2,376
Views
0
CrossRef citations to date
0
Altmetric
Commentary

Parallels in the Pathogenesis of SARS-CoV-2 and M. Tuberculosis: a synergistic or Antagonistic Alliance?

, ORCID Icon & ORCID Icon
Pages 1691-1695 | Received 13 Jul 2020, Accepted 08 Dec 2020, Published online: 06 Jan 2021

“SARS-CoV-2 and M. tuberculosis infections have some important parallels with regards to cell entry, immune response and immune evasion. Thus, TB-endemic regions may have exacerbated pathologies in the context of SARS-CoV-2 infections; something that the scientific community will need to pay attention to.”

The world is facing a major challenge of the new pneumonia condition termed COVID-19 caused by SARS-CoV-2, the seventh member of human coronaviruses. The global burden of COVID-19 is rising daily and as of 10 November 2020, there were over 1,275,122 deaths (https://www.worldometers.info/coronavirus/).

Coronaviruses are enveloped, positive-sense and single-stranded RNA viruses [Citation1]. Based on 96.2% nucleotide sequence identity with a bat-borne coronavirus (BatCoV RaTG13) that has been identified in Rhinolophus affinis bat species, it is likely that SARS-CoV-2 originated from a bat [Citation2]. COVID-19 is primarily defined by an acute viral pneumonia and cytokine storm leading to respiratory failure [Citation3]. The main transmission route of this virus is droplets blown out through cough and sneezing by an infected person. The common symptoms of COVID-19 include cough, fever, shortness of breath and tiredness [Citation4]. Severe cases manifest in symptoms that are associated with cellular immune deficiency, coagulation activation, myocardia, multiple organ dysfunction and septic shock [Citation5]. SARS-CoV-2 is highly pathogenic in persons with underlying medical conditions that reduce their immune competence such as TB [Citation6].

TB, caused by Mycobacterium tuberculosis and transmitted through infected air droplets from cough or sneezing, is one of the top ten causes of death due to infectious diseases globally, with an estimated 10 million infections and 1.5 million deaths in 2018 [Citation7]. Indeed, since TB affects the respiratory system, it could prove catastrophic if present in comorbidity with COVID-19 [Citation6].

In this commentary, we discuss the current state of knowledge on the parallels in the pathogenesis of SARS-CoV-2 and M. tuberculosis and the potential implications of co-infection on the clinical outcomes.

SARS-CoV-2 & M. tuberculosis: targeting same cells?

Spike (S) glycoprotein on the surface of SARS-CoV-2 facilitates viral entry into target cells. The S protein consists of three receptor-binding S1 heads which bind to the cellular receptor to facilitate viral attachment to target cells [Citation8]. Moreover, the S protein is required to be proteolytically active by being cleaved at the multibasic S1/S2 cleavage site by the host’s cell protease furin to facilitate protein-mediated cell-cell fusion [Citation9]. The receptor binding domain of the S protein binds to angiotensin-converting enzyme 2 (ACE2) receptor and invade the host cell through clathrin-mediated endocytosis [Citation4]. Type I interferons (IFN) stimulate the expression of SARS-CoV-2 entry receptors and enable the virus to be endocytosed [Citation4]. The high human ACE2 binding affinity of the receptor binding domain and furin activation of the spike helps SARS-CoV-2 to successfully enter the human lung cell while evading the immune response [Citation10]. The virus infects and replicates in ciliated and mucus-secreting cells of bronchial epithelium, type II pneumocytes in the lungs [Citation11] and macrophages [Citation12].

Like SARS-CoV-2, M. tuberculosis also infects type II pneumocytes in the lungs [Citation13,Citation14] via pattern recognition receptors of the host cells, including Toll-like receptors, complement receptors, dendritic cell-specific intercellular adhesion molecule grabbing nonintegrin (DC-SIGN), mannose receptors, CD14 receptors, scavenger receptors and FCγ receptors [Citation15]. Furthermore, the bacterium is internalized through phagocytosis by the resident macrophages and further translocated to lysosomes for destruction [Citation16].

Co-infection of SARS-CoV-2 and M. tuberculosis may exacerbate the pathologies that are associated with each pathogen. The co-infection in macrophages can increase the production of pro- and anti-inflammatory cytokines and therefore play a crucial role in pathogenesis. Severe infection of type II pneumocytes with SARS-CoV-2 can cause regeneration failure of the cells and lead to lesions in the lungs, accelerated bronchial damage and ultimately lung disease [Citation17].

Dysregulating the immune system: the shared story of SARS-CoV-2 & M. tuberculosis

The human immune system responds to SARS-CoV-2 by differentiating the monocytes into pro-inflammatory macrophages through activation of JAK–STAT pathways, an activation that is mediated by production of IL-1β and/ or IL-18 [Citation18]. The activation can lead to the production of pro-inflammatory cytokines and resulting in COVID-19-associated cytokine storm. Cytokine storm causes rapid replication of SARS-CoV-2 virus and results in the infection of more alveolar epithelial cells [Citation19]. Significant elevation of inflammatory cytokines such as IL-6, IL-1β and IP10 have been detected in COVID-19 patients [Citation20]. The rapidly increasing cytokines induce excess production of neutrophils, which infiltrate lung tissue and cause lung injury [Citation21]. Cytokine storm is also induced in M. tuberculosis infection and is driven by an overwhelming immune response from alveolar macrophages, monocytes and dendritic cells in the lungs and lymph nodes [Citation22]. These cells release an array of cytokines, including IL-1, IL-18, IFN-α, IL-6, IL-10 [Citation23] and therefore co-infections of SARS-CoV-2 with M. tuberculosis will likely result in the production of high amounts of cytokines and lead to exacerbated pathogenicity.

Although the two pathogens share some features of cytokine storm, it is important to note that the magnitude of response differs, with M. tuberculosis having a lower response. Moreover, the outcomes associated with cytokine storm are different for the two pathogens. The COVID-19 patients experience acute respiratory distress syndrome whereas in M. tuberculosis chronic wasting and slow long destruction ensue from cytokine storm.

Another marked feature in the immune response against SARS-CoV-2 is lymphocytopenia [Citation24]. Lymphocytes count of less than 1.5 × 109 per liter has been shown to increase the risk of severe COVID-19 [Citation25]. It is believed that the virus directly infects lymphocytes through the ACE2 receptor and kill them leading to lymphocytopenia [Citation26]. The direct damage of organs such as the lymphatic system by SARS-CoV-2 may also result in a reduction of lymphocytes. Production of TNFα, IL-6 and other pro-inflammatory cytokines during SARS-CoV-2 can also induce lymphocyte apoptosis [Citation26].

The dysregulation of T lymphocytes has also been demonstrated in TB patients [Citation27]. It has been reported that protective immunity to TB depends on CD4+ cells through cell-mediated responses to allow control of M. tuberculosis infection [Citation28]. A depletion of the CD4+ cells increases risks to reactivation of TB [Citation29], and is also associated with severe radiological lesions, increased paradoxical reaction or death [Citation27,Citation30]. The depletion of CD4+ by TB may also result in immunosuppression, which may increase severity on patients with comorbidity of TB and SARS-CoV-2 [Citation31].

Potential protective effects of BCG vaccine against COVID-19

BCG is a live attenuated vaccine that has been used to protect children against TB and other viral infections [Citation32]. The vaccine induces immune response through production of antibodies that prevent the proliferation of the pathogen [Citation33]. It is suggested that BCG vaccine might boost the immune system and help reduce the severity of COVID-19 [Citation34]. BCG vaccination has a nonspecific or heterologous effect, which reduces morbidity and mortality to subsequent infections [Citation35]. The vaccine activates CD4+ and CD8+ memory cells with nontargeted antigens, thus modulating T helper (Th) 1 and Th17 responses for secondary infections [Citation36]. BCG also induces immunological memory in innate immune cells including natural killer (NK) cells, monocytes and macrophages [Citation37]. All these result in bystander response including enhanced production of pro-inflammatory cytokines such as IL-1β, IFN-γ, TNF and IL-6, macrophage activity, T-cell responses and enhanced antibody titers [Citation35].

Based on the nonspecific nature of BCG and its capacity to elicit both innate and adaptive immunity, BCG vaccine has been suggested as a preventive measure against SARS-CoV-2 infection or to reduce COVID-19 severity, especially among children who have already been vaccinated with BCG [Citation38,Citation39]. Healthy individuals vaccinated with BCG will have a boosted immune system, which upon infection with SARS-CoV-2, will lead to inhibited viral replication, decreased viral loads and consequently less inflammation and reduced possibility of cytokine storm production and lymphopenia [Citation38]. Countries in Africa, Asia and South America, that are implementing mandatory national BCG vaccination for their populations, have demonstrated lower COVID-19 cases and mortality [Citation39,Citation40]. Although, a direct link between the low COVID-19 cases and BCG is yet to be established, it is probable that BCG vaccination induces protection against SARS-CoV-2 via trained innate immunity. BCG vaccination results in epigenetically trained populations of monocytes and/or NK cells, which robustly responds to and clears a broad spectrum of viruses, including SARS-CoV-2 [Citation41]. More scientific evidence is needed to establish the protective nature of BCG to SARS-CoV-2 specifically, and to which group of individuals – children and/or adults – should be targeted to realize the full benefits of this trained immunity. Moreover, considering that heightened immune response such as cytokine response is attributed to complications in COVID-19 patients [Citation42], the potential negative effects of elevated immune response by the BCG’s trained immunity warrants further investigation.

Conclusion & future perspective

SARS-CoV-2 and M. tuberculosis infections have some important parallels with regards to cell entry, immune response and immune evasion. Thus, TB-endemic regions may have exacerbated pathologies in the context of SARS-CoV-2 infections; something that the scientific community will need to pay attention to. At the moment, recommending BCG vaccine for combating COVID-19 must be done with caution. Moreover, the vaccine does not offer a lifetime protection and there is paucity of evidence confirming its protective role against SARS-CoV-2. More research should be directed toward a deeper understanding of how BCG might protect against COVID-19. A thorough understanding of the pathophysiology of COVID-19 in the context of M. tuberculosis co-infection is warranted to inform on better ways to manage such cases and avert high COVID-19-related mortalities that are likely to occur in the high TB endemic areas.

Financial & competing interests disclosure

K Tapela and CO Olwal were supported by a WACCBIP-World Bank ACE PhD fellowship (ACE02-WACCBIP: Awandare) and a DELTAS Africa grant (DEL-15-007: Awandare). The DELTAS Africa Initiative is an independent funding scheme of the African Academy of Sciences (AAS)’s Alliance for Accelerating Excellence in Science in Africa (AESA) and supported by the New Partnership for Africa’s Development, Planning and Coordinating Agency (NEPARD Agency) with funding from the Welcome Trust (107755/Z15/Z: Awandare). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

Additional information

Funding

K Tapela and CO Olwal were supported by a WACCBIP-World Bank ACE PhD fellowship (ACE02-WACCBIP: Awandare) and a DELTAS Africa grant (DEL-15-007: Awandare). The DELTAS Africa Initiative is an independent funding scheme of the African Academy of Sciences (AAS)’s Alliance for Accelerating Excellence in Science in Africa (AESA) and supported by the New Partnership for Africa’s Development, Planning and Coordinating Agency (NEPARD Agency) with funding from the Welcome Trust (107755/Z15/Z: Awandare). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

References

  • Yan R , ZhangY , LiY , XiaL , GuoY , ZhouQ. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science367, 1444–1448 (2020).
  • Zheng J . SARS-CoV-2: an emerging coronavirus that causes a global threat. Int. J. Biol. Sci.16(10), 1678–1685 (2020).
  • Hung IF , LungK , TsoEYet al. Articles Triple combination of interferon beta-1b, lopinavir – ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, Phase II trial. Lancet6736(20), 1–10 (2020).
  • Yang P , WangX. COVID-19: a new challenge for human beings. Cell. Mol. Immunol.17(5), 555–557 (2020).
  • Wang Y , WangY , ChenY , QinQ. Unique epidemiological and clinical features of the emerging 2019 novel coronavirus pneumonia (COVID-19) implicate special control measures. J. Med. Virol.92(6), 568–576 (2020).
  • Demes L . Doctor’ s note: coronavirus and patients with TB and HIV poor socioeconomic conditions (2020). http://www.aljazeera.com/features/2020/4/8/doctors-note-coronavirus-and-patients-with-tb-and-hiv
  • WHO . Global Tuberculosis Report 2019 New data on tuberculosis trends in 202 countries. http://www.who.int/tb/global-report-2019
  • Hoffmann M , Kleine-WeberH , SchroederS , MullerMA , DrostenC , PohlmannS. SARS-CoV-2 Cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell181, 271–280 (2020).
  • Walls AC , ParkYJ , TortoriciMA , WallA , McGuireAT , VeeslerD. Structure, function and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell181(2), 281–292.e6 (2020).
  • Shang J , WanY , LuoCet al. Cell entry mechanisms of SARS-CoV-2. Proc. Natl Acad. Sci. USA117(21), 11727–11734 (2020).
  • Mason RJ . Pathogenesis of COVID-19 from a cell biology perspective. Eur. Respir. J.55(2000607), 9–11 (2020).
  • Abassi Z , KnaneyY , KarramT , HeymanSN. The lung macrophage in SARS-CoV-2 infection: a friend or a foe?Front. Immunol.11(June), 1–5 (2020).
  • Bowdish DME , SakamotoK , KimMJet al. MARCO, TLR2 and CD14 are required for macrophage cytokine responses to mycobacterial trehalose dimycolate and Mycobacterium tuberculosis. PLoS Pathog.5(6), 1–14 (2009).
  • Ganbat D , SeehaseS , RichterEet al. Mycobacteria infect different cell types in the human lung and cause species dependent cellular changes in infected cells. BMC Pulm. Med.16(1), 1–16 (2016).
  • Hossain MM , NorazmiMN. Pattern recognition receptors and cytokines in Mycobacterium tuberculosis infection - the double-edged sword?Biomed. Res. Int.2013, 1–18 (2013).
  • Russell DG . Mycobacterium tuberculosis: here today, and here tomorrow. Nat. Rev. Mol. Cell. Biol.2, 1–9 (2001).
  • Pinky L , González-parraG , DobrovolnyHM. Superinfection and cell regeneration can lead to chronic viral coinfections Lubna. J. Theor. Biol.466, 24–38 (2019).
  • Merad M , MartinJC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat. Rev. Immunol.20, 355–362 (2020).
  • Coperchini F , ChiovatoL , CroceL , MagriF , RotondiM. The cytokine storm in COVID-19: an overview of the involvement of the chemokine/chemokine-receptor system. Cytokine Growth Factor Rev.53, 25–32 (2020).
  • Liu B , LiM , ZhouZ , GuanX , XiangY. Can we use interleukin-6 (IL-6) blockade for coronavirus disease 2019 (COVID-19)-induced cytokine release syndrome (CRS)?J. Autoimmun.102452, 0896–8411 (2020).
  • Xie P , MaW , TangH , LiuD. Severe COVID-19: a review of recent progress with a look toward the future. Front. Public Health8(189), 1–7 (2020).
  • Etna MP , GiacominiE , SeveraM , CocciaEM. Pro-and anti-inflammatory cytokines in tuberculosis: a two-edged sword in TB pathogenesis. Semin. Immunol.26(6), 543–551 (2014).
  • Flesch IEA , KaufmannSHE. Role of cytokines in tuberculosis. Immunobiology189(3–4), 316–339 (1993).
  • Zheng M , GaoY , WangGet al. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Int. J. Infect. Dis.17(5), 533–535 (2020).
  • Zhao Q , MengM , KumarRet al. Lymphopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: a systemic review and meta-analysis. Int. J. Infect. Dis.96, 131–135 (2020).
  • Tan L , WangQ , ZhangDet al. Lymphopenia predicts disease severity of COVID-19: a descriptive and predictive study. Signal Transduct. Target Ther.5(1), 16–18 (2020).
  • Al-aska AI , Al-anaziAR , Al-subaeiSSet al. CD4+ T-lymphopenIa in HIV negative tuberculosis patients at King Khalid University hospital in Riyadh, Saudi Arabia. Eur. J. Med. Res.16(6), 285–288 (2011).
  • Rahman S , GudettaB , FinkJet al. Compartmentalization of immune responses in human tuberculosis: Few CD8+ effector T cells but elevated levels of FoxP3+ regulatory T cells in the granulomatous lesions. Am. J. Pathol.174(6), 2211–2224 (2009).
  • Fehri SM , RacilH , HabibechSet al. Lymphocytopenia in pulmonary tuberculosis: does it have a clinical value? Eur. Respir. J. 48, PA2667 (2016).
  • Etna MP , GiacominiE , SeveraM , CocciaEM. Pro-and anti-inflammatory cytokines in tuberculosis: a two-edged sword in TB pathogenesis. Semin. Immunol.26(6), 543–551 (2014).
  • Vizcarra P , Pérez-ElíasMJ , QueredaCet al. Description of COVID-19 in HIV-infected individuals: a single-centre, prospective cohort. Lancet HIV3018(20), 1–11 (2020).
  • Salem A , NofalA , HosnyD. Treatment of common and plane warts in children with topical viable Bacillus Calmette–Guerin. Pediatr. Dermatol.30(1), 60–63 (2013).
  • Roland M . The immunopathology of tuberculosis, the mode of action of the Bacillus Calmette–Guérin, of the tuberculin and of the immunotherapy. BBRJ3, 67–76 (2019).
  • WHO . Bacille Calmette-Guérin (BCG) vaccination and COVID-19 (2020). http://www.who.int/news-room/commentaries/detail/bacille-calmette-guérin-(bcg)-vaccination-and-covid-19
  • Moorlag SJCFM , ArtsRJW , van CrevelR , NeteaMG. Nonspecific effects of BCG vaccine on viral infections. Clin. Microbiol. Infect.25(12), 1473–1478 (2019).
  • Kleinnijenhuis J , QuintinJ , PreijersFet al. Long-lasting effects of BCG vaccination on both heterologous th1/th17 responses and innate trained immunity. J. Innate. Immun.6(2), 152–158 (2014).
  • Netea MG , JoostenLAB , LatzEet al. Trained immunity: a program of innate immune memory in health and disease. Science352(6284), 427 (2016).
  • O'Neill LAJ , NeteaMG. BCG-induced trained immunity: can it offer protection against COVID-19?Nat. Rev. Immunol.20(6), 335–337 (2020).
  • Urashima M , OtaniK , HasegawaY , AkutsuT. BCG vaccination and mortality of COVID-19 across 173 countries: an ecological study. Int. J. Environ. Res. Public Health17(15), 1–20 (2020).
  • Wickramasinghe D , WickramasingheN , KamburugamuwaSA , ArambepolaC , SamarasekeraDN. Correlation between immunity from BCG and the morbidity and mortality of COVID-19. Trop. Dis. Travel Med. Vac.6(17), 1–15 (2020).
  • O'Neill LAJ , NeteaMG. BCG-induced trained immunity: can it offer protection against COVID-19?Nat. Rev. Immunol.20(6), 335–337 (2020).
  • Mehta P , McAuleyDF , BrownM , SanchezE , TattersallRS , MansonJJ. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet395(10229), 1033–1034 (2020).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.