Advances in the Diagnosis and Treatment of Rotavirus Infections: Narrative Review
Abstract
1. Introduction
2. Diagnostic Methods for RV Infections
2.1. Clinical Diagnosis
2.2. Laboratory Diagnosis
2.3. Radiological Diagnosis
3. Innovative Diagnosis Methods for RV Infections
3.1. Enzyme Immunoassay (EIA)
3.2. Digital PCR (dPCR)
3.3. Sequencing Methods
4. Symptomatic Treatment and Antiviral Therapy
4.1. Hydration and Dietary Management Based on ESPGHAN/ESPID Recommendations
4.2. Pharmacological Therapy—ESPGHAN/ESPID Recommendations
4.2.1. Zinc
4.2.2. Probiotics Supplementation
4.2.3. Oral Immunoglobulins
4.2.4. Natural Compounds—Quercetin
4.2.5. Antiviral Compounds
5. Materials and Methods
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Crawford, S.E.; Ramani, S.; Tate, J.E.; Parashar, U.D.; Svensson, L.; Hagbom, M.; Franco, M.A.; Greenberg, H.B.; O’Ryan, M.; Kang, G.; et al. Rotavirus infection. Nat. Rev. Dis. Primers 2017, 3, 17083. [Google Scholar] [CrossRef]
- Maldonado, Y.A.; Yolken, R.H. Rotavirus. Baillieres Clin. Gastroenterol. 1990, 4, 609–625. [Google Scholar] [CrossRef]
- Parashar, U.D.; Nelson, E.A.; Kang, G. Diagnosis, management, and prevention of rotavirus gastroenteritis in children. BMJ 2013, 347, f7204. [Google Scholar] [CrossRef]
- Kim, J.S.; Lee, S.K.; Ko, D.H.; Hyun, J.; Kim, H.S. Performance evaluation of the automated fluorescent immunoassay system rotavirus assay in clinical samples. Ann. Lab. Med. 2019, 39, 50–57. [Google Scholar] [CrossRef]
- ICTV. Current ICTV Taxonomy Release: Rotavirus. Available online: https://ictv.global/taxonomy (accessed on 11 September 2025).
- Staat, M.A.; Azimi, P.H.; Berke, T.; Roberts, N.; Bernstein, D.I.; Ward, R.L.; Pickering, L.K.; Matson, D.O. Clinical presentations of rotavirus infection among hospitalized children. Pediatr. Infect. Dis. J. 2002, 21, 221–227. [Google Scholar] [CrossRef] [PubMed]
- Le, X.; Tao, Y.; Wang, B.; Hou, Y.; Ning, Y.; Hou, J.; Wang, R.; Li, Q.; Xia, X. Diversity and potential cross-species transmission of rotavirus A in wild animals in Yunnan, China. Microorganisms 2025, 13, 145. [Google Scholar] [CrossRef]
- Pawłuszkiewicz, K.; Ryglowski, P.J.; Idzik, N.; Błaszczyszyn, K.; Kucharczyk, E.; Gaweł-Dąbrowska, D.; Siczek, M.; Widelski, J.; Paluch, E. Rotavirus infections: Pathophysiology, symptoms, and vaccination. Pathogens 2025, 14, 480. [Google Scholar] [CrossRef]
- Lee, K.Y. Rotavirus infection–associated central nervous system complications: Clinicoradiological features and potential mechanisms. Clin. Exp. Pediatr. 2022, 65, 484–491. [Google Scholar]
- Medycyna Praktyczna. Diarrhea. Available online: https://www.mp.pl/interna/chapter/B16.I.1.2 (accessed on 18 June 2025).
- LeClair, C.E.; McConnell, K.A. Rotavirus. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023; Volume 1. [Google Scholar]
- Xu, X.; Luo, Y.; He, C.; Dian, Z.; Mi, H.; Yang, J.; Feng, Y.; Miao, Z.; Xia, X. Increased Risk of Neurological Disease Following Pediatric Rotavirus Infection: A Two-Center Case-Control Study. J. Infect. Dis. 2023, 227, 1313–1321. [Google Scholar] [CrossRef] [PubMed]
- Ramig, R.F. Pathogenesis of intestinal and systemic rotavirus infection. J. Virol. 2004, 78, 10213–10220. [Google Scholar] [CrossRef] [PubMed]
- Aghsaeifard, Z.; Heidari, G.; Alizadeh, R. Understanding the use of oral rehydration therapy: A narrative review from clinical practice to main recommendations. Health Sci. Rep. 2022, 5, e827. [Google Scholar] [CrossRef]
- Barr, W.; Smith, A. Acute diarrhea. Am. Fam. Physician 2014, 89, 180–189. [Google Scholar]
- Hahn, S.; Kim, Y.; Garner, P. Reduced osmolarity oral rehydration solution for treating dehydration due to diarrhoea in children: Systematic review. BMJ 2001, 323, 81–85. [Google Scholar] [CrossRef]
- Ruuska, T.; Vesikari, T. RV disease in Finnish children: Use of numerical scores for clinical severity of diarrhoeal episodes. Scand. J. Infect. Dis. 1990, 22, 259–267. [Google Scholar] [CrossRef]
- Clark, H.F.; Borian, F.E.; Bell, L.M.; Modesto, K.; Gouvea, V.; Plotkin, S.A. Protective effect of WC3 vaccine against RV diarrhea in infants during a predominantly serotype 1 RV season. J. Infect. Dis. 1988, 158, 570–587. [Google Scholar] [CrossRef]
- Friedman, J.N.; Goldman, R.D.; Srivastava, R.; Parkin, P.C. Development of a clinical dehydration scale for use in children between 1 and 36 months of age. J. Pediatr. 2004, 145, 201–207. [Google Scholar] [CrossRef]
- Levine, A.C.; Glavis-Bloom, J.; Modi, P.; Nasrin, S.; Rege, S.; Chu, C.; Schmid, C.H.; Alam, N.H. Empirically derived dehydration scoring and decision tree models for children with diarrhea: Assessment and internal validation in a prospective cohort study in Dhaka, Bangladesh. Glob. Health Sci. Pract. 2015, 3, 405–418. [Google Scholar] [CrossRef] [PubMed]
- St Jean, D.T.; Chilyabanyama, O.N.; Bosomprah, S.; Asombang, M.; Velu, R.M.; Chibuye, M.; Mureithi, F.; Sukwa, N.; Chirwa, M.; Mokha, P.; et al. Development of a diarrhoea severity scoring scale in a passive health facility-based surveillance system. PLoS ONE 2022, 17, e0272981. [Google Scholar] [CrossRef] [PubMed]
- Gorelick, M.; Shaw, K.; Murphy, K. Validity and reliability of clinical signs in the diagnosis of dehydration in children. Pediatrics 1997, 99, e6. [Google Scholar] [CrossRef]
- World Health Organization (WHO). The Treatment of Diarrhoea: A Manual for Physicians and Other Senior Health Workers, 4th ed.; World Health Organization: Geneva, Switzerland, 2005.
- Aslan, A.; Kurugol, Z.; Cetin, H.; Karakaşlilar, S.; Koturoğlu, G. Comparison of Vesikari and Clark scales regarding the definition of severe rotavirus gastroenteritis in children. Infect. Dis. 2015, 47, 332–337. [Google Scholar] [CrossRef] [PubMed]
- Kang, G. Rotavirus genotypes and severity of diarrheal disease. Clin. Infect. Dis. 2006, 43, 315–316. [Google Scholar] [CrossRef][Green Version]
- Jauregui, J.; Nelson, D.; Choo, E.; Stearns, B.; Levine, A.C.; Liebmann, O.; Shah, S.P.; Carlo, W.A. External validation and comparison of three pediatric clinical dehydration scales. PLoS ONE 2014, 9, e95739. [Google Scholar] [CrossRef]
- Falszewska, A.; Szajewska, H.; Dziechciarz, P. Diagnostic accuracy of three clinical dehydration scales: A systematic review. Arch. Dis. Child. 2018, 103, 383–388. [Google Scholar] [CrossRef]
- Isanaka, S.; Guindo, O.; Langendorf, C.; Seck, A.M.; Plikaytis, B.D.; Sayinzoga-Makombe, N.; McNeal, M.M.; Meyer, N.; Adehossi, E.; Djibo, A.; et al. Efficacy of a Low-Cost, Heat-Stable Oral Rotavirus Vaccine in Niger. N. Engl. J. Med. 2017, 376, 1121–1130. [Google Scholar] [CrossRef]
- Schnadower, D.; I Tarr, P.; Casper, T.C.; Gorelick, M.H.; Dean, M.J.; O’cOnnell, K.J.; Mahajan, P.; Chun, T.H.; Bhatt, S.R.; Roskind, C.G.; et al. Randomised controlled trial of Lactobacillus rhamnosus (LGG) versus placebo in children presenting to the emergency department with acute gastroenteritis: The PECARN probiotic study protocol. BMJ Open 2017, 7, e018115. [Google Scholar] [CrossRef]
- Lewis, K. Vesikari Clinical Severity Scoring System Manual. 2011. Available online: https://www.path.org/our-impact/resources/vesikari-clinical-severity-scoring-system-manual/ (accessed on 3 September 2025).
- Wikswo, M.E.; Weinberg, G.A.; Szilagyi, P.G.; Selvarangan, R.; Harrison, C.J.; Klein, E.J.; A Englund, J.; Sahni, L.C.; A Boom, J.; Halasa, N.B.; et al. Evaluation of a Modified Vesikari Severity Score as a Research Tool for Assessing Pediatric Acute Gastroenteritis. J. Pediatr. Infect. Dis. Soc. 2024, 13, 547–550. [Google Scholar] [CrossRef] [PubMed]
- Schnadower, D.; Tarr, P.I.; Gorelick, M.H.; O’COnnell, K.; Roskind, C.G.; Powell, E.C.; Rao, J.; Bhatt, S.; Freedman, S.B. Validation of the modified Vesikari score in children with gastroenteritis in 5 US emergency departments. J. Pediatr. Gastroenterol. Nutr. 2013, 57, 514–519. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.-L.; Chen, S.-Y.; Tsai, C.-N.; Chao, H.-C.; Lai, M.-W.; Chang, Y.-J.; Chen, C.-L.; Chiu, C.-H. Complicated norovirus infection and assessment of severity by a modified Vesikari disease score system in hospitalized children. BMC Pediatr. 2016, 16, 162. [Google Scholar] [CrossRef] [PubMed]
- Sowmyanarayanan, T.V.; Ramani, S.; Sarkar, R.; Arumugam, R.; Warier, J.P.; Moses, P.D.; Simon, A.; Agarwal, I.; Bose, A.; Arora, R.; et al. Severity of RV Gastroenteritis in Indian Children Requiring Hospitalization. Vaccine 2012, 30, 167–172. [Google Scholar] [CrossRef]
- Armah, G.E.; Sow, S.O.; Breiman, R.F.; Dallas, M.J.; Tapia, M.D.; Feikin, D.R.; Binka, F.N.; Steele, A.D.; Laserson, K.F.; Ansah, N.A.; et al. Efficacy of Pentavalent RV Vaccine against Severe RV Gastroenteritis in Infants in Developing Countries in Sub-Saharan Africa: A Randomised, Double-Blind, Placebo-Controlled Trial. Lancet 2010, 376, 606–614. [Google Scholar] [CrossRef]
- Madhi, S.A.; Cunliffe, N.A.; Steele, D.; Witte, D.; Kirsten, M.; Louw, C.; Ngwira, B.; Victor, J.C.; Gillard, P.H.; Cheuvart, B.B.; et al. Effect of Human RV Vaccine on Severe Diarrhea in African Infants. N. Engl. J. Med. 2010, 362, 289–298. [Google Scholar] [CrossRef] [PubMed]
- Guarino, A.; Ashkenazi, S.; Gendrel, D.; Vecchio, A.L.; Shamir, R.; Szajewska, H. European Society for Pediatric Gastroenterology, Hepatology, and Nutrition/European Society for Pediatric Infectious Diseases evidence-based guidelines for the management of acute gastroenteritis in children in Europe: Update 2014. J. Pediatr. Gastroenterol. Nutr. 2014, 59, 132–152. [Google Scholar] [CrossRef] [PubMed]
- Hoxha, T.; Xhelili, L.; Azemi, M.; Avdiu, M.; IsmailiJaha, V.; EfendijaBeqa, U.; GrajcevciUka, V. Comparing the Accuracy of the Three Dehydration Scales in Children with Acute Diarrhea in a Developing Country of Kosovo. Mater. Sociomed. 2015, 27, 140–143. [Google Scholar] [CrossRef] [PubMed]
- Manouana, G.P.; Nguema-Moure, P.A.; Tomazatos, A.; Maloum, M.N.; Bock, C.-T.; Kremsner, P.G.; Velavan, T.P.; Adegnika, A.A.; Niendorf, S. RT-PCR-based assessment of the SD Bioline Rota/Adeno antigen-based test in infants with and without diarrhea. Virol. J. 2023, 20, 40. [Google Scholar] [CrossRef]
- Wang, Y.; Zheng, Y.; Li, Y.; Zhang, S.; Wang, X.; Zong, H.; Huang, W.; Kong, D.; Jiang, Y.; Liu, P.; et al. Development of a rapid homogeneous immunoassay for detection of rotavirus in stool samples. Front. Public Health 2022, 10, 975720. [Google Scholar] [CrossRef]
- Medycyna Praktyczna. Microbiological, Immunological, and Molecular Diagnostics of the Most Common Foodborne Infections in Children. Available online: https://www.mp.pl/pediatria/praktyka-kliniczna/badania-laboratoryjne/180277 (accessed on 16 June 2025).
- World Health Organization. Manual of Rotavirus Detection and Characterization Methods. Available online: https://iris.who.int/bitstream/handle/10665/342905/WER9628-301-319-eng-fre.pdf?sequence=1 (accessed on 16 June 2025).
- The Public Health Laboratory Network. Rotavirus—Laboratory Case Definition. Available online: https://www.health.gov.au/sites/default/files/documents/2022/06/rotavirus-laboratory-case-definition.pdf (accessed on 18 June 2025).
- Hughes, J.H.; Tuomari, A.V.; Mann, D.R.; Hamparian, V.V. Latex immunoassay for rapid detection of rotavirus. J. Clin. Microbiol. 1984, 20, 441–447. [Google Scholar] [CrossRef]
- Long, S.S.; Prober, C.G.; Fischer, M.; Kimberlin, D. Principles and Practice of Pediatric Infectious Diseases, 6th ed.; Elsevier Health Sciences: Amsterdam, The Netherlands, 2022. [Google Scholar]
- James, V.L.A.; Lambden, P.R.; Caul, E.O.; Clarke, I.N. Enzyme-linked immunosorbent assay based on recombinant human group C rotavirus inner capsid protein (VP6) to detect human group C rotaviruses in fecal samples. J. Clin. Microbiol. 1998, 36, 3178–3181. [Google Scholar] [CrossRef]
- Hung, P.J.; Chen, C.C. Diagnostic accuracy of rotavirus antigen tests in children: A systematic review and meta-analysis. Trop. Med. Int. Health 2023, 28, 72–79. [Google Scholar] [CrossRef] [PubMed]
- Merrill, C.R.; Goldman, D.; Sedman, S.A.; Ebert, M.H. Ultrasensitive stain for proteins in polyacrylamide gels shows regional variation in cerebrospinal fluid proteins. Science 1981, 211, 1437–1438. [Google Scholar] [CrossRef]
- Oakley, B.R.; Kirsch, D.R.; Morris, N.R. A simplified ultrasensitive silver stain for detecting proteins in polyacrylamide gels. Anal. Biochem. 1980, 105, 361–363. [Google Scholar] [CrossRef]
- Dash, S.K.; Kumar, K.; Tewari, A.; Varshney, P.; Goel, A.; Bhatia, A.K. Detection of rotavirus from hospitalized diarrheic children in Uttar Pradesh, India. Indian J. Microbiol. 2012, 52, 472–477. [Google Scholar] [CrossRef][Green Version]
- Hammami, S.; Castro, A.E.; Osburn, B.I. Comparison of polyacrylamide gel electrophoresis, an enzyme-linked immunosorbent assay, and an agglutination test for the direct identification of bovine rotavirus from feces and coelectrophoresis of viral RNAs. J. Vet. Diagn. Investig. 1990, 2, 184–190. [Google Scholar] [CrossRef]
- Kasempimolporn, S.; Louisirirotchanakul, S.; Sinarachatanant, P.; Wasi, C. Polyacrylamide gel electrophoresis and silver staining for detection of rotavirus in stools from diarrheic patients in Thailand. J. Clin. Microbiol. 1988, 26, 158–160. [Google Scholar] [CrossRef] [PubMed]
- Kusumakar, A.L.; Savita; Malik, Y.; Minakshi; Prasad, G. Detection of human rotavirus in hospitalized diarrheic children in central India. Indian J. Microbiol. 2007, 47, 373–376. [Google Scholar] [CrossRef][Green Version]
- Chen, C.C.; Hsiao, K.Y.; Bai, C.H.; Wang, Y.H. Investigation of the diagnostic performance of the SARS-CoV-2 saliva antigen test: A meta-analysis. J. Microbiol. Immunol. Infect. 2022, 55, 84–93. [Google Scholar] [CrossRef]
- Centers for Disease Control and Prevention (CDC). Clinical Overview of Rotavirus. Rotavirus. Available online: https://www.cdc.gov/rotavirus/hcp/clinical-overview/?CDC_AAref_Val=https://www.cdc.gov/rotavirus/clinical.html (accessed on 16 June 2025).
- Carossino, M.; Barrandeguy, M.E.; Erol, E.; Li, Y.; Balasuriya, U.B.R. Development and evaluation of a one-step multiplex real-time TaqMan® RT-qPCR assay for the detection and genotyping of equine G3 and G14 rotaviruses in fecal samples. Virol. J. 2019, 16, 49. [Google Scholar] [CrossRef]
- Steyer, A.; Mičetić-Turk, D.; Fijan, S. The efficacy of probiotics as antiviral agents for the treatment of rotavirus gastrointestinal infections in children: An updated overview of literature. Microorganisms 2022, 10, 2392. [Google Scholar] [CrossRef]
- Kosiak, W.; Swieton, D.; Piskunowicz, M. Sonographic inferior vena cava/aorta diameter index, a new approach to the body fluid status assessment in children and young adults in emergency ultrasound—Preliminary study. Am. J. Emerg. Med. 2008, 26, 320–325. [Google Scholar] [CrossRef] [PubMed]
- Octavius, G.S.; Imanuelly, M.; Wibowo, J.; Heryadi, N.K.; Widjaja, M. Inferior vena cava to aorta ratio in dehydrated pediatric patients: A systematic review and meta-analysis. Clin. Exp. Pediatr. 2023, 66, 477–484. [Google Scholar] [CrossRef]
- El Amrousy, D.; Gamal, R.; Elrifaey, S.; Hassan, S. Non-invasive Assessment of Significant Dehydration in Infants Using the Inferior Vena Cava to Aortic Ratio: Is it Useful? J. Pediatr. Gastroenterol. Nutr. 2018, 66, 882–886. [Google Scholar] [CrossRef]
- Paketçi, C.; Edem, P.; Okur, D.; Sarıoğlu, F.C.; Güleryüz, H.; Bayram, E.; Kurul, S.H.; Yiş, U. Rotavirus encephalopathy with concomitant acute cerebellitis: Report of a case and review of the literature. Turk. J. Pediatr. 2020, 62, 119–124. [Google Scholar] [CrossRef] [PubMed]
- Dennehy, P.H.; Gauntlett, D.R.; Spangenberger, S.E. Choice of reference assay for the detection of rotavirus in fecal specimens: Electron microscopy versus enzyme immunoassay. J. Clin. Microbiol. 1990, 28, 1280–1283. [Google Scholar] [CrossRef] [PubMed]
- Marchlewicz, B.; Spiewak, M.; Lampinen, J. Evaluation of Abbott TESTPACK ROTAVIRUS with clinical specimens. J. Clin. Microbiol. 1988, 26, 2456–2458. [Google Scholar] [CrossRef]
- Brooks, R.G.; Brown, L.; Franklin, R.B. Comparison of a new rapid test (TestPack Rotavirus) with standard enzyme immunoassay and electron microscopy for the detection of rotavirus in symptomatic hospitalized children. J. Clin. Microbiol. 1989, 27, 775–777. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Liu, M.; Sugata, K.; Wang, Y.; Hull, J.; Foytich, K.; Jiang, B. Development of a new enzyme immunoassay for improved detection of rotavirus in fecal specimens of vaccinated infants. J. Clin. Virol. 2018, 99–100, 44–49. [Google Scholar] [CrossRef]
- Kumar, N.; Malik, Y.S.; Kumar, S.; Sharma, K.; Sircar, S.; Saurabh, S.; Gulati, B.R.; Singh, N.; Singh, A.K.; Joshi, V.G.; et al. Peptide-Recombinant VP6 Protein Based Enzyme Immunoassay for the Detection of Group A Rotaviruses in Multiple Host Species. PLoS ONE 2016, 11, e0159027. [Google Scholar] [CrossRef]
- Moon, S.-S.; Wang, H.; Brown, K.; Wang, Y.; Bessy, T.; Greenberg, H.B.; Jiang, B. Development and validation of a VP7-specific EIA for determining the potency and stability of inactivated rotavirus vaccine. J. Virol. Methods 2025, 332. [Google Scholar] [CrossRef]
- Malik, Y.S.; Verma, A.K.; Kumar, N.; Touil, N.; Karthik, K.; Tiwari, R.; Bora, D.P.; Dhama, K.; Ghosh, S.; Hemida, M.G.; et al. Advances in diagnostic approaches for viral etiologies of diarrhea: From the lab to the field. Front. Microbiol. 2019, 10, 1957. [Google Scholar] [CrossRef]
- Mirabile, A.; Sangiorgio, G.; Bonacci, P.G.; Bivona, D.; Nicitra, E.; Bonomo, C.; Bongiorno, D.; Stefani, S.; Musso, N. Advancing pathogen identification: The role of digital PCR in enhancing diagnostic power in different settings. Diagnostics 2024, 14, 1598. [Google Scholar] [CrossRef]
- Kiulia, N.M.; Gonzalez, R.; Thompson, H.; Aw, T.G.; Rose, J.B. Quantification and trends of rotavirus and enterovirus in untreated sewage using reverse transcription droplet digital PCR. Food Environ. Virol. 2021, 13, 154–169. [Google Scholar] [CrossRef]
- Huge, B.J.; North, D.; Mousseau, C.B.; Bibby, K.; Dovichi, N.J.; Champion, M.M. Comparison of RT-dPCR and RT-qPCR and the effects of freeze-thaw cycle and glycine release buffer for wastewater SARS-CoV-2 analysis. Sci. Total Environ. 2022, 12, 20641. [Google Scholar] [CrossRef]
- Matthijnssens, J.; Ciarlet, M.; Heiman, E.; Arijs, I.; Delbeke, T.; McDonald, S.M.; Palombo, E.A.; Iturriza-GómAra, M.; Maes, P.; Patton, J.T.; et al. Full genome-based classification of rotaviruses reveals a common origin between human Wa-like and porcine rotavirus strains and human DS-1-like and bovine rotavirus strains. J. Virol. 2008, 82, 3204–3219. [Google Scholar] [CrossRef]
- Minami-Fukuda, F.; Nagai, M.; Takai, H.; Murakami, T.; Ozawa, T.; Tsuchiaka, S.; Okazaki, S.; Katayama, Y.; Oba, M.; Nishiura, N.; et al. Detection of bovine group A rotavirus using rapid antigen detection kits, RT-PCR and next-generation DNA sequencing. J. Vet. Med. Sci. 2013, 75, 1651–1655. [Google Scholar] [CrossRef][Green Version]
- Dung, T.T.N.; Duy, P.T.; Sessions, O.M.; Sangumathi, U.K.; Phat, V.V.; Tam, P.T.T.; To, N.T.N.; Phuc, T.M.; Chau, T.T.H.; Chau, N.N.M.; et al. A universal genome sequencing method for rotavirus A from human fecal samples which identifies segment reassortment and multi-genotype mixed infection. BMC Genom. 2017, 18, 324. [Google Scholar] [CrossRef] [PubMed]
- Kuba, Y.; Takemae, N.; Kawato, S.; Oba, K.; Taniguchi, K.; Kageyama, T. Evaluation of rapid amplicon-based nanopore sequencing using the latest chemistry for accurate whole genome analysis of influenza A virus in clinical samples. Jpn. J. Infect. Dis. 2025. [Google Scholar] [CrossRef] [PubMed]
- Yandle, Z.; Gonzalez, G.; Carr, M.; Matthijnssens, J.; De Gascun, C. A viral metagenomic protocol for nanopore sequencing of group A rotavirus. J. Virol. Methods 2023, 312, 114664. [Google Scholar] [CrossRef] [PubMed]
- Krasnikov, N.; Gulyukin, A.; Aliper, T.; Yuzhakov, A. Complete genome characterization by nanopore sequencing of rotaviruses A, B, and C circulating on large-scale pig farms in Russia. Virol. J. 2024, 21, 289. [Google Scholar] [CrossRef]
- Dunkin, M.A. Blood in Stool. WebMD. Available online: https://www.webmd.com/digestive-disorders/blood-in-stool (accessed on 18 June 2025).
- Spandorfer, P.R. Oral versus intravenous rehydration of moderately dehydrated children: A randomized, controlled trial. Pediatrics 2005, 115, 295–301. [Google Scholar] [CrossRef]
- Fonseca, B.K.; Holdgate, A.; Craig, J.C. Enteral vs intravenous rehydration therapy for children with gastroenteritis. Arch. Pediatr. Adolesc. Med. 2004, 158, 483–490. [Google Scholar] [CrossRef]
- Civra, A.; Francese, R.; Donalisio, M.; Tonetto, P.; Coscia, A.; Sottemano, S.; Balestrini, R.; Faccio, A.; Cavallarin, L.; Moro, G.E.; et al. Human Colostrum and Derived Extracellular Vesicles Prevent Infection by Human Rotavirus and Respiratory Syncytial Virus in Vitro. J. Hum. Lact. 2021, 37, 122–134. [Google Scholar] [CrossRef]
- Campo, J.J.; Seppo, A.E.; Randall, A.Z.; Pablo, J.; Hung, C.; Teng, A.; Shandling, A.D.; Truong, J.; Oberai, A.; Miller, J.; et al. Human milk antibodies to global pathogens reveal geographic and interindividual variations in IgA and IgG. J. Clin. Investig. 2024, 134, e168789. [Google Scholar] [CrossRef] [PubMed]
- Superti, F.; Ammendolia, M.G.; Valenti, P.; Seganti, L. Antirotaviral activity of milk proteins: Lactoferrin prevents rotavirus infection in the enterocyte-like cell line HT-29. Med. Microbiol. Immunol. 1997, 186, 83–91. [Google Scholar] [CrossRef]
- Valenti, P.; Antonini, G. Lactoferrin: An important host defence against microbial and viral attack. Cell Mol. Life Sci. 2005, 62, 2576–2587. [Google Scholar] [CrossRef]
- Jeong, Y.Y.; Lee, G.Y.; Yoo, Y.C. Bovine lactoferricin induces intestinal epithelial cell activation through phosphorylation of FAK and paxillin and prevents rotavirus infection. J. Microbiol. Biotechnol. 2021, 31, 1175–1182. [Google Scholar] [CrossRef]
- Owens, J.R.; Broadhead, R.; Hendrickse, R.G.; Jaswal, O.P.; Gangal, R.N. Loperamide in the 13ktreatment of acute gastroenteritis in early childhood: Report of a two-centre, double-blind, controlled clinical trial. Ann. Trop. Paediatr. 1981, 1, 135–141. [Google Scholar] [CrossRef]
- Hagbom, M.; Novak, D.; Ekström, M.; Khalid, Y.; Andersson, M.; Lindh, M.; Nordgren, J.; Svensson, L.; Mansur, A. Ondansetron treatment reduces rotavirus symptoms—A randomized double-blinded placebo-controlled trial. PLoS ONE 2017, 12, e0186824. [Google Scholar] [CrossRef]
- Kang, G.; Thuppal, S.V.; Srinivasan, R.; Sarkar, R.; Subashini, B.; Venugopal, S.; Sindhu, K.; Anbu, D.; Parez, N.; Svensson, L.; et al. Racecadotril in the management of rotavirus and non-rotavirus diarrhea in under-five children: Two randomized, double-blind, placebo-controlled trials. Indian Pediatr. 2016, 53, 595–600. [Google Scholar] [CrossRef]
- Dewi, M.R.; Soenarto, Y.; Karyana, I.P.G. Efficacy of synbiotic treatment in children with acute rotavirus diarrhea. Paediatr. Indones. 2015, 55, 74–78. [Google Scholar] [CrossRef][Green Version]
- Jiang, L.; Tang, A.; Song, L.; Tong, Y.; Fan, H. Advances in the development of antivirals for rotavirus infection. Front. Immunol. 2023, 14, 1041149. [Google Scholar] [CrossRef] [PubMed]
- Jiang, C.X.; Xu, C.D.; Yang, C.Q. Therapeutic effects of zinc supplement as adjunctive therapy in infants and young children with rotavirus enteritis. Zhongguo Dang Dai Er Ke Za Zhi 2016, 18, 826–830. [Google Scholar]
- Cai, Y.; Wang, X.; Li, C.; Li, F.; Yan, Z.; Ma, N.; Sun, M. Probiotics combined with zinc and selenium preparation in the treatment of child rotavirus enteritis. Am. J. Transl. Res. 2022, 14, 1043–1050. [Google Scholar]
- Xu, N.; Zhang, W.; Huo, J.; Tao, R.; Jin, T.; Zhang, Y.; Wang, Y.; Zhu, L.; Li, J.; Yao, Q.; et al. Characterization of changes in the intestinal microbiome following combination therapy with zinc preparation and conventional treatment for children with rotavirus enteritis. Front. Cell. Infect. Microbiol. 2023, 13, 1153701. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.-F.; Liu, P.-Y.; Chen, Y.-Y.; Nong, B.-R.; Huang, I.-F.; Hsieh, K.-S.; Chen, K.-T. Three-combination probiotics therapy in children with salmonella and rotavirus gastroenteritis. J. Clin. Gastroenterol. 2014, 48, 37–42. [Google Scholar] [CrossRef]
- Aggarwal, S.; Upadhyay, A.; Shah, D.; Teotia, N.; Agarwal, A.; Jaiswal, V. Lactobacillus GG for treatment of acute childhood diarrhoea: An open labelled, randomized controlled trial. Indian J. Med. Res. 2014, 139, 379–385. [Google Scholar]
- Sindhu, K.N.C.; Sowmyanarayanan, T.V.; Paul, A.; Babji, S.; Ajjampur, S.S.R.; Priyadarshini, S.; Sarkar, R.; Balasubramanian, K.A.; Wanke, C.A.; Ward, H.D.; et al. Immune response and intestinal permeability in children with acute gastroenteritis treated with Lactobacillus rhamnosus GG: A randomized, double-blind, placebo-controlled trial. Clin. Infect. Dis. 2014, 58, 1107–1115. [Google Scholar] [CrossRef] [PubMed]
- Lee, D.K.; Park, J.E.; Kim, M.J.; Seo, J.G.; Lee, J.H.; Ha, N.J. Probiotic bacteria, B. longum and L. acidophilus inhibit infection by rotavirus in vitro and decrease the duration of diarrhea in pediatric patients. Clin. Res. Hepatol. Gastroenterol. 2015, 39, 237–244. [Google Scholar] [CrossRef]
- Sobouti, B.; Noorbakhsh, S.; Ashraf, H.; Ashraf-Talesh, S. Use of probiotic for the treatment of acute rotavirus diarrhea in children: A randomized single-blind controlled trial. Int. J. Child Adolesc. 2016, 2, 5–9. [Google Scholar]
- Das, S.; Gupta, P.K.; Das, R.R. Efficacy and safety of Saccharomyces boulardii in acute rotavirus diarrhea: Double-blind randomized controlled trial from a developing country. J. Trop. Pediatr. 2016, 62, 464–470. [Google Scholar] [CrossRef][Green Version]
- Park, M.S.; Kwon, B.; Ku, S.; Ji, G.E. The Efficacy of Bifidobacterium longum BORI and Lactobacillus acidophilus AD031 Probiotic Treatment in Infants with Rotavirus Infection. Nutrients 2017, 9, 887. [Google Scholar] [CrossRef] [PubMed]
- Hong Chau, T.T.; Minh Chau, N.N.; Hoang Le, N.T.; Chung The, H.; Voong Vinh, P.; Nguyen To, N.T.; Ngoc, N.M.; Tuan, H.M.; Chau Ngoc, T.L.; Kolader, M.-E.; et al. A Double-Blind, Randomized, Placebo-Controlled Trial of Lactobacillus acidophilus for the Treatment of Acute Watery Diarrhea in Vietnamese Children. Pediatr. Infect. Dis. J. 2018, 37, 35–42. [Google Scholar] [CrossRef]
- Shin, D.Y.; Yi, D.Y.; Jo, S.; Lee, Y.M.; Kim, J.-H.; Kim, W.; Park, M.R.; Yoon, S.M.; Kim, Y.; Yang, S.; et al. Effect of a New Lactobacillus plantarum Product, LRCC5310, on Clinical Symptoms and Virus Reduction in Children with Rotaviral Enteritis. Medicine 2020, 99, e22192. [Google Scholar] [CrossRef] [PubMed]
- He, Z.; Lan, L.; Chen, N.; Cheng, Y.; Gong, S. The efficacy of lactobacillus reuteri in conjunction with racecadotril in managing pediatric rotavirus enteritis and its impact on intestinal mucosa and immune function. Afr. Health Sci. 2025, 25, 38–45. [Google Scholar] [CrossRef] [PubMed]
- Szymański, H.; Szajewska, H. Lack of Efficacy of Lactobacillus reuteri DSM 17938 for the Treatment of Acute Gastroenteritis: A Randomized Controlled Trial. Pediatr. Infect. Dis. J. 2019, 38, e237–e242. [Google Scholar] [CrossRef]
- Mourey, F.; Sureja, V.; Kheni, D.; Shah, P.; Parikh, D.; Upadhyay, U.; Satia, M.; Shah, D.; Troise, C.; Decherf, A. A Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial of Saccharomyces boulardii in Infants and Children with Acute Diarrhea. Pediatr. Infect. Dis. J. 2020, 39, e347–e351. [Google Scholar] [CrossRef]
- Xie, Y.M.; Wang, L.Y.; Gao, S.; Wang, Z.L. Effect of ingested immunoglobulin on sIgA expression in pediatric rotavirus enteritis. Sichuan Da Xue Xue Bao Yi Xue Ban 2015, 46, 71–74. [Google Scholar]
- Williams, D. Treatment of Rotavirus-Associated Diarrhea Using Enteral Immunoglobulins for Pediatric Stem Cell Transplant Patients. J. Oncol. Pharm. Pract. 2015, 21, 238–240. [Google Scholar] [CrossRef]
- Flerlage, T.; Hayden, R.; Cross, S.J.; Dallas, R.; Srinivasan, A.; Tang, L.; Sun, Y.; Maron, G. Rotavirus Infection in Pediatric Allogeneic Hematopoietic Cell Transplant Recipients: Clinical Course and Experience Using Nitazoxanide and Enterally Administered Immunoglobulins. Pediatr. Infect. Dis. J. 2018, 37, 176–181. [Google Scholar] [CrossRef]
- Banerjee, S.; Sarkar, R.; Mukherjee, A.; Miyoshi, S.I.; Kitahara, K.; Halder, P.; Koley, H.; Chawla-Sarkar, M. Quercetin, a Flavonoid, Combats Rotavirus Infection by Deactivating Rotavirus-Induced Pro-Survival NF-κB Pathway. Front. Microbiol. 2022, 13, 951716. [Google Scholar] [CrossRef]
- Van Dycke, J.; Arnoldi, F.; Papa, G.; Vandepoele, J.; Burrone, O.R.; Mastrangelo, E.; Tarantino, D.; Heylen, E.; Neyts, J.; Rocha-Pereira, J. A Single Nucleoside Viral Polymerase Inhibitor Against Norovirus, Rotavirus, and Sapovirus-Induced Diarrhea. J. Infect. Dis. 2018, 218, 1753–1758. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.; Ding, S.; Yin, Y.; Xu, L.; Li, P.; Peppelenbosch, M.P.; Pan, Q.; Wang, W. Suppression of Pyrimidine Biosynthesis by Targeting DHODH Enzyme Robustly Inhibits Rotavirus Replication. Antivir. Res. 2019, 167, 35–44. [Google Scholar] [CrossRef]
- Zhou, X.; Li, Y.; Li, T.; Cao, J.; Guan, Z.; Xu, T.; Jia, G.; Ma, G.; Zhao, R. Portulaca oleracea L. Polysaccharide Inhibits Porcine Rotavirus In Vitro. Animals 2023, 13, 2306. [Google Scholar] [CrossRef]
- Kim, J.-H.; Kim, K.; Kim, W. Genipin Inhibits Rotavirus-Induced Diarrhea by Suppressing Viral Replication and Regulating Inflammatory Responses. Sci. Rep. 2020, 10, 15836. [Google Scholar] [CrossRef]
- Eichwald, C.; De Lorenzo, G.; Schraner, E.M.; Papa, G.; Bollati, M.; Swuec, P.; de Rosa, M.; Milani, M.; Mastrangelo, E.; Ackermann, M.; et al. Identification of a Small Molecule That Compromises the Structural Integrity of Viroplasms and Rotavirus Double-Layered Particles. J. Virol. 2018, 92, e01943-17. [Google Scholar] [CrossRef]
- La Frazia, S.; Ciucci, A.; Arnoldi, F.; Coira, M.; Gianferretti, P.; Angelini, M.; Belardo, G.; Burrone, O.R.; Rossignol, J.-F.; Santoro, M.G. Thiazolides, a New Class of Antiviral Agents Effective Against Rotavirus Infection, Target Viral Morphogenesis, Inhibiting Viroplasm Formation. J. Virol. 2013, 87, 11096–11106. [Google Scholar] [CrossRef]
- Tohmé, M.J.; Giménez, M.; Peralta, A.; Colombo, M.; Delgui, L. Ursolic Acid: A Novel Antiviral Compound Inhibiting Rotavirus Infection In Vitro. Int. J. Antimicrob. Agents 2019, 54, 601–609. [Google Scholar] [CrossRef]
- Santos-Ferreira, N.; Van Dycke, J.; Chiu, W.; Neyts, J.; Matthijnssens, J.; Rocha-Pereira, J. Molnupiravir Inhibits Human Norovirus and Rotavirus Replication in 3D Human Intestinal Enteroids. Antivir. Res. 2024, 223, 105839. [Google Scholar] [CrossRef]
- Shen, Z.; He, H.; Wu, Y.; Li, J. Cyclosporin A Inhibits Rotavirus Replication and Restores Interferon-Beta Signaling Pathway In Vitro and In Vivo. PLoS ONE 2013, 8, e71815. [Google Scholar] [CrossRef]
- Chanda, S.D.; Banerjee, A.; Chawla, S.M. Cordycepin, an Adenosine Analogue, Executes Anti-Rotaviral Effect by Stimulating Induction of Type I Interferon. J. Virol. Antivir. Res. 2015, 4, 2. [Google Scholar] [CrossRef]
- Hardy, M.E.; Hendricks, J.M.; Paulson, J.M.; Faunce, N.R. 18 β-Glycyrrhetinic Acid Inhibits Rotavirus Replication in Culture. Virol. J. 2012, 9, 96. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Hu, X.; Wu, J.; Lin, X.; Chen, R.; Lu, C.; Song, X.; Leng, Q.; Li, Y.; Kuang, X.; et al. ML241 Antagonizes ERK 1/2 Activation and Inhibits Rotavirus Proliferation. Viruses 2024, 16, 623. [Google Scholar] [CrossRef] [PubMed]
- Huang, H.; Liao, D.; He, B.; Pu, R.; Cui, Y.; Zhou, G. Deoxyshikonin inhibited rotavirus replication by regulating autophagy and oxidative stress through SIRT1/FoxO1/Rab7 axis. Microb. Pathog. 2023, 178, 106065. [Google Scholar] [CrossRef] [PubMed]
- Zhang, R.; Feng, C.; Luo, D.; Zhao, R.; Kannan, P.R.; Yin, Y.; Iqbal, M.Z.; Hu, Y.; Kong, X. Metformin Hydrochloride significantly inhibits rotavirus infection in Caco2 cell line, intestinal organoids, and mice. Pharmaceuticals 2023, 16, 1279. [Google Scholar] [CrossRef]
- Cui, J.; Fu, X.; Xie, J.; Gao, M.; Hong, M.; Chen, Y.; Su, S.; Li, S. Critical role of cellular cholesterol in bovine rotavirus infection. Virol. J. 2014, 11, 98. [Google Scholar] [CrossRef]
- Dou, X.; Li, Y.; Han, J.; Zarlenga, D.S.; Zhu, W.; Ren, X.; Dong, N.; Li, X.; Li, G. Cholesterol of lipid rafts is a key determinant for entry and post-entry control of porcine rotavirus infection. BMC Vet. Res. 2018, 14, 45. [Google Scholar] [CrossRef]
- Patra, U.; Mukhopadhyay, U.; Sarkar, R.; Mukherjee, A.; Chawla-Sarkar, M. RA-839, a selective agonist of Nrf2/ARE pathway, exerts potent anti-rotaviral efficacy in vitro. Antivir. Res. 2019, 161, 53–62. [Google Scholar] [CrossRef]
- López, T.; López, S.; Arias, C.F. The tyrosine kinase inhibitor genistein induces the detachment of rotavirus particles from the cell surface. Virus Res. 2015, 210, 141–148. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Ding, S.; Feng, N.; Mooney, N.; Ooi, Y.S.; Ren, L.; Diep, J.; Kelly, M.R.; Yasukawa, L.L.; Patton, J.T.; et al. Drebrin restricts rotavirus entry by inhibiting dynamin-mediated endocytosis. Proc. Natl. Acad. Sci. USA 2017, 114, E3642–E3651. [Google Scholar] [CrossRef]
- Yin, Y.; Chen, S.; Hakim, M.S.; Wang, W.; Xu, L.; Dang, W.; Qu, C.; Verhaar, A.P.; Su, J.; Fuhler, G.M.; et al. 6-Thioguanine inhibits rotavirus replication through suppression of Rac1 GDP/GTP cycling. Antivir. Res. 2018, 156, 92–101. [Google Scholar] [CrossRef]
- Tian, Z.; Zhang, J.; He, H.; Li, J.; Wu, Y.; Shen, Z. MiR-525-3p mediates antiviral defense to rotavirus infection by targeting nonstructural protein 1. Biochim. Biophys. Acta 2017, 1863, 3212–3225. [Google Scholar] [CrossRef]
- Zhou, Y.; Chen, L.; Du, J.; Hu, X.; Xie, Y.; Wu, J.; Lin, X.; Yin, N.; Sun, M.; Li, H. MicroRNA-7 inhibits rotavirus replication by targeting viral NSP5 in vivo and in vitro. Viruses 2020, 12, 209. [Google Scholar] [CrossRef] [PubMed]
- Yin, Y.; Dang, W.; Zhou, X.; Xu, L.; Wang, W.; Cao, W.; Chen, S.; Su, J.; Cai, X.; Xiao, S.; et al. PI3K-Akt-mTOR axis sustains rotavirus infection via the 4E-BP1 mediated autophagy pathway and represents an antiviral target. Virulence 2017, 9, 83–98. [Google Scholar] [CrossRef] [PubMed]
- Crawford, S.E.; Desselberger, U. Lipid droplets form complexes with viroplasms and are crucial for rotavirus replication. Curr. Opin. Virol. 2016, 19, 11–15. [Google Scholar] [CrossRef]
- Contin, R.; Arnoldi, F.; Mano, M.; Burrone, O.R. Rotavirus replication requires a functional proteasome for effective assembly of viroplasms. J. Virol. 2011, 85, 2781–2792. [Google Scholar] [CrossRef]
- López, T.; Silva-Ayala, D.; López, S.; Arias, C.F. Replication of the rotavirus genome requires an active ubiquitin-proteasome system. J. Virol. 2011, 85, 11964–11971. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.; Wang, Y.; Li, P.; Yin, Y.; Bijvelds, M.J.; de Jonge, H.R.; Peppelenbosch, M.P.; Kainov, D.E.; Pan, Q. Drug screening identifies gemcitabine inhibiting rotavirus through alteration of pyrimidine nucleotide synthesis pathway. Antivir. Res. 2020, 180, 104823. [Google Scholar] [CrossRef] [PubMed]
The Vesikari Scoring System (VSS) [17] | ||||||||
Characteristics/Points | 1 | 2 | 3 | |||||
Number of stools/days | 1–3 | 4–5 | ≥6 | |||||
Duration of diarrhea (days) | 1–4 | 5 | ≥6 | |||||
Number of emesis events/day | 1 | 2–4 | ≥5 | |||||
Duration of emesis (days) | 1 | 2 | ≥3 | |||||
Rectal temperature (°C) | 37.1–38.4 | 38.5–38.9 | ≥39 | |||||
Dehydration | - | 1–5% | ≥6% | |||||
Treatment | Rehydration | Hospitalization | - | |||||
The Clark Scale [18] | ||||||||
Characteristics/Points | 1 | 2 | 3 | |||||
Number of stools/days | 2–4 | 5–7 | ≥8 | |||||
Duration of diarrhea (days) | 1–4 | 5–7 | ≥8 | |||||
Number of emesis events/day | 1–3 | 4–6 | ≥7 | |||||
Duration of emesis (days) | 2 | 3–5 | ≥6 | |||||
Rectal temperature (°C) | 38.1–38.2 | 38.3–38.7 | ≥38.8 | |||||
Temperature duration (days) | 1–2 | 3–4 | ≥5 | |||||
Behavioral symptoms | Irritable/less playful | Lethargic/listless | Seizures | |||||
Duration of behavioral symptoms (days) | 1–2 | 3–4 | ≥5 | |||||
Clinical Dehydration Scale (CDS) [19] | ||||||||
Characteristics/Points | 1 | 2 | 3 | |||||
General appearance | Normal | Thirsty, restless, or lethargic but irritable when touched | Cold, drowsy, limp, sweaty, or comatose | |||||
Eyes | Normal | Slightly sunken | Extremely sunken | |||||
Mucous membrane (tondue) | Sticky | - | ||||||
Tears | Normal | Decreased tears | Absent tears | |||||
The Dehydration: Assessing Kids Accurately (DHAKA) Scale [20] | ||||||||
Characteristics/Points | 0 | 2 | 4 | |||||
General appearance | Normal | Restless, irritable | Lethargic, unconscious | |||||
Tears | Normal | Decreased | Absent | |||||
Skin pinch | Normal | Slow | Very slow | |||||
Respirations | Normal | Deep | - | |||||
Centre for Infectious Disease Research in Zambia (CIDRZ) Scale [21] | ||||||||
Characteristics/Points | 1 | 2 | 3 | Characteristics/Points | ||||
Frequency of vomiting | 2–3 episodes/day | 4–5 episodes/day | ≥6 episodes/day | |||||
General appearance | - | - | Restless/irritable | Lethargic | ||||
Skin pinch test | Normal (instant recoil) | Slow recoil (>2 s) | Very slow | |||||
Tears | Present | - | Absent | - | ||||
Respirations | Normal | - | Deep | - | ||||
The 10- and 4-point Gorelick Scale for dehydration (4-point scale with) [22] | ||||||||
Characteristics | No or minimal dehydration | Moderate to severe dehydration | ||||||
General appearance | Alert | Restless, lethargic, unconscious | ||||||
Tears | Present | Absent | ||||||
Mucous membranes | Moist | Dry, very dry | ||||||
Capillary refill | Normal | Prolonged or minimal | ||||||
Eyes | Normal | Sunken | ||||||
Quality of pulses | Normal | Thready; weak or impalpable | ||||||
Heart rate | Normal | Tachycardia | ||||||
Urine output | Normal | Reduced; not passed in many hours | ||||||
Breathing | Present | Deep; deep and rapid | ||||||
Skin elasticity | Instant recoil | Recoil slowly; recoil > 2 s | ||||||
The World Health Organization (WHO) scale for dehydration [23] | ||||||||
Characteristics/Points | A | B | C | |||||
General appearance | Good, active | Irritable, restless | Lethargic or unconscious | |||||
Eyes | Normal | Sunken | Sunken | |||||
Thirst | Not thirsty, drinks normally | Thirsty, drinks eagerly | Not able to drink or drinks poorly | |||||
Skin turgor | Instant recoil | Slow return to normal | Very slow return to normal |
Symptom or Sign | Vesikari Score System (VSS) | Modified Vesikari Severity Score (MVSS) |
---|---|---|
Max. no. diarrheal stools/24 h | ||
1–3 | 1 | 1 |
4–5 | 2 | 2 |
≥6 | 3 | |
Duration of diarrhea in days | ||
1–4 | 1 | 1 |
5 | 2 | 2 |
≥6 | 3 | |
Max. no. vomiting episodes/24 h | ||
1 | 1 | 1 |
2–4 | 2 | |
≥5 | 3 | 2 |
Duration of vomiting in days | ||
1 | 1 | 1 |
2 | 2 | |
≥3 | 3 | 2 |
Fever | ||
<37.0 °C | 0 | 0 |
37.1–38.4 °C | 1 | 1 |
38.5–38.9 °C | 2 | 2 |
≥39 °C | 3 | 3 |
Dehydration | ||
Little to mild | 1 | 1 |
Mild to moderate (1–5%) | 2 | 2 |
Severe (≥6%) | 3 | 3 |
Treatment | ||
None | 0 | 0 |
Rehydration | 1 | 1 |
Hospitalization | 2 | 2 |
Duration of fever in days | ||
0 | Not assessed | 0 |
1–3 | Not assessed | 1 |
≥3 | Not assessed | 2 |
Gastrointestinal hemorrhage | ||
None | Not assessed | 0 |
Occult blood in stool only | Not assessed | 1 |
Gross bloody stool | Not assessed | 2 |
Convulsion | ||
None | Not assessed | 0 |
Yes | Not assessed | 1 |
With recurrence | Not assessed | 2 |
Assessment of abdomen pain or flatulence | ||
None | Not assessed | 0 |
Flatulence | Not assessed | 1 |
Irritability or pain | Not assessed | 2 |
Total Score | Max 20 | Max 24 |
Scoring Scale | Area Under Curve (AUC) | 95% CI |
---|---|---|
Vesikari Scale | 0.26 | −0.17, 0.56 |
Clark Scale | 0.18 | −0.17, 0.56 |
DHAKA Score | 0.59 | 0.11, 1.00 |
CIDRZ Scale | 0.59 | −0.13, 0.39 |
Dehydration Scale | AUC | 95% CI |
WHO Scale (≥5% dehydration) | 0.71 | 0.65–0.77 |
Gorelick 4-point | 0.71 | 0.63–0.78 |
Gorelick 10-point | 0.74 | 0.68–0.81 |
CDS | 0.54 | 0.45–0.63 |
Type of Test | Suitable Specimen | Target |
---|---|---|
Culture | Fluid stool | Bottles after trypsin pre-treatment. RV has been cultured in primary African green monkey kidney and MA104 cells. |
Latex Agglutination | Fluid stool | Detection of the VP6 antigen of group A RVs. |
Enzyme immunoassay (EIA) | Fluid stool | Detection of the VP6 antigen of group A RVs. |
Immunochromatographic Tests (ICAs) | Fluid stool | A strip with immobilized monoclonal antibodies against the VP6 protein of RV. |
Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) | Fluid stool | PCR assays for RV detection commonly target the VP6 gene, while genotyping assays focus on the VP7 and VP4 genes. |
Serological Tests | Clotted blood 1 week after illness | RV-specific IgM appears in serum about one week after symptom onset. Serum IgA is the primary serologic marker for infection and the most reliable indicator of reinfection in vaccine studies. |
Sanger Sequencing | Fluid stool (RNA extracted) | Partial sequencing of VP4 and VP7 genes for genotyping; highly accurate for identifying known strains but low throughput. |
Next-Generation Sequencing (NGS)—Illumina MiSeq | Fluid stool (RNA extracted, cDNA synthesized) | Whole genome sequencing of all 11 segments; detects mixed infections, point mutations, and reassortment events. Requires relatively high viral load for optimal coverage. |
Next-Generation Sequencing (NGS)—Oxford Nanopore MinION | Fluid stool (RNA extracted) | Long-read sequencing of all RV segments; useful for rapid, real-time detection and full genome assembly directly from clinical specimens. Effective in identifying rare or reassortant strains even in low-resource settings. |
Name of Drug | ESPGHAN/ESPID (2014) Recommendation | ESPGHAN/ESPID (2014) Strength of Recommendation/Quality of Evidence | Research Associated with RV (If Possible) Infection Published from 2014 to 2025 |
---|---|---|---|
Ondansetron (antiemetic drug) | Administered either orally or intravenously—appears potentially effective in managing vomiting in young children with AGE; nevertheless, confirmation of its safety in pediatric populations is required prior to issuing conclusive recommendations. | (II, B) * (strong recommendation, low-quality evidence) ** | In a randomized trial involving 104 children with AGE, a single oral dose of ondansetron (0.15 mg/kg) significantly reduced symptom duration in RV-positive cases (median: 2 days, p = 0.014) and decreased diarrheal episodes in children symptomatic for >3 days (p = 0.028). These findings suggest that ondansetron, by attenuating gastrointestinal symptoms, may facilitate oral rehydration and potentially reduce the need for hospitalization in children with RV AGE [87]. |
Dexamethasone, dimenhydrinate, granisetron, and metoclopramide (antiemetics drugs) | The use of other antiemetic agents is not supported by current evidence. | (II, B) * (strong recommendation, low-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
Loperamide (antimotility and antiperistaltic drug) | Not recommended in the management of AGE in children. | (II, B) * (strong recommendation, very low-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
Diosmectite | Can be considered. | (II, B) * (weak recommendation, moderate-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
Diosmectite Plus LGG | Since Smectite plus LGG and LGG alone provide similar therapeutic outcomes in young children with AGE, using both together is not supported. | (II, B) * (weak recommendation, low-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
Other absorbents (kaolin–pectin and attapulgite-activated charcoal) | Currently not recommended for managing AGE in pediatric patients. | (III, C) * (weak recommendation, very low-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
Racecadotril (antisecretory drugs) | Currently not recommended for managing AGE in pediatric patients. | (II, B) * (weak recommendation, moderate-quality evidence) ** | In two randomized, double-blind, placebo-controlled trials conducted in community and hospital settings in Vellore, India, racecadotril (1.5 mg/kg TID for 3 days) was evaluated in children aged 3–59 months with acute diarrhea. A total of 326 children completed the trials. Racecadotril showed no significant benefit over placebo in reducing diarrheal duration, stool output, or fluid intake, regardless of RV status. Median diarrhea duration remained comparable between groups in both settings. These findings do not support the use of racecadotril for managing AGE in young children [88]. |
Bismuth Subsalicylate | Currently not recommended for managing AGE in pediatric patients. | (III, C) * (strong recommendation, low-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
Zinc | The therapeutic value of zinc in treating AGE is mainly observed in children from developing countries; in regions with sufficient zinc intake, its benefit is minimal. | (I, A) * (strong recommendation, moderate-quality evidence) ** | Described in Section 4.2.1. |
Probiotics | Probiotics, when used with oral rehydration, help reduce the severity of symptoms in children with AGE (I, A). LGG and S. boulardii are recommended as adjuncts to rehydration (I, A) | (I, A) * (strong recommendation, moderate-quality evidence) about probiotics ** and (I, A) * (strong recommendation, low-quality evidence) about LGG and S. boulardii ** | Described in Section 4.2.2. |
Synbiotics | Not recommended until supported by well-documented studies. | (II, B) * (weak recommendation, low-quality evidence) ** | In a study involving 69 children aged 6–59 months with acute diarrhea, 34 received a synbiotic formulation comprising L. casei (4 × 108 CFU), L. rhamnosus (3.5 × 108), S. thermophilus (1 × 108), B. breve (5 × 107), L. acidophilus (5 × 107), B. infantis (4 × 107), L. bulgaricus (1 × 107), and 990 mg of FOS (1 × 109 CFU total) once daily for five days, alongside ORS/IV rehydration and zinc therapy, while 35 children served as controls (Placebo + ORS/IV rehydration therapy + Zn). Significant decrease in diarrhea recovery time. No impact on hospital stay [89]. |
Prebiotics | Currently not recommended for managing AGE in pediatric patients. | (II, B) * (weak recommendation, low-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
Micronutrients like folic acid | Currently not recommended for managing AGE in pediatric patients. | (II, B) * (weak recommendation, very low-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
Gelatine Tannate | Currently not recommended for managing AGE in pediatric patients. | (III, C) * (weak recommendation, very low-quality evidence) ** | No new research available on PubMed and Scopus from 2014 to 2025. |
ORV | Can be considered for hospitalized children with RV AGE. | (III, C) * (weak recommendation, very low-quality evidence) ** | Described in Section 4.2.3. |
Study Short Methodology | Type of Study | Key Findings | Authors and the Date |
---|---|---|---|
In a randomized study of 103 pediatric patients with RV enteritis, participants received either standard therapy alone (n = 52) or standard therapy plus zinc gluconate supplementation for 10 days (n = 51). Clinical outcomes were assessed at 72 h, including symptom resolution and recovery of extra-intestinal organ involvement. A 3-month follow-up evaluated diarrhea recurrence. | Randomized Controlled Trial | Zinc supplementation significantly improved clinical outcomes in infants with RV enteritis, with a higher response rate (90% vs. 75%, p < 0.05) and shorter duration of diarrhea, fever, and vomiting compared to standard therapy alone (p < 0.05). Moreover, zinc reduced both the recurrence and severity of post-treatment diarrhea over a 3-month follow-up (p < 0.05). | Jiang et al. (2016) [91] |
A total of 85 patients with RV AGE were divided into two groups based on treatment strategy: the control group (n = 42) received probiotics alone, while the experimental group (n = 43) received a combination of probiotics with zinc and selenium. Clinical efficacy, stool frequency, and adverse event incidence were evaluated to determine therapeutic effectiveness. | Original Article | Combined probiotic, zinc, and selenium therapy significantly enhanced clinical efficacy compared to probiotics alone (88.4% vs. 50%, p < 0.05), with faster symptom resolution and greater reductions in stool frequency. Post-treatment levels of myocardial enzymes (CK, CK-MB, AST) and inflammatory markers (IL-6, IL-8, hsCRP) were markedly lower in the combination group (p < 0.05). | Cai et al. (2022) [92] |
This study included 45 pediatric patients hospitalized with RV enteritis. Patients were assigned to three groups (n = 15 each): untreated controls, conventional therapy (oral smectite), and conventional therapy plus oral zinc gluconate. Fecal samples were collected 6 h post-fasting. Treatment lasted 7 days, with age-adjusted dosing for both smectite and zinc. | Original Research Article | Zinc combined with conventional therapy improved clinical recovery in pediatric RV enteritis, shortening symptom duration and enhancing gut microbiota diversity. Beneficial genera (e.g., Faecalibacterium, Bacteroides) increased, while inflammatory markers (IL-6, TNF-α, CRP) showed negative correlations with key commensals. Zinc adjunctive therapy accelerates recovery, supports microbiota balance, and may reduce inflammation, informing clinical management strategies. | Xu et al. (2023) [93] |
Study Short Methodology | Type of Probiotics | Population | Primary Outcomes | Key Findings | Authors and the Date |
---|---|---|---|---|---|
Single-center, open-label, randomized, controlled trial included 159 patients (age range, 3 mo to 14 yo). | Bacillus mesentericus TO-A (1.1 × 107 CFU), Clostridium butyricum TO-A (2.0 × 107 CFU), Enterococcus faecalis T-110 (3.17 × 108 CFU) (BIO-THREE) 3 times daily for 7 days | 61 children (RV and Salmonella) | Duration of diarrhea, duration of fever, length of hospital stay | Probiotic therapy significantly shortened the duration of diarrhea and fever and reduced hospitalization time compared to control. Benefits were observed in both RV and salmonella infections, indicating broad therapeutic potential (p < 0.0001). | Huang et al. (2014) [94] |
An open-label, randomized, controlled trial included 200 patients with AGE, aged between 6 months and 5 years. | LGG in dose of 10 × 109 CFU/day for five days (LGG group) or no probiotic medication (control group) | 100 children in control group and 100 children in LGG group | Duration of diarrhea, number of stools per day | LGG administration in children with AGE significantly shortened duration of diarrhea compared to the control group [60 (54–72) h vs. 78 (72–90) h; p < 0.001]. The number of stools per day was significantly lower in the LGG group compared with the control group (p < 0.001). | Aggarwal S et al. (2014) [95] |
Children with AGE aged 6 months to 5 years, testing positive for either RV in stool (coinfections were excluded), were randomized to LGG (ATCC 53103) or placebo, once daily for 4 weeks. Baseline demographic and clinical details were obtained. | LGG (ATCC 53103) 1 × 1010 CFU | 140 children with acute AGE (RV positive subgroup) | Recurrence of diarrhea, intestinal function/permeability, serum IgG levels | In children with RV AGE, LGG reduced recurrent episodes (25% vs. 46%; p = 0.048) and impaired intestinal function (48% vs. 72%; p = 0.027). A significant rise in anti-RV IgG was observed (2215 vs. 456 EU; p = 0.003). | Sindhu et al. (2014) [96] |
In vitro antiviral activities of probiotic isolates on RV infection were investigated in the Vero cell using a plaque reduction assay. Then several probiotic strains with the high antiviral activity were chosen for further clinical trials. Twenty-nine pediatric patients who presented with symptoms of viral AGE were enrolled in a double-blind trial and randomly assigned at admission to receive six probiotic strains. | Bifidobacterium longum, B. lactis, Lactobacillus acidophilus, L. rhamnosus, L. plantarum, and Pediococcus pentosaceus at a dose of 109 CFU/g or a comparable placebo twice daily for 1 week. | Children with RV AGE; cell line studies | Duration of diarrhea (clinical); inhibition of viral replication (in vitro) | In vitro results confirmed inhibition of RV replication by the probiotics. Clinically, children treated with probiotics experienced a statistically significant reduction in the duration of diarrhea. | Lee et al. (2015) [97] |
Single-blind, placebo-controlled, randomized trial. | Combined product: L. casei, L. rhamnosus, S. termophilus, B. breve, L. acidophilus, L. bulgaricus, B. infantis + ORS/IV rehydration therapy ORS/IV rehydration therapy Dosage: 1 × 109 CFU/g 2×/day for 5 days | 64 children with confirmed RV AGE | Duration of diarrhea, adverse effects | LGG group had significantly shorter mean duration of diarrhea (4.1 vs. 6.2 days). Findings confirm symptomatic benefit of LGG in acute RV. | Sobouti et al., 2016 [98] |
Children (3 months to 5 years) with WHO-defined acute watery diarrhea and RV positive stool (n = 60) were randomized into intervention (n = 30) and control (n = 30) groups. The intervention group received SB. | SB (500 mg/day) for 5 days | 100 children with confirmed RV | Diarrhea duration, safety | S. boulardii significantly reduced diarrhea duration. Safe and effective adjunct to standard therapy in acute RV. | Das et al. (2016) [99] |
The children were randomly divided into the study’s two treatment groups: three days of the oral administration of a probiotics formula containing both Bifidobacterium longum BORI and Lactobacillus acidophilus AD031 or a placebo and the standard therapy for diarrhea. | Bifidobacterium longum BORI (2 × 1010 CFU/g) and Lactobacillus acidophilus AD031 (2 × 109 CFU/g) | Infants with RV infection | Diarrhea duration, viral load | When the 57 cases completed the protocol, the duration of the patients’ diarrhea was significantly shorter in the probiotics group (4.38 ± 1.29, N = 28) than the placebo group (5.61 ± 1.23, N = 29), with a p-value of 0.001. There were no serious, adverse events and no differences in the frequency of adverse events in both groups. Probiotic combination reduced both diarrhea duration and viral shedding. Clinically and virologically beneficial. | Park et al. (2017) [100] |
Double-blind, placebo-controlled randomized controlled trial. | L. acidophilus (2 daily oral doses of 2 × 108 for 5 days) + ORS + Zn + antimicrobials were needed Placebo + ORS + Zn + antimicrobials were needed. | 300 Vietnamese children with acute watery diarrhea (RV-positive subgroup) | Duration of diarrhea, treatment failure | No significant difference in duration of diarrhea between treatment and placebo groups. Subgroup analysis (RV cases) also showed no benefit. | Hong Chau et al., 2018 [101] |
Randomized, controlled clinical trial with retrospective comparison, conducted in hospitalized children with RV AGE. | Lactobacillus plantarum LRCC5310 | 50 children (15 probiotic group, 8 control, 27 retrospective control) hospitalized with confirmed RV AGE | Duration of diarrhea, number of stools, viral load in stool samples | LRCC5310 significantly reduced diarrhea frequency and duration (p = 0.033, p = 0.003, and p = 0.012, respectively), improved Vesikari scores (p = 0.076, p = 0.061, and p = 0.036, respectively), and inhibited viral replication compared with controls; no adverse effects reported. | Shin et al. (2020) [102] |
Randomized controlled trial comparing racecadotril (1.5 mg/kg, given three times daily for 7 days) alone vs. racecadotril plus Lactobacillus reuteri in children with rotavirus enteritis. | Lactobacillus reuteri (administered daily at 1 × 108 CFU for 7 days) | 85 children with confirmed RV AGE (43 control, 42 probiotic + racecadotril) | RV conversion rate, intestinal mucosal barrier function, immune response (CD4+, CD8+), intestinal microbiota composition | Combination therapy led to significantly higher RV clearance rates at days 3, 5, and 7 (p < 0.05), reaching 61.90%, 76.19%, and 92.86%, respectively, improved mucosal barrier function, increased CD4+ and beneficial microbiota, and reduced endotoxins, AGEs, D-lactic acid, and CD8+ levels compared with controls. | He et al. (2025) [103] |
Study Short Methodology | Type of Oral Immunoglobuline with Dosage | Type of Study | Key Findings | Authors and the Date |
---|---|---|---|---|
A total of 100 pediatric patients with RV enteritis divided into control and immunoglobulin treated group. All patients received fluid replacement. | A randomized trial included two groups of children aged 3 months to 3 years. The control group (n = 50) received oral placebo, while the treatment group (n = 50) received anti-RV IgY from egg yolk (1 g, three times daily for 3 days). Baseline characteristics, including age, sex, and pre-treatment diarrhea frequency, showed no significant differences between groups (p > 0.05). | Clinical trial, randomized controlled trial | Orally administered anti-RV IgY significantly reduced diarrhea duration (4.5 ± 0.92 vs. 5.8 ± 1.68 days, p = 0.015), decreased stool frequency (p < 0.05), increased fecal SIgA levels with earlier doubling (day 3 vs. day 5), and lowered viral shedding compared to placebo (p < 0.05). | Xie et al. (2015) [106] |
A total of 4 pediatric hematopoietic stem cell transplant patients with confirmed RV infection. | Patients received human immunoglobulin (Gamunex-C, 10%, Grifols Therapeutics, LLC, Research Triangle Park, NC, USA). Two with acute myeloid leukemia (ages 10 months and 5 years) and one with Shwachman–Diamond syndrome (14 years) received 20 mg/kg/dose QID. A patient with Wiskott–Aldrich syndrome (14 months) received 76 mg/kg/dose QID. | Case report | In three of four treatment episodes, stool frequency and/or consistency improved within a median of three days after starting enteral immunoglobulin. Symptom resolution was faster than in historical controls. One patient was later diagnosed with GI graft-versus-host disease after 22 months. These results indicate potential benefits of oral immunoglobulin for managing RV diarrhea in HSCT recipients. | Williams et al. (2015) [107] |
A total of 36 pediatric allogeneic HCT recipients with total 49 discrete episodes of RV infection (positive stool RV antigen assay). | Nitazoxanide and/or enterally administered immunoglobulins. One patient received 16 doses of immunoglobulin over 5 days at 0.34 g/kg/dose; all other patients received a single initial dose. | Retrospective single-center study | Median duration of diarrhea: 17.5 days (range 4–122). After initiation of treatment, the median duration of clinical symptoms are 11 days (nitazoxanide), 23 days (immunoglobulins), and 26 days (combination). No adverse effects observed. Efficacy not confirmed. | Flerlage et al., (2018) [108] |
Stage of Viral Life Cycle | Compound | Class of Inhibitor | Mechanism of Action | Type of Study |
---|---|---|---|---|
Viral entry | Cordycepin | Adenosine analog | Modulates PI3K/Akt pathway, promotes apoptosis, and suppresses viral replication. | Investigational in vitro and in vivo study (BALB/c infant mice) [119]. |
Cyclosporine | Cyclic peptide (host factor modulator) | Increases expression of type I interferons, enhancing antiviral response. | Investigational in vitro and in vivo study (BALB/c infant mice) [118]. | |
Drebin | Cytoskeleton-associated protein | Binds VP4 and its VP5 fragment, hindering viral entry into host cells. | In vitro study; also evaluated in vivo: human primary enteroids and C57BL/6 mouse model [128]. | |
Genipin | Aglycone (from geniposide) | Inhibits viral attachment and penetration (early stage) and also assembly and release (late stage). | In vitro studies [113]. | |
Genistein | Flavonoid, tyrosine kinase inhibitor | Inhibits integrin phosphorylation, reduces RV binding affinity to integrins and entry. | In vitro studies [127]. | |
Methyl-β-cyclodextrin | Pharmacological cholesterol-sequestering agent | Disrupts cholesterol rafts and blocks RV receptor-mediated endocytosis. | In vitro studies [124,125]. | |
Viral entry, Genome replication | ML241 | Small-molecule MAPK pathway inhibitor | Inhibits ERK1/2 phosphorylation and IκBα/NF-κB signaling, suppressing viral entry, replication, and cytotoxicity. | In vitro and in vivo study using BALB/c infant mice [121]. |
Genome replication | 6-Thioguanine (6-TG) | Thio-analog of guanine | GTP-Rac1 inhibitor | In vitro study; tested in vivo: human enteroids; 6-TG has been clinically used since the 1950s as an anticancer and immunosuppressive agent [22]. |
Brequinar | Quinolinecarboxylic acid | Inhibits pyrimidine synthesis via DHODH inhibition. | In vitro studies [111]. | |
Leflunomide | Isoxazole derivative | Inhibits pyrimidine synthesis via DHODH inhibition. | In vitro studies [111]. | |
Metformin hydrochloride | Biguanide (metabolic regulator) | Reduces viral mRNA levels and RV replication in Caco-2 cells and intestinal organoids. | In vitro and in vivo study using BALB/c infant mice [123]. | |
MiR-525-3p | Cellular microRNA | Binds the 3′ UTR of RV NSP1, enhancing interferon and cytokine levels. | In vitro studies [130]. | |
miRNA-7 | Cellular microRNA | Targets RV gene segment 11 encoding NSP5, disrupting viroplasm formation. | In vitro and in vivo study using BALB/c infant mice [131]. | |
POL-P (Portulaca oleracea L.) | Polysaccharide (plant-derived immunomodulator) | Upregulates IFN-α expression, suppressing viral replication. | In vitro studies [112]. | |
Formation of Viroplasm | Deoxyshikonin | Naphthoquinone derivative | Activates SIRT1, ac-FoxO1, and Rab7; lowers VP6 expression and viral titers; induces autophagy and oxidative stress. | In vitro studies [122]. |
MG132, bortezomib, and lactacystin | Tripeptide aldehyde, dipeptidylboronate, and antibiotic, respectively | Proteasome inhibitors; disrupt VP formation and alter VP1 localization. | In vitro studies [134,135]. | |
Molnupiravir | Cytidine nucleoside analog | Inhibits viroplasm formation without altering structure. | In vitro studies [117]. | |
Nitazoxanide | Thiazolide | Inhibits VP7 maturation, disrupts viroplasm assembly, and impairs viral morphogenesis. | Clinical trial for RV treatment; clinically approved anti-infective treatment [115]. | |
TOFA, triacsin C, C75, A922500, betulinic acid, CI-976, PHB, and isoproterenol/IBMX | Pharmacological enzyme inhibitors of the lipid metabolism pathway | Modulates lipid droplet biogenesis and degradation. | In vitro studies [133]. | |
Ursolic acid | Triterpenoid | Reduces lipid droplet availability, limiting VP formation. | In vitro studies [116]. | |
Infectious particle genesis | 18β-Glycyrrhetinic acid (18βGRA) | Aglycone (PI3K/Akt pathway; antiviral activity) | Inhibits viral replication by modulating apoptosis and signaling pathways. | In vitro studies, tested in a mouse model (C57BL/6 male mice) [120]. |
RA839 | Small molecule | Activates the Nrf2/ARE pathway, enhancing the cellular redox response. | In vitro studies [126]. | |
Rapamycin, LY294002, and BEZ235 | Macrolide, morpholine-containing chemical compound, and imidazoquinoline derivative, respectively | mTOR and PI3K inhibitors; activate the autophagy cascade. | In vitro study; tested in vivo: human enteroids; rapamycin is FDA-approved for transplant recipients [25]. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Pawłuszkiewicz, K.; Kucharczyk, E.; Korgiel, M.; Busłowicz, T.; Faltus, A.; Kucharczyk, N.; Paluch, E. Advances in the Diagnosis and Treatment of Rotavirus Infections: Narrative Review. Int. J. Mol. Sci. 2025, 26, 9175. https://doi.org/10.3390/ijms26189175
Pawłuszkiewicz K, Kucharczyk E, Korgiel M, Busłowicz T, Faltus A, Kucharczyk N, Paluch E. Advances in the Diagnosis and Treatment of Rotavirus Infections: Narrative Review. International Journal of Molecular Sciences. 2025; 26(18):9175. https://doi.org/10.3390/ijms26189175
Chicago/Turabian StylePawłuszkiewicz, Karolina, Emilia Kucharczyk, Matylda Korgiel, Tomasz Busłowicz, Anita Faltus, Natalia Kucharczyk, and Emil Paluch. 2025. "Advances in the Diagnosis and Treatment of Rotavirus Infections: Narrative Review" International Journal of Molecular Sciences 26, no. 18: 9175. https://doi.org/10.3390/ijms26189175
APA StylePawłuszkiewicz, K., Kucharczyk, E., Korgiel, M., Busłowicz, T., Faltus, A., Kucharczyk, N., & Paluch, E. (2025). Advances in the Diagnosis and Treatment of Rotavirus Infections: Narrative Review. International Journal of Molecular Sciences, 26(18), 9175. https://doi.org/10.3390/ijms26189175