restoration in HIV-infected patients receiving long-term antiretroviral treatment. Clin Infect Dis. 2009;48:787–94.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Ensoli B, Bellino S, Tripiciano A, Longo O, Francavilla V, Marcotullio S, Cafaro A, Picconi O, Paniccia G, Scoglio A, et al. Therapeutic immunization with HIV-1 Tat reduces immune activation and loss of regulatory T-cells and improves immune function in subjects on HAART. PLoS ONE. 2010;5:e13540.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Lee YW, Eum SY, Nath A, Toborek M. Estrogen-mediated protection against HIV Tat protein-induced inflammatory pathways in human vascular endothelial cells. Cardiovasc Res. 2004;63:139–48.

    Article  CAS  PubMed  Google Scholar 

  48. Duan M, Yao H, Hu G, Chen X, Lund AK, Buch S. HIV Tat induces expression of ICAM-1 in HUVECs: implications for miR-221/-222 in HIV-associated cardiomyopathy. PLoS ONE. 2013;8:e60170.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gibellini D, Miserocchi A, Tazzari PL, Ricci F, Clo A, Morini S, Ponti C, Pasquinelli G, Bon I, Pagliaro P, et al. Analysis of the effects of HIV-1 Tat on the survival and differentiation of vessel wall-derived mesenchymal stem cells. J Cell Biochem. 2012;113:1132–41.

    Article  CAS  PubMed  Google Scholar 

  50. Hofman FM, Wright AD, Dohadwala MM, Wong-Staal F, Walker SM. Exogenous tat protein activates human endothelial cells. Blood. 1993;82:2774–80.

    CAS  PubMed  Google Scholar 

  51. Correale M, Palmiotti GA, Lo Storto MM, Montrone D, Foschino Barbaro MP, Di Biase M, Lacedonia D. HIV-associated pulmonary arterial hypertension: from bedside to the future. Eur J Clin Invest. 2015;45:515–28.

    Article  PubMed  Google Scholar 

  52. Bigna JJ, Sime PS, Koulla-Shiro S. HIV related pulmonary arterial hypertension: epidemiology in Africa, physiopathology, and role of antiretroviral treatment. AIDS Res Ther. 2015;12:36.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Feinstein MJ, Bahiru E, Achenbach C, Longenecker CT, Hsue P, So-Armah K, Freiberg MS, Lloyd-Jones DM. Patterns of Cardiovascular Mortality for HIV-Infected Adults in the United States: 1999 to 2013. Am J Cardiol. 2016;117:214–20.

    Article  PubMed  Google Scholar 

  54. Pal J, Sen K, Sarkar G, Mandal A, Chakraborty S, Deb A. Effect of antiretroviral therapy on pulmonary hypertension in HIV patients. J Indian Med Assoc. 2013;111:845–6,849.

    Google Scholar 

  55. Spikes L, Dalvi P, Tawfik O, Gu H, Voelkel NF, Cheney P, O’Brien-Ladner A, Dhillon NK. Enhanced pulmonary arteriopathy in simian immunodeficiency virus-infected macaques exposed to morphine. Am J Respir Crit Care Med. 2012;185:1235–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Dalvi P, Sharma H, Chinnappan M, Sanderson M, Allen J, Zeng R, Choi A, O’Brien-Ladner A, Dhillon NK. Enhanced autophagy in pulmonary endothelial cells on exposure to HIV-Tat and morphine: role in HIV-related pulmonary arterial hypertension. Autophagy. 2016;12:2420–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Rosenstiel P, Gharavi A, D’Agati V, Klotman P. Transgenic and infectious animal models of HIV-associated nephropathy. J Am Soc Nephrol. 2009;20:2296–304.

    Article  PubMed  Google Scholar 

  58. Gupta MK, Kaminski R, Mullen B, Gordon J, Burdo TH, Cheung JY, Feldman AM, Madesh M, Khalili K. HIV-1 Nef-induced cardiotoxicity through dysregulation of autophagy. Sci Rep. 2017;7:8572.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Vogel J, Hinrichs SH, Reynolds RK, Luciw PA, Jay G. The HIV tat gene induces dermal lesions resembling Kaposi’s sarcoma in transgenic mice. Nature. 1988;335:606–11.

    Article  CAS  PubMed  Google Scholar 

  60. Altavilla G, Caputo A, Trabanelli C, Brocca Cofano E, Sabbioni S, Menegatti MA, Barbanti-Brodano G, Corallini A. Prevalence of liver tumours in HIV-1 tat-transgenic mice treated with urethane. Eur J Cancer. 2004;40:275–83.

    Article  CAS  PubMed  Google Scholar 

  61. Vogel J, Hinrichs SH, Napolitano LA, Ngo L, Jay G. Liver cancer in transgenic mice carrying the human immunodeficiency virus tat gene. Cancer Res. 1991;51:6686–90.

    CAS  PubMed  Google Scholar 

  62. Corallini A, Altavilla G, Pozzi L, Bignozzi F, Negrini M, Rimessi P, Gualandi F, Barbanti-Brodano G. Systemic expression of HIV-1 tat gene in transgenic mice induces endothelial proliferation and tumors of different histotypes. Cancer Res. 1993;53:5569–75.

    CAS  PubMed  Google Scholar 

  63. Kim BO, Liu Y, Ruan Y, Xu ZC, Schantz L, He JJ. Neuropathologies in transgenic mice expressing human immunodeficiency virus type 1 Tat protein under the regulation of the astrocyte-specific glial fibrillary acidic protein promoter and doxycycline. Am J Pathol. 2003;162:1693–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Garza HH Jr, Prakash O, Carr DJ. Aberrant regulation of cytokines in HIV-1 TAT72-transgenic mice. J Immunol. 1996;156:3631–7.

    CAS  PubMed  Google Scholar 

  65. Vellutini C, Horschowski N, Philippon V, Gambarelli D, Nave KA, Filippi P. Development of lymphoid hyperplasia in transgenic mice expressing the HIV tat gene. AIDS Res Hum Retroviruses. 1995;11:21–9.

    Article  CAS  PubMed  Google Scholar 

  66. Cafaro A, Caputo A, Fracasso C, Maggiorella MT, Goletti D, Baroncelli S, Pace M, Sernicola L, Koanga-Mogtomo ML, Betti M, et al. Control of SHIV-89.6P-infection of cynomolgus monkeys by HIV-1 Tat protein vaccine. Nat Med. 1999;5:643–50.

    Article  CAS  PubMed  Google Scholar 

  67. Cafaro A, Bellino S, Titti F, Maggiorella MT, Sernicola L, Wiseman RW, Venzon D, Karl JA, O’Connor D, Monini P, et al. Impact of viral dose and major histocompatibility complex class IB haplotype on viral outcome in mauritian cynomolgus monkeys vaccinated with Tat upon challenge with simian/human immunodeficiency virus SHIV89.6P. J Virol. 2010;84:8953–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ensoli F, Cafaro A, Casabianca A, Tripiciano A, Bellino S, Longo O, Francavilla V, Picconi O, Sgadari C, Moretti S, et al. HIV-1 Tat immunization restores immune homeostasis and attacks the HAART-resistant blood HIV DNA: results of a randomized phase II exploratory clinical trial. Retrovirology. 2015;12:33.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Huet T, Dazza MC, Brun-Vezinet F, Roelants GE, Wain-Hobson S. A highly defective HIV-1 strain isolated from a healthy Gabonese individual presenting an atypical western blot. AIDS. 1989;3:707–15.

    Article  CAS  PubMed  Google Scholar 

  70. Loret E. HIV extracellular Tat: myth or reality? Curr HIV Res. 2015;13:90–7.

    Article  CAS  PubMed  Google Scholar 

  71. Loret EP, Darque A, Jouve E, Loret EA, Nicolino-Brunet C, Morange S, Castanier E, Casanova J, Caloustian C, Bornet C, et al. Intradermal injection of a Tat Oyi-based therapeutic HIV vaccine reduces of 1.5 log copies/mL the HIV RNA rebound median and no HIV DNA rebound following cART interruption in a phase I/II randomized controlled clinical trial. Retrovirology. 2016;13:21.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Hu S, Wang T, Pei X, Cai H, Chen J, Zhang X, Wan Q, Wang J. Synergistic enhancement of antitumor efficacy by PEGylated Multi-walled carbon nanotubes modified with cell-penetrating peptide TAT. Nanoscale Res Lett. 2016;11:452.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Ali A, Banerjea AC. Curcumin inhibits HIV-1 by promoting Tat protein degradation. Sci Rep. 2016;6:27539.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Wongcharoen W, Phrommintikul A. The protective role of curcumin in cardiovascular diseases. Int J Cardiol. 2009;133:145–51.

    Article  PubMed  Google Scholar 

Download references

Authors’ contributions

YB designed and edited the paper; YJ and LC wrote the paper. FMB edited the paper. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Funding

This project was supported by National Natural Science Foundation of China (Grant No. 81673426) and National Natural Science Foundation of Heilongjiang Province (Grant No. LC2015034).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yunlong Bai.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jiang, Y., Chai, L., Fasae, M.B. et al. The role of HIV Tat protein in HIV-related cardiovascular diseases. J Transl Med 16, 121 (2018). https://doi.org/10.1186/s12967-018-1500-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-018-1500-0

Keywords