Skip to main content

Hemorrhagic cystitis induced by JC polyomavirus infection following COVID-19: a case report

Abstract

JC polyomavirus (JCPyV) is a human polyomavirus that can establish lifelong persistent infection in the majority of adults. It is typically asymptomatic in immunocompetent individuals. However, there is a risk of developing progressive multifocal leukoencephalopathy (PML) in immunocompromised or immunosuppressed patients. Though JCPyV commonly resides in the kidney-urinary tract, its involvement in urinary system diseases is extremely rare. Here, we reported a case of a 60-year-old male patient with coronavirus disease 2019 (COVID-19) infection who developed hemorrhagic cystitis after receiving treatment with nirmatrelvir 300 mg/ritonavir 100 mg quaque die (QD). Subsequent metagenomic next-generation sequencing (mNGS) confirmed the infection to be caused by JCPyV type 2. Then, human immunoglobulin (PH4) for intravenous injection at a dose of 25 g QD was administered to the patient. Three days later, the hematuria resolved. This case illustrates that in the setting of compromised host immune function, JCPyV is not limited to causing central nervous system diseases but can also exhibit pathogenicity in the urinary system. Moreover, mNGS technology facilitates rapid diagnosis of infectious etiology by clinical practitioners, contributing to precise treatment for patients.

Peer Review reports

Introduction

Coronavirus disease 2019 (COVID-19) is an illness caused by the novel coronavirus SARS-CoV-2 and has resulted in a global pandemic with over 700 million confirmed cases and approximately 7 million deaths [1]. Studies have found that patients with COVID-19 infection exhibit both elevated pro-inflammatory mediators and significant immune suppression, which may be attributed to immune dysregulation in response to the infection [2]. In the state of immunological derangement, polyomavirus can transition from a latent phase to a pathogenic phase. Among the known human polyomaviruses, the most extensively studied are BK polyomavirus (BKPyV) and JC polyomavirus (JCPyV). These are currently the only two viruses associated with extracellular vesicles [3,4,5]. Both BKPyV and JCPyV establish persistent infections in the kidneys, but only BKPyV is typically pathogenic at this site, leading to hemorrhagic cystitis and nephropathy [6]. JCPyV is not only well-known for causing fatal progressive multifocal leukoencephalopathy (PML) but is also associated with other rare neurological disorders such as JC virus granule-cell neuronopathy, JC virus encephalopathy, and JC virus meningitis [6, 7].

JCPyV possesses a closed circular double-stranded DNA genome with a length of 5130 bp, divided into early and late genes, separated by the non-coding control region (NCCR) containing the replication origin (ORI), promoter, and enhancer elements. The genome encodes six major viral proteins (large T and small T antigens, VP1, VP2, VP3, and agnoprotein), as well as several splice variants of the T antigen [8,9,10]. The large T antigen is the most crucial protein in JCPyV, participating in the transcription and replication of the viral genome. T proteins drive host cells into the S phase for viral replication, regulate the transcription of host and viral genomes, directly participate in viral DNA replication, and interact with numerous cellular proteins to facilitate these processes [9, 10]. Subsequently, the JCPyV agnoprotein acts as a viral channel protein, participating in the release of progeny viral particles and promoting JCPyV reproduction [11,12,13].

Research indicates that polyomaviruses may undergo periodic reactivation in both individuals with normal immune function and immunocompromised patients, as evidenced by asymptomatic viral urine shedding. It is noteworthy that compared to BKPyV, which is rarely found in the urine of healthy adults, JCPyV viral shedding is more common and increases with age [14]. A study involving 400 healthy blood donors found that JCPyV viral shedding was significantly more frequent and had higher viral loads compared to BKPyV viral shedding (19% vs. 7%, P < 0.0001) [15]. Research suggests that asymptomatic viral shedding may progress to hematuria and develop into symptoms of cystitis as tissue damage increases, with hemorrhagic cystitis being more common with BKPyV infection. JCPyV is a latent infection in the majority of humans, typically persisting in an asymptomatic state [16]. However, in cases of immune dysregulation, JCPyV can be reactivated, transitioning from latency to a pathogenic phase [17, 18]. This study reports, for the first time, a case of immunocompromised patient, due to COVID-19 infection, in whom JCPyV infection was confirmed to be activated leading to hemorrhagic cystitis through metagenomics next-generation sequencing (mNGS) results.

Case presentation

A 60-year-old male presented to our hospital on May 29, 2023, complaining of fever and sore throat for two days. Upon admission, the nucleic acid test for the novel coronavirus was positive, leading to the consideration of hospitalization in the Infectious Pulmonary Disease Department for the treatment of COVID-19 infection. Twenty-four years prior (Surgery performed in 1999), the patient underwent mitral valve and tricuspid valve replacement, as well as pulmonary valve repair. He had been receiving long-term oral anticoagulation therapy with warfarin 2.5 mg quaque die (QD). Additionally, he had an 8-year history of hypertension (Diagnosed in 2015). On the day of admission, the patient complained of low-grade fever, sore throat, fatigue, and poor appetite, without symptoms such as chest tightness, shortness of breath, abdominal pain, diarrhea, urinary frequency, urgency, dysuria, or hematuria. Physical examination revealed no positive signs. Blood tests upon admission showed a white blood cell count of 12.7 × 109/L and a C-reaction protein (CRP) level of 17.9 mg/L, with other parameters within normal limits. Coagulation function tests showed a prothrombin time (PT) of 21.3 s, an activated partial thromboplastin time (APTT) of 44.5 s, and an international normalized ratio (INR) of 1.82R. Urinalysis, cardiac enzyme panel, renal function, liver function, and other indicators were normal. Chest CT showed chronic inflammation in both lungs. The patient was started on nirmatrelvir 300 mg/ritonavir 100 mg QD and methylprednisolone injection 40 mg QD for anti-COVID-19 treatment while continuing the anticoagulation therapy with warfarin 2.5 mg QD. With the anti-COVID-19 treatment, the patient’s body temperature gradually normalized, and symptoms improved. On the 5th day (June 2, 2023) of hospitalization, nirmatrelvir/ritonavir and methylprednisolone were discontinued.

On the 8th day (June 5, 2023) of hospitalization, the patient suddenly developed hematuria, prompting discontinuation of warfarin. On the 9th day (June 6, 2023) of hospitalization, urinalysis revealed 3 + occult blood, with a red blood cell count of 1022 cells/μL and a white blood cell count of 5 cells/μL. Blood tests showed a white blood cell count of 10.4 × 109/L and a CRP level of 34.57 mg/L, with other parameters within normal limits. Coagulation function tests showed a PT of 17.8 s, an APTT of 43.6 s, and an INR of 1.51R. Renal, ureteral, bladder and prostate ultrasound revealed no abnormalities. A possible urinary tract infection was considered. Therefore, on the 9th day (June 6, 2023) of hospitalization, the patient was started on levofloxacin tablets 0.5 g QD for antimicrobial therapy. However, the treatment response was poor, with the gradually worsened hematuria symptoms and intermittent low-grade fever (around 37.5 ℃). On the 11th day (June 8, 2023) of hospitalization, the urinalysis was counterchecked, showing 3 + occult blood, a red blood cell count of 19,502 cells/μL, and a white blood cell count of 152 cells/μL. Repeated blood routine examination showed a white blood cell count of 10.6 × 109/L and a CRP level of 16.7 mg/L, with other parameters within normal limits. Coagulation function tests demonstrated a PT of 16.4 s, an APTT of 36.4 s, and an INR of 1.39R. Renal function, liver function, and cardiac enzyme panel were normal.

The urine culture results were negative, and we could not identify the cause of hematuria. We recognize that this may be due to limitations in laboratory testing conditions, making it challenging to detect the pathogenic microorganisms responsible. To expedite the diagnosis, on the 12th day (June 9, 2023) of hospitalization, a midstream urine sample from the patient was sent to a third-party clinical laboratory (Zhejiang Luoxi Medical Laboratory Co., Ltd) for mNGS testing. Details of the mNGS method and quality control are provided in Supplementary file 1. The results revealed JCPyV type 2 with a sequence count of 919 and a relative abundance of 96.84%. We considered the patient to have hemorrhagic cystitis caused by JCPyV, unrelated to the treatment of warfarin. Consequently, we reinitiated the patient’s warfarin therapy at a dose of 2.5 mg QD and discontinued levofloxacin. Starting from the 13th day (June 10, 2023) of hospitalization, human immunoglobulin (PH4) for intravenous injection at a dosage of 25 g QD was commenced to treat hemorrhagic cystitis induced by JCPyV infection. After 3 days (June 10–12) of treatment, the patient’s body temperature normalized, and there was no further hematuria, leading to discontinuation of the medication. The patient was discharged on the 15th day (June 12, 2023) of hospitalization and continued long-term anticoagulation therapy with warfarin at a dose of 2.5 mg QD (for anticoagulation following previous cardiac valve replacement surgery). One month after discharge, the patient had a follow-up visit at the outpatient clinic. Urinalysis revealed a presence of 56 fungi/μl. Considering that the patient had a normal body temperature and no signs of urinary tract infection such as hematuria, chills, or febrile episodes, the result was considered a possible contamination of the urine specimen. Therefore, no treatment was administered. The follow-up was concluded (Fig. 1).

Fig. 1
figure 1

Timeline of significant events before and after patient admission. *Day 0: COVID-19 testing was conducted, with a positive result. The patient was admitted for treatment on the same day (Day 1). Therefore, Day 0 and Day 1 refer to the same day

Discussion

JCPyV is a non-enveloped DNA virus with a seroprevalence of 50% to 70% in adults [15, 19, 20]. As a conditional pathogen, JCPyV virus can cause various diseases in the central nervous system, with the most prominent being PML, a demyelinating disease involving lytic infection of glial cells [21,22,23]. The prerequisite for PML is profound suppression of cell-mediated immunity, whether disease-related, such as with HIV or lymphoproliferative malignancies, or due to immunosuppressive or immunomodulatory therapies (multiple sclerosis or rheumatoid arthritis) or a combination of both (systemic lupus erythematosus) [21]. Additionally, JCPyV can infect meningeal and choroid plexus cells, leading to JCPyV meningitis (JCVM) [24]. Studies have found that JCPyV VP1 capsid protein deletion mutants contribute to facilitating JCV entry or replication into granule cell neurons, causing JC virus granule cell neuronopathy (JCV GCN) [25, 26]. The fulminant JCPyV encephalopathy (JCE) can also involve cortical pyramidal neurons, characterized by cortical gray matter infection and dissolution [27]. Furthermore, after the initial infection, JCPyV can remain latent in different sites, such as B cells, the brain, spleen, and notably, the urogenital tract (as the most epidemiologically relevant latent site) [14].

The JCPyV virus undergoes persistent latent infection in the kidneys and is shed into the urine [28]. In healthy individuals, the urinary shedding of JCPyV is usually asymptomatic and intermittent, with a higher prevalence and a higher viral load compared to BKPyV [15]. Conversely, in immunocompromised patients, BKPyV urinary shedding is more common [29]. Despite the high viral load in urine, most patients remain asymptomatic, suggesting effective compensatory mechanisms in the cytopathic loss of urothelial cells. Notably, evident diseases induced by JCPyV are observed only under specific circumstances. For example, approximately 50% of kidney transplant recipients develop Polyomavirus-associated nephropathy (PVAN) caused by BKPyV, ultimately leading to graft failure [30, 31]. BKPyV-induced hemorrhagic cystitis occurs in 57% of bone marrow transplant recipients [32]. Interestingly, we observed that JCPyV-induced kidney disease is rare. JCPyV-related nephropathy is a severe but extremely rare complication in kidney transplant recipients, with an incidence rate of only 0.9% in a cohort of 103 kidney transplant recipients [28, 33]. Despite the close homology between BKPyV and JCPyV, there are significant biological differences in their pathogenesis [34].

Although persistent microscopic hematuria caused by JCPyV infection has been reported by Chiarinotti D et al. [35] and Di Maida F et al. [36], as well as JCPyV-induced PVAN being reported [37, 38], these instances are limited to individual case reports. Unlike the more common urological manifestations caused by BKPyV, cases of hemorrhagic cystitis induced by JCPyV are exceptionally rare. JCPyV, as a conditional pathogenic agent, rarely causes hemorrhagic cystitis. Recent literature suggests that although replication of JCPyV in the urinary tract is common, it is associated with a lower incidence of renal disease, indicating that JCPyV viruria may be a protective factor against kidney diseases [39, 40]. It is believed that the long-term coevolution of human polyomaviruses with the host has led to the loss of their pathogenicity, while unique selective pressures encountered in immunocompromised hosts have driven host intramolecular evolution, resulting in the emergence of pathogenic polyomaviruses [41]. In other words, changes in the host’s immune status can reactivate the virus and cause significant pathological reactions in a few cases. This may partially explain the occurrence of JCPyV-associated hemorrhagic cystitis in the case we reported, who had recently experienced a COVID-19 infection prior to the onset of hematuria. It is well known that COVID-19 infection disrupts the host immune homeostasis and stimulates an excessive inflammatory response [42], with severe cases even experiencing a “cytokine storm”, leading to severe clinical complications [43]. In the elderly population, the severity of COVID-19 is exacerbated due to underlying comorbidities and immune senescence [44]. The case we reported involved a 60-year-old patient with hypertension and a history of heart valve replacement surgery, with an elevated CRP index above the normal range (> 10 mg/L) after admission. CRP is a major acute-phase protein [45], and its levels increase in response to injury, infection, and inflammation [46]. In our report, there was an increase in CRP levels (34.57 mg/L vs 17.9 mg/L) during the occurrence of hematuria, indicating an exacerbation of systemic infection or inflammation. This indirectly suggests that the occurrence of JCPyV-induced hemorrhagic cystitis in this case may be related to the disruption of host immune homeostasis following COVID-19 infection, leading to reactivation of JCPyV. However, we did not perform testing for T lymphocyte subtypes, complement, and other immune response molecules, which limits our comprehensive understanding of the specific immune changes in the patient’s body following COVID-19 infection. In addition, an early study in Wuhan, China, reported that among 701 COVID-19 patients, the incidence of acute kidney injury was only 5.1%, while proteinuria (43.9%) and hematuria (26.7%) were very common [47]. In the pathophysiology of COVID-19 infection, ACE2 receptors facilitate intracellular entry and replication of the SARS-CoV-2 virus [48]. Research also suggests a close association between ACE2 receptors and organ damage related to COVID-19 [49]. This may also be one of the reasons for the hematuria observed in this case.

In 2022, a study titled “Seroprevalence of JCV during the SARS COVID-19 Pandemic” assessed the seroprevalence of JCPyV in the first year following the COVID-19 outbreak and compared it with the seroprevalence of previous years. The results indicated a threefold increase in seroprevalence after the pandemic, although statistically nonsignificant, it was notably elevated [50]. The research also found that JCPyV could induce blood coagulation in human O-type red blood cells, fostering seroepidemiological investigations that have led to the global identification of JCPyV. A substantial proportion of the population experiences seroconversion before adulthood [51, 52], and healthy individuals, including pregnant women, can produce immunoglobulin G (IgG) antibodies against JCPyV [53, 54]. However, current seroepidemiological research predominantly focuses on immunocompromised patients with JCPyV reactivation causing PML [55, 56]. Research on JCPyV-induced urological diseases is relatively limited, and future studies should strengthen serological investigations to delve into the underlying pathogenic mechanisms.

In this report, we employed mNGS as a rapid diagnostic tool for JCPyV infection. Conventional diagnostic methods were unable to identify the pathogen in this patient. Our report effectively demonstrated the potential of mNGS to improve the identification of rare pathogenic agents. Previous methods for viral detection have included antigen–antibody analysis [57] and polymerase chain reaction (PCR)-based techniques [58]. mNGS, a recently popularized high-throughput sequencing approach, has been utilized to assist in pathogen detection from various body fluids, such as cerebrospinal fluid, bronchoalveolar lavage fluid, and plasma, offering unique advantages in comprehensive profiling of known and unknown pathogens [59, 60]. Due to its high degree of specialization and cost, mNGS cannot currently be considered a diagnostic tool integrated into routine clinical practice. However, with technological advancements, it is imperative to reduce costs and standardize procedures. It is believed that once mNGS achieves a comprehensive transition from scientific research to clinical practice, it will significantly transform disease diagnosis and treatment approaches.

Currently, there are no specific antiviral drugs or other options available for JCPyV, apart from immune reconstitution [23]. In this report, effective control of hemorrhagic cystitis was achieved by administering human immunoglobulin (PH4) for intravenous injection to the patient for three days. Human immunoglobulin (PH4) for intravenous injection is a therapeutic preparation of normal human Immunoglobulin G (IgG) obtained from healthy blood donors, which can be used as a replacement therapy or an immunomodulator in patients with primary or secondary immunodeficiencies [61]. Antiviral drugs such as chlorpromazine, cytarabine, and topotecan are also widely used in patients with JCPyV-associated diseases [34]. Furthermore, researchers are developing vaccines targeting JCPyV [19], aiming to prevent the development of JCPyV-associated diseases. Recently, the immune checkpoint inhibitor pembrolizumab has been reported to treat a patient with JCPyV-induced PML, demonstrating the great therapeutic potential of anti-PD-1 therapy in PML patients [62]. With the advancement of technology, there will be more treatment options available for JCPyV infection.

In this case, under the condition of immune dysregulation, JCPyV induces the occurrence of hemorrhagic cystitis, challenging the previous understanding that JCPyV in urine is non-pathogenic [63], thus enhancing our understanding of the outcomes of JCPyV infection. Moreover, mNGS technology has been reported as a novel and effective tool for rapidly diagnosing infectious etiologies, emphasizing its potential contribution to precise treatment decisions by clinical practitioners.

Availability of data and materials

The data and materials in the current study are available from the corresponding author on reasonable request.

References

  1. WHO. WHO Coronavirus (COVID-19) dashboard. 2023.

  2. Limmer A, Engler A, Kattner S, Gregorius J, Pattberg KT, Schulz R, Schwab J, Roth J, Vogl T, Krawczyk A, Witzke O, Zelinskyy G, Dittmer U, Brenner T, Berger MM. Patients with SARS-CoV-2-induced viral sepsis simultaneously show immune activation, impaired immune function and a procoagulatory disease state. Vaccines (Basel). 2023;11(2):435.

    Article  CAS  PubMed  Google Scholar 

  3. Morris-Love J, Gee GV, O’Hara BA, Assetta B, Atkinson AL, Dugan AS, Haley SA, Atwood WJ. JC polyomavirus uses extracellular vesicles to infect target cells. mBio. 2019;10(2):e00379-19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. O’Hara BA, Morris-Love J, Gee GV, Haley SA, Atwood WJ. JC Virus infected choroid plexus epithelial cells produce extracellular vesicles that infect glial cells independently of the virus attachment receptor. PLoS Pathog. 2020;16(3):e1008371.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Handala L, Blanchard E, Raynal PI, Roingeard P, Morel V, Descamps V, Castelain S, Francois C, Duverlie G, Brochot E, Helle F. BK polyomavirus hijacks extracellular vesicles for En Bloc transmission. J Virol. 2020;94(6):e01834-19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Boothpur R, Brennan DC. Human polyoma viruses and disease with emphasis on clinical BK and JC. J Clin Virol. 2010;47(4):306–12.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Tan CS, Koralnik IJ. Progressive multifocal leukoencephalopathy and other disorders caused by JC virus: clinical features and pathogenesis. Lancet Neurol. 2010;9(4):425–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Morris-Love J, Atwood WJ. Complexities of JC polyomavirus receptor-dependent and -independent mechanisms of infection. Viruses. 2022;14(6):1130.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Assetta B, Atwood WJ. The biology of JC polyomavirus. Biol Chem. 2017;398(8):839–55.

    Article  CAS  PubMed  Google Scholar 

  10. Ferenczy MW, Marshall LJ, Nelson CD, Atwood WJ, Nath A, Khalili K, Major EO. Molecular biology, epidemiology, and pathogenesis of progressive multifocal leukoencephalopathy, the JC virus-induced demyelinating disease of the human brain. Clin Microbiol Rev. 2012;25(3):471–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Suzuki T, Orba Y, Okada Y, Sunden Y, Kimura T, Tanaka S, Nagashima K, Hall WW, Sawa H. The human polyoma JC virus agnoprotein acts as a viroporin. PLoS Pathog. 2010;6(3):e1000801.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Khalili K, White MK, Sawa H, Nagashima K, Safak M. The agnoprotein of polyomaviruses: a multifunctional auxiliary protein. J Cell Physiol. 2005;204(1):1–7.

    Article  CAS  PubMed  Google Scholar 

  13. Suzuki T, Okada Y, Semba S, Orba Y, Yamanouchi S, Endo S, Tanaka S, Fujita T, Kuroda S, Nagashima K, Sawa H. Identification of FEZ1 as a protein that interacts with JC virus agnoprotein and microtubules: role of agnoprotein-induced dissociation of FEZ1 from microtubules in viral propagation. J Biol Chem. 2005;280(26):24948–56.

    Article  CAS  PubMed  Google Scholar 

  14. Costa C, Cavallo R. Polyomavirus-associated nephropathy. World J Transplant. 2012;2(6):84–94.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Egli A, Infanti L, Dumoulin A, Buser A, Samaridis J, Stebler C, Gosert R, Hirsch HH. Prevalence of polyomavirus BK and JC infection and replication in 400 healthy blood donors. J Infect Dis. 2009;199(6):837–46.

    Article  CAS  PubMed  Google Scholar 

  16. May D, Bellizzi A, Kassa W, Cipriaso JM, Caocci M, Wollebo HS. IFNα and β mediated JCPyV suppression through C/EBPβ-LIP isoform. Viruses. 2021;13(10):1937.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kartau M, Auvinen E, Verkkoniemi-Ahola A, Mannonen L, Helanterä I, Anttila VJ. JC polyomavirus DNA detection in clinical practice. J Clin Virol. 2022;146:105051.

    Article  CAS  PubMed  Google Scholar 

  18. Cook L, Polyomaviruses. Microbiol Spectr. 2016;4(4). https://journals.asm.org/doi/10.1128/microbiolspec.dmih2-0010-2015

  19. Ray U, Cinque P, Gerevini S, Longo V, Lazzarin A, Schippling S, Martin R, Buck CB, Pastrana DV. JC Polyomavirus mutants escape antibody-mediated neutralization. Sci Transl Med. 2015;7(306):306ra151.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Antonsson A, Green AC, Mallitt KA, O’Rourke PK, Pawlita M, Waterboer T, Neale RE. Prevalence and stability of antibodies to the BK and JC polyomaviruses: a long-term longitudinal study of australians. J Gen Virol. 2010;91(Pt 7):1849–53.

    Article  CAS  PubMed  Google Scholar 

  21. Kartau M, Sipilä JO, Auvinen E, Palomäki M, Verkkoniemi-Ahola A. Progressive multifocal leukoencephalopathy: current insights. Degener Neurol Neuromuscul Dis. 2019;9:109–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Cortese I, Muranski P, Enose-Akahata Y, Ha SK, Smith B, Monaco M, Ryschkewitsch C, Major EO, Ohayon J, Schindler MK, Beck E, Reoma LB, Jacobson S, Reich DS, Nath A. Pembrolizumab treatment for progressive multifocal leukoencephalopathy. N Engl J Med. 2019;380(17):1597–605.

    Article  CAS  PubMed  Google Scholar 

  23. Atkinson AL, Atwood WJ. Fifty years of JC polyomavirus: a brief overview and remaining questions. Viruses. 2020;12(9):969.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Corbridge SM, Rice RC, Bean LA, Wüthrich C, Dang X, Nicholson DA, Koralnik IJ. JC virus infection of meningeal and choroid plexus cells in patients with progressive multifocal leukoencephalopathy. J Neurovirol. 2019;25(4):520–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dang X, Koralnik IJ. A granule cell neuron-associated JC virus variant has a unique deletion in the VP1 gene. J Gen Virol. 2006;87(Pt 9):2533–7.

    Article  CAS  PubMed  Google Scholar 

  26. Koralnik IJ, Wüthrich C, Dang X, Rottnek M, Gurtman A, Simpson D, Morgello S. JC virus granule cell neuronopathy: a novel clinical syndrome distinct from progressive multifocal leukoencephalopathy. Ann Neurol. 2005;57(4):576–80.

    Article  PubMed  Google Scholar 

  27. Wüthrich C, Dang X, Westmoreland S, McKay J, Maheshwari A, Anderson MP, Ropper AH, Viscidi RP, Koralnik IJ. Fulminant JC virus encephalopathy with productive infection of cortical pyramidal neurons. Ann Neurol. 2009;65(6):742–8.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Delbue S, Ferraresso M, Ghio L, Carloni C, Carluccio S, Belingheri M, Edefonti A, Ferrante P. A review on JC virus infection in kidney transplant recipients. Clin Dev Immunol. 2013;2013:926391.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Wiseman AC. Polyomavirus nephropathy: a current perspective and clinical considerations. Am J Kidney Dis. 2009;54(1):131–42.

    Article  PubMed  Google Scholar 

  30. Hirsch HH, Steiger J. Polyomavirus BK. Lancet Infect Dis. 2003;3(10):611–23.

    Article  PubMed  Google Scholar 

  31. Hirsch HH. BK virus: opportunity makes a pathogen. Clin Infect Dis. 2005;41(3):354–60.

    Article  PubMed  Google Scholar 

  32. Arthur RR, Shah KV, Baust SJ, Santos GW, Saral R. Association of BK viruria with hemorrhagic cystitis in recipients of bone marrow transplants. N Engl J Med. 1986;315(4):230–4.

    Article  CAS  PubMed  Google Scholar 

  33. Drachenberg CB, Hirsch HH, Papadimitriou JC, Gosert R, Wali RK, Munivenkatappa R, Nogueira J, Cangro CB, Haririan A, Mendley S, Ramos E. Polyomavirus BK versus JC replication and nephropathy in renal transplant recipients: a prospective evaluation. Transplantation. 2007;84(3):323–30.

    Article  PubMed  Google Scholar 

  34. Diotti RA, Nakanishi A, Clementi N, Mancini N, Criscuolo E, Solforosi L, Clementi M. JC polyomavirus (JCV) and monoclonal antibodies: friends or potential foes? Clin Dev Immunol. 2013;2013:967581.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Chiarinotti D, Ruva CE, David P, Capurro F, Brustia M, Omodeo Zorini E, Benigni E, Turello E, De Leo M, Verzetti G. Polyomavirus infection in an immunocompetent patient and literature overview. G Ital Nefrol. 2002;19(1):74–8.

    CAS  PubMed  Google Scholar 

  36. Di Maida F, Viola L, Lambertini L, Mari A, Mencarini J, Borchi B, Carini M, Zammarchi L, Minervini A. A rare urinary JC virus reactivation after long-term therapy with rituximab. Int J Infect Dis. 2021;103:447–9.

    Article  PubMed  Google Scholar 

  37. Janphram C, Worawichawong S, Disthabanchong S, Sumethkul V, Rotjanapan P. Absence of JC Polyomavirus (JCPyV) viremia in early post-transplant JCPyV nephropathy: a case report. Transpl Infect Dis. 2017;19(6):e12761. https://onlinelibrary.wiley.com/doi/10.1111/tid.12761.

  38. Sharma N, Abdulkhalek S. Kidney allograft dysfunction due to John Cunningham (JC) virus nephropathy. Cureus. 2022;14(11):e32021.

    PubMed  PubMed Central  Google Scholar 

  39. Divers J, Langefeld CD, Lyles DS, Ma L, Freedman BI. Protective association between JC polyoma viruria and kidney disease. Curr Opin Nephrol Hypertens. 2019;28(1):65–9.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Kruzel-Davila E, Divers J, Russell GB, Kra-Oz Z, Cohen MS, Langefeld CD, Ma L, Lyles DS, Hicks PJ, Skorecki KL, Freedman BI, Family Investigation of N.; diabetes. Viruria is associated with reduced risk of diabetic kidney disease. J Clin Endocrinol Metab. 2019;104(6):2286–94.

    Article  PubMed  PubMed Central  Google Scholar 

  41. McIlroy D, Halary F, Bressollette-Bodin C. Intra-patient viral evolution in polyomavirus-related diseases. Philos Trans R Soc Lond B Biol Sci. 2019;374(1773):20180301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Meng H, Wang S, Tang X, Guo J, Xu X, Wang D, Jin F, Zheng M, Yin S, He C, Han Y, Chen J, Han J, Ren C, Gao Y, Liu H, Wang Y, Jin R. Respiratory immune status and microbiome in recovered COVID-19 patients revealed by metatranscriptomic analyses. Front Cell Infect Microbiol. 2022;12:1011672.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Montazersaheb S, Hosseiniyan Khatibi SM, Hejazi MS, Tarhriz V, Farjami A, Ghasemian Sorbeni F, Farahzadi R, Ghasemnejad T. COVID-19 infection: an overview on cytokine storm and related interventions. Virol J. 2022;19(1):92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Abul Y, Leeder C, Gravenstein S. Epidemiology and clinical presentation of COVID-19 in older adults. Infect Dis Clin North Am. 2023;37(1):1–26.

    Article  PubMed  Google Scholar 

  45. Black S, Kushner I, Samols D. C-reactive protein. J Biol Chem. 2004;279(47):48487–90.

    Article  CAS  PubMed  Google Scholar 

  46. Sproston NR, Ashworth JJ. Role of C-Reactive protein at sites of inflammation and infection. Front Immunol. 2018;9:754.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Cheng Y, Luo R, Wang K, Zhang M, Wang Z, Dong L, Li J, Yao Y, Ge S, Xu G. Kidney disease is associated with in-hospital death of patients with COVID-19. Kidney Int. 2020;97(5):829–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sarier M, Demir M, Emek M, Usta SS, Soylu A, Konuk EY, Turgut H. Comparison of spermiograms of infertile men before and during the COVID-19 pandemic. Rev Assoc Med Bras (1992). 2022;68(2):191–5.

    Article  PubMed  Google Scholar 

  49. Lores E, Wysocki J, Batlle D. ACE2, the kidney and the emergence of COVID-19 two decades after ACE2 discovery. Clin Sci (Lond). 2020;134(21):2791–805.

    Article  CAS  PubMed  Google Scholar 

  50. Vigiser I, Piura Y, Kolb H, Shiner T, Komarov I, Karni A, Regev K. JCV seroconversion rate during the SARS COVID-19 pandemic. Multiple Scler Relat Disorders. 2022;68:104244.

    Article  CAS  Google Scholar 

  51. Rziha HJ, Bornkamm GW, zur Hausen H. BK virus: I. Seroepidemiologic studies and serologic response to viral infection. Med Microbiol Immunol. 1978;165(2):73–81.

    Article  CAS  PubMed  Google Scholar 

  52. Walker DL, Padgett BL. The epidemiology of human polyomaviruses. Prog Clin Biol Res. 1983;105:99–106.

    CAS  PubMed  Google Scholar 

  53. Andrews CA, Daniel RW, Shah KV. Serologic studies of papovavirus infections in pregnant women and renal transplant recipients. Prog Clin Biol Res. 1983;105:133–41.

    CAS  PubMed  Google Scholar 

  54. Daniel R, Shah K, Madden D, Stagno S. Serological investigation of the possibility of congenital transmission of papovavirus JC. Infect Immun. 1981;33(1):319–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. De Gascun CF, Carr MJ. Human polyomavirus reactivation: disease pathogenesis and treatment approaches. Clin Dev Immunol. 2013;2013:373579.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Elphick GF, Querbes W, Jordan JA, Gee GV, Eash S, Manley K, Dugan A, Stanifer M, Bhatnagar A, Kroeze WK, Roth BL, Atwood WJ. The human polyomavirus, JCV, uses serotonin receptors to infect cells. Sci (New York N Y). 2004;306(5700):1380–3.

    Article  CAS  Google Scholar 

  57. Amanat F, Stadlbauer D, Strohmeier S, Nguyen THO, Chromikova V, McMahon M, Jiang K, Arunkumar GA, Jurczyszak D, Polanco J, Bermudez-Gonzalez M, Kleiner G, Aydillo T, Miorin L, Fierer DS, Lugo LA, Kojic EM, Stoever J, Liu STH, Cunningham-Rundles C, Felgner PL, Moran T, Garcia-Sastre A, Caplivski D, Cheng AC, Kedzierska K, Vapalahti O, Hepojoki JM, Simon V, Krammer F. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat Med. 2020;26(7):1033–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Smyrlaki I, Ekman M, Lentini A, Rufino de Sousa N, Papanicolaou N, Vondracek M, Aarum J, Safari H, Muradrasoli S, Rothfuchs AG, Albert J, Hogberg B, Reinius B. Massive and rapid COVID-19 testing is feasible by extraction-free SARS-CoV-2 RT-PCR. Nat Commun. 2020;11(1):4812.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Duan H, Li X, Mei A, Li P, Liu Y, Li X, Li W, Wang C, Xie S. The diagnostic value of metagenomic next rectanglegeneration sequencing in infectious diseases. BMC Infect Dis. 2021;21(1):62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wu X, Li Y, Zhang M, Li M, Zhang R, Lu X, Gao W, Li Q, Xia Y, Pan P, Li Q. Etiology of severe community-acquired pneumonia in adults based on metagenomic next-generation sequencing: a prospective multicenter study. Infect Dis Ther. 2020;9(4):1003–15.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Mouthon L. Intravenous immunoglobulin therapy. Rev Prat. 2005;55(10):1049–56.

    PubMed  Google Scholar 

  62. Mohn N, Wattjes MP, Adams O, Nay S, Tkachenko D, Salge F, Heine J, Pars K, Hoglinger G, Respondek G, Stangel M, Skripuletz T, Jacobs R, Suhs KW. PD-1-inhibitor pembrolizumab for treatment of progressive multifocal leukoencephalopathy. Ther Adv Neurol Disord. 2021;14:1756286421993684.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Yogo Y, Kitamura T, Sugimoto C, Ueki T, Aso Y, Hara K, Taguchi F. Isolation of a possible archetypal JC virus DNA sequence from nonimmunocompromised individuals. J Virol. 1990;64(6):3139–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This research received no specific grant from any funding agency in the public, commercial, or not for profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

YJ L conceived of the study, and participated in its design and interpretation and helped to draft the manuscript. XP L participated in the design and interpretation of the data and drafting/revising the manuscript. YJ L and XP L performed the statistical analysis and revised the manuscript critically. All the authors read and approved the final manuscript.

Corresponding author

Correspondence to Yuanjie Lv.

Ethics declarations

Ethics approval and consent to participate

This study was reviewed and approved by Hospital of Traditional Chinese Medicine, Xinchang County Ethics Committee (No.2023-LW-01).

Consent for publication

We have obtained the patient’s written informed consent to publish the case report.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lv, Y., Liu, X. Hemorrhagic cystitis induced by JC polyomavirus infection following COVID-19: a case report. BMC Urol 24, 87 (2024). https://doi.org/10.1186/s12894-024-01464-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12894-024-01464-1

Keywords