Washed microbiota transplantation vs. manual fecal microbiota transplantation: clinical findings, animal studies and in vitro screening

Ting Zhang, Gaochen Lu, Zhe Zhao, Yafei Liu, Quan Shen, Pan Li, Yaoyao Chen, Haoran Yin, Huiquan Wang, Cicilia Marcella, Bota Cui, Lei Cheng, Guozhong Ji, Faming Zhang, Ting Zhang, Gaochen Lu, Zhe Zhao, Yafei Liu, Quan Shen, Pan Li, Yaoyao Chen, Haoran Yin, Huiquan Wang, Cicilia Marcella, Bota Cui, Lei Cheng, Guozhong Ji, Faming Zhang

Abstract

Fecal microbiota transplantation (FMT) by manual preparation has been applied to treat diseases for thousands of years. However, this method still endures safety risks and challenges the psychological endurance and acceptance of doctors, patients and donors. Population evidence showed the washed microbiota preparation with microfiltration based on an automatic purification system followed by repeated centrifugation plus suspension for three times significantly reduced FMT-related adverse events. This washing preparation makes delivering a precise dose of the enriched microbiota feasible, instead of using the weight of stool. Intraperitoneal injection in mice with the fecal microbiota supernatant obtained after repeated centrifugation plus suspension for three times induced less toxic reaction than that by the first centrifugation following the microfiltration. The toxic reactions that include death, the change in the level of peripheral white blood cells, and the proliferation of germinal center in secondary lymphoid follicles in spleen were noted. The metagenomic next-generation sequencing (NGS) indicated the increasing types and amount of viruses could be washed out during the washing process. Metabolomics analysis indicated metabolites with pro-inflammatory effects in the fecal microbiota supernatant such as leukotriene B4, corticosterone, and prostaglandin G2 could be removed by repeated washing. Near-infrared absorption spectroscopy could be served as a rapid detection method to control the quality of the washing-process. In conclusion, this study for the first time provides evidence linking clinical findings and animal experiments to support that washed microbiota transplantation (WMT) is safer, more precise and more quality-controllable than the crude FMT by manual.

Keywords: adverse event; fecal microbiota transplantation; infection; metabolomics; safety; spectroscopy; transplant; virus; washed microbiota transplantation.

Conflict of interest statement

Faming Zhang invented the concept of GenFMTer, TET and the related devices. Ting Zhang, Gaochen Lu, Zhe Zhao, Yafei Liu, Quan Shen, Pan Li, Yaoyao Chen, Haoran Yin, Huiquan Wang, Cicilia Marcella, Bota Cui, Lei Cheng, and Guozhong Ji declare that they have no competing interests. All procedures followed were in accordance with the ethical standards of the institutional review board of the Second Affiliated Hospital of Nanjing Medical University and with the Helsinki Declaration of 1975, as revised in 2000. Informed consent was obtained from all patients for being included in the study. All institutional and national guidelines for the care and use of laboratory animals were followed.

Figures

Figure 1
Figure 1
Flow chart of the study
Figure 2
Figure 2
Evidence related to fecal microbiota preparation from human population to animal. (A–C) AEs related to manual and automatic preparation for fecal microbiota in patients. (D–F) Relationship between fecal weight and enriched microbiota from FMT donors. (H) Death time and rate after intraperitoneal injection of fecal supernatant in mice (i.p., intraperitoneal injection). Statistical comparisons are performed using chi-square test; *P < 0.05, **P < 0.01, ***P < 0.001. Correlation analysis was performed using Spearman correlation analysis. Data are presented as mean ± standard deviation (SD)
Figure 3
Figure 3
Changes of peripheral blood cells in the five groups of mice after 6 h of intraperitoneal injection of fecal microbiota supernatant. (A) Changes of WBC (n = 8 animals/group). (B) Changes of RBC (n = 8 animals/group). (C) Changes of PLT (n = 8 animals/group). Statistical comparisons are performed using one-way ANOVA; *P < 0.05, **P < 0.01, ***P < 0.001. Data are presented as mean ± SD
Figure 4
Figure 4
Changes of peripheral blood cells of mice after intraperitoneal injection of fecal microbiota supernatant. (A) Changes of WBC in the five groups of mice after 24 h (Supernatant 1, n = 3; other groups, n = 8). (B) Changes of RBC in the five groups of mice after 24 h (Supernatant 1, n = 3; other groups, n = 8). (C) Changes of PLT in the five groups of mice after 24 h (Supernatant 1, n = 3; other groups, n = 8). (D) Changes of WBC at 6 h and 24 h after injection with Supernatant 1 (Supernatant 1 6 h, n = 8; Supernatant 1 24 h, n = 3). (E) Changes in the percentage of NEUT at 6 h and 24 h after injection with Supernatant 1 (Supernatant 1 6 h, n = 8; Supernatant 1 24 h, n = 3). (F) Changes of WBC at 6 h and 24 h after injection with Supernatant 3 (n = 8/group). (H) Changes in the percentage of NEUT at 6 h and 24 h after injection with Supernatant 3 (n = 8/group). Statistical comparisons (A–C) are performed using one-way ANOVA; statistical comparisons (D–H) are performed using unpaired t-tests; *P < 0.05, **P < 0.01, ***P < 0.001. Data are presented as mean ± SD
Figure 5
Figure 5
HE staining of spleen in mice at 6 h after intraperitoneal injection with the fecal microbiota supernatant. (A) No proliferation of primary follicles was observed in the group of normal saline (×200). (B) The proliferation of secondary follicles was observed in the group of Supernatant 1 (×200). (C) No proliferation of primary follicles was observed in the group of Supernatant 3 (×200). (D) NEUT infiltration in the group of normal saline (×400). (E) NEUT infiltration in the group of Supernatant 1 (×400). (F) NEUT infiltration in the group of Supernatant 3 (×400) . White arrow noted: germinal center of secondary follicles; yellow arrow noted: NEUT infiltration
Figure 6
Figure 6
Differential screening of virus changing during the washing process. (A) Changes of virus types. (B) Changes of metagenomic NGS reads number. (C and D) Changes of the same virus in the washed fecal supernatant. (E) Top ten viruses with the most significant fold change
Figure 7
Figure 7
Top five viruses in each donor
Figure 8
Figure 8
Differential screening of metabolomics during the washing process. Group 1: Supernatant 1; Group 2: Supernatant 3. (A) A score map of the PCA model. (B) Cluster analysis of differential metabolites. Each row represented a differential metabolite with each column representing a sample, and the color from green to red corresponding expression from low to high. (C) A score map of the PLS-DA model. (D) Volcanic map of differential metabolites. The down-regulated differential metabolites were labeled green, the up-regulated ones were marked in red, and the undifferentiated metabolites were labeled as purple-grey. (E) The response ranking check chart of the PLS-DA model. (F) Bubble map of metabolic pathway enrichment analysis. The X-axis enrichment factor (RichFactor) is the value of differential metabolites annotated to the pathway divided by all identified metabolites annotated to the pathway. The larger value indicated the greater proportion of differential metabolites annotated in the pathway. The dot size represented the number of differential metabolites annotated to the pathway
Figure 9
Figure 9
Differential testing by near-infrared absorption (NIRS) spectroscopy. (A) Incident surface of Supernatant 1, 2, 3, 4, 5 and normal saline. (B) Exit surface of Supernatant 1, 2, 3, 4, 5 and normal saline. (C) The light intensity of fecal microbiota supernatant by near-infrared absorption spectroscopy. (D) The absorbance of fecal microbiota supernatant by near-infrared absorption spectroscopy. (E) Euclidean distance between the spectrum of the supernatant of different washing times and the spectrum of the normal saline. (F) Correlation coefficient between the spectrum of the supernatant of different washing times and the spectrum of the normal saline

References

    1. Aboul-Ata AA, Vitti A, Nuzzaci M, El-Attar AK, Piazzolla G, Tortorella C, Harandi AM, Olson O, Wright SA, Piazzolla P. Plant-based vaccines: novel and low-cost possible route for Mediterranean innovative vaccination strategies. Adv Virus Res. 2014;89:1–37.
    1. Afarideh M, Behdadnia A, Noshad S, Mirmiranpour H, Mousavizadeh M, Khajeh E, Rad MV, Mazaheri T, Nakhjavani M, Esteghamati A. Association of peripheral 5-hydroxyindole-3-acetic acid, a serotonin derivative, with metabolic syndrome and low-grade inflammation. Endocr Pract. 2015;21:711–718.
    1. Alhouayek M, Muccioli GG. COX-2-derived endocannabinoid metabolites as novel inflammatory mediators. Trends Pharmacol Sci. 2014;35:284–292.
    1. Alhouayek M, Masquelier J, Cani PD, Lambert DM, Muccioli GG. Implication of the anti-inflammatory bioactive lipid prostaglandin D2-glycerol ester in the control of macrophage activation and inflammation by ABHD6. Proc Natl Acad Sci U S A. 2013;110:17558–17563.
    1. Allegretti JR, Mullish BH, Kelly C, Fischer M. The evolution of the use of faecal microbiota transplantation and emerging therapeutic indications. Lancet. 2019;394:420–431.
    1. Austenaa LM, Carlsen H, Hollung K, Blomhoff HK, Blomhoff R. Retinoic acid dampens LPS-induced NF-kappaB activity: results from human monoblasts and in vivo imaging of NF-kappaB reporter mice. J Nutr Biochem. 2009;20:726–734.
    1. Bajaj JS, Kassam Z, Fagan A, Gavis EA, Liu E, Cox IJ, Kheradman R, Heuman D, Wang J, Gurry T, et al. Fecal microbiota transplant from a rational stool donor improves hepatic encephalopathy: a randomized clinical trial. Hepatology. 2017;66:1727–1738.
    1. Bigagli E, Cinci L, Paccosi S, Parenti A, D’Ambrosio M, Luceri C. Nutritionally relevant concentrations of resveratrol and hydroxytyrosol mitigate oxidative burst of human granulocytes and monocytes and the production of pro-inflammatory mediators in LPS-stimulated RAW 264.7 macrophages. Int Immunopharmacol. 2017;43:147–155.
    1. Boltana S, Sanhueza N, Donoso A, Aguilar A, Crespo D, Vergara D, Arriagada G, Morales-Lange B, Mercado L, Rey S, et al. The expression of TRPV channels, prostaglandin E2 and pro-inflammatory cytokines during behavioural fever in fish. Brain Behav Immun. 2018;71:169–181.
    1. Brandt LJ. Editorial commentary: fecal microbiota transplantation: patient and physician attitudes. Clin Infect Dis. 2012;55:1659–1660.
    1. Cai J, Peng Y, Chen T, Liao H, Zhang L, Chen Q, He Y, Wu P, Xie T, Pan Q. Chloral hydrate treatment induced apoptosis of macrophages via Fas signaling pathway. Med Sci Monit. 2016;22:4836–4843.
    1. Cammarota G, Ianiro G, Tilg H, Rajilic-Stojanovic M, Kump P, Satokari R, Sokol H, Arkkila P, Pintus C, Hart A, et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut. 2017;66:569–580.
    1. Cammarota G, Ianiro G, Kelly CR, Mullish BH, Allegretti JR, Kassam Z, Putignani L, Fischer M, Keller JJ, Costello SP, et al. International consensus conference on stool banking for faecal microbiota transplantation in clinical practice. Gut. 2019;68(12):2111–2121.
    1. Costello SP, Tucker EC, La Brooy J, Schoeman MN, Andrews JM. Establishing a fecal microbiota transplant service for the treatment of Clostridium difficile infection. Clin Infect Dis. 2016;62:908–914.
    1. Costello SP, Hughes PA, Waters O, Bryant RV, Vincent AD, Blatchford P, Katsikeros R, Makanyanga J, Campaniello MA, Mavrangelos C, et al. Effect of fecal microbiota transplantation on 8-week remission in patients with ulcerative colitis: a randomized clinical trial. JAMA. 2019;321:156–164.
    1. Cui B, Li P, Xu L, Zhao Y, Wang H, Peng Z, Xu H, Xiang J, He Z, Zhang T, et al. Step-up fecal microbiota transplantation strategy: a pilot study for steroid-dependent ulcerative colitis. J Transl Med. 2015;13:298.
    1. Cui B, Li P, Xu L, Peng Z, Xiang J, He Z, Zhang T, Ji G, Nie Y, Wu K, et al. Step-up fecal microbiota transplantation (FMT) strategy. Gut Microbes. 2016;7:323–328.
    1. DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, Turbett S, Chung RT, Chen YB, Hohmann EL. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381:2043–2050.
    1. Ding X, Li Q, Li P, Zhang T, Cui B, Ji G, Lu X, Zhang F. Long-term safety and efficacy of fecal microbiota transplant in active ulcerative colitis. Drug Saf. 2019;42:869–880.
    1. He Z, Li P, Zhu J, Cui B, Xu L, Xiang J, Zhang T, Long C, Huang G, Ji G, et al. Multiple fresh fecal microbiota transplants induces and maintains clinical remission in Crohn’s disease complicated with inflammatory mass. Sci Rep. 2017;7:4753.
    1. Huang HL, Chen HT, Luo QL, Xu HM, He J, Li YQ, Zhou YL, Yao F, Nie YQ, Zhou YJ. Relief of irritable bowel syndrome by fecal microbiota transplantation is associated with changes in diversity and composition of the gut microbiota. J Dig Dis. 2019;20:401–408.
    1. Jilma-Stohlawetz P, Kliegel T, Kantner-Schlifke I, Strasser-Marsik C, Mayr FB, Jilma B. Upregulation of cytokine mRNA in circulating leukocytes during human endotoxemia. Eur Cytokine Netw. 2017;28:19–26.
    1. Kang DW, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, Khoruts A, Geis E, Maldonado J, McDonough-Means S, et al. Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017;5:10.
    1. Kim YS, Park ZY, Kim SY, Jeong E, Lee JY. Alteration of Toll-like receptor 4 activation by 4-hydroxy-2-nonenal mediated by the suppression of receptor homodimerization. Chem Biol Interact. 2009;182:59–66.
    1. Kim SR, Jung YR, Kim DH, An HJ, Kim MK, Kim ND, Chung HY. Caffeic acid regulates LPS-induced NF-kappaB activation through NIK/IKK and c-Src/ERK signaling pathways in endothelial cells. Arch Pharm Res. 2014;37:539–547.
    1. Konig J, Siebenhaar A, Hogenauer C, Arkkila P, Nieuwdorp M, Noren T, Ponsioen CY, Rosien U, Rossen NG, Satokari R, et al. Consensus report: faecal microbiota transfer—clinical applications and procedures. Aliment Pharmacol Ther. 2017;45:222–239.
    1. Kurhaluk N, Zaitseva OV, Sliuta A, Kyriienko S, Winklewski PJ. Melatonin diminishes oxidative stress in plasma, retains erythrocyte resistance and restores white blood cell count after low dose lipopolysaccharide exposure in mice. Gen Physiol Biophys. 2018;37:571–580.
    1. Kwon SY, Ro M, Kim JH. Mediatory roles of leukotriene B4 receptors in LPS-induced endotoxic shock. Sci Rep. 2019;9:5936.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. Lin L, Chen X, Zhou Q, Huang P, Jiang S, Wang H, Deng Y. Synaptic structure and alterations in the hippocampus in neonatal rats exposed to lipopolysaccharide. Neurosci Lett. 2019;709:134364.
    1. Liu Y, Fang S, Li X, Feng J, Du J, Guo L, Su Y, Zhou J, Ding G, Bai Y, et al. Aspirin inhibits LPS-induced macrophage activation via the NF-kappaB pathway. Sci Rep. 2017;7:11549.
    1. Lu J, Huang G, Wang Z, Zhuang S, Xu L, Song B, Xiong Y, Guan S. Tyrosol exhibits negative regulatory effects on LPS response and endotoxemia. Food Chem Toxicol. 2013;62:172–178.
    1. Ma Y, Yang J, Cui B, Xu H, Xiao C, Zhang F. How Chinese clinicians face ethical and social challenges in fecal microbiota transplantation: a questionnaire study. BMC Med Ethics. 2017;18:39.
    1. Mattila E, Uusitalo-Seppala R, Wuorela M, Lehtola L, Nurmi H, Ristikankare M, Moilanen V, Salminen K, Seppala M, Mattila PS, et al. Fecal transplantation, through colonoscopy, is effective therapy for recurrent Clostridium difficile infection. Gastroenterology. 2012;142:490–496.
    1. McDonald LC, Gerding DN, Johnson S, Bakken JS, Carroll KC, Coffin SE, Dubberke ER, Garey KW, Gould CV, Kelly C, et al. Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA) Clin Infect Dis. 2018;66:987–994.
    1. McSweeney B, Allegretti JR, Fischer M, Xu H, Goodman KJ, Monaghan T, McLeod C, Mullish BH, Petrof EO, Phelps EL, et al. In search of stool donors: a multicenter study of prior knowledge, perceptions, motivators, and deterrents among potential donors for fecal microbiota transplantation. Gut Microbes. 2019 doi: 10.1080/19490976.2019.1611153.
    1. Metsola J, Maksimow M, Ojaniemi M, Metsola H, Marttila-Ichihara F, Vuolteenaho R, Yegutkin GG, Salmi M, Hallman M, Jalkanen S. Postnatal development and LPS responsiveness of pulmonary adenosine receptor expression and of adenosine-metabolizing enzymes in mice. Pediatr Res. 2014;76:515–521.
    1. Miller S, Naccache SN, Samayoa E, Messacar K, Arevalo S, Federman S, Stryke D, Pham E, Fung B, Bolosky WJ, et al. Laboratory validation of a clinical metagenomic sequencing assay for pathogen detection in cerebrospinal fluid. Genome Res. 2019;29:831–842.
    1. Moayyedi P, Surette MG, Kim PT, Libertucci J, Wolfe M, Onischi C, Armstrong D, Marshall JK, Kassam Z, Reinisch W, et al. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology. 2015;149(102–109):e106.
    1. Ng SC, Kamm MA, Yeoh YK, Chan PKS, Zuo T, Tang W, Sood A, Andoh A, Ohmiya N, Zhou Y, et al. Scientific frontiers in faecal microbiota transplantation: joint document of Asia-Pacific Association of Gastroenterology (APAGE) and Asia-Pacific Society for Digestive Endoscopy (APSDE) Gut. 2019;69(1):83–91.
    1. Nilius B, Owsianik G. The transient receptor potential family of ion channels. Genome Biol. 2011;12:218.
    1. Paramsothy S, Kamm MA, Kaakoush NO, Walsh AJ, van den Bogaerde J, Samuel D, Leong RWL, Connor S, Ng W, Paramsothy R, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet. 2017;389:1218–1228.
    1. Park L, Mone A, Price JC, Tzimas D, Hirsh J, Poles MA, Malter L, Chen LA. Perceptions of fecal microbiota transplantation for Clostridium difficile infection: factors that predict acceptance. Ann Gastroenterol. 2017;30:83–88.
    1. Pilgrim MJ, Kasman L, Grewal J, Bruorton ME, Werner P, London L, London SD. A focused salivary gland infection with attenuated MCMV: an animal model with prevention of pathology associated with systemic MCMV infection. Exp Mol Pathol. 2007;82:269–279.
    1. Qi X, Li X, Zhao Y, Wu X, Chen F, Ma X, Zhang F, Wu D. Treating steroid refractory intestinal acute graft-vs.-host disease with fecal microbiota transplantation: a pilot study. Front Immunol. 2018;9:2195.
    1. Satokari R, Mattila E, Kainulainen V, Arkkila PE. Simple faecal preparation and efficacy of frozen inoculum in faecal microbiota transplantation for recurrent Clostridium difficile infection–an observational cohort study. Aliment Pharmacol Ther. 2015;41:46–53.
    1. Sood A, Mahajan R, Juyal G, Midha V, Grewal CS, Mehta V, Singh A, Joshi MC, Narang V, Kaur K, et al. Efficacy of fecal microbiota therapy in steroid dependent ulcerative colitis: a real world intention-to-treat analysis. Intest Res. 2019;17:78–86.
    1. Suffredini AF, Noveck RJ. Human endotoxin administration as an experimental model in drug development. Clin Pharmacol Ther. 2014;96:418–422.
    1. Sung J, Jeon H, Kim IH, Jeong HS, Lee J. Anti-inflammatory effects of stearidonic acid mediated by suppression of NF-kappaB and MAP-kinase pathways in macrophages. Lipids. 2017;52:781–787.
    1. Symonds EM, Nguyen KH, Harwood VJ, Breitbart M. Pepper mild mottle virus: a plant pathogen with a greater purpose in (waste)water treatment development and public health management. Water Res. 2018;144:1–12.
    1. Tan ZX, Chen YH, Xu S, Qin HY, Wang H, Zhang C, Xu DX, Zhao H. Calcitriol inhibits tumor necrosis factor alpha and macrophage inflammatory protein-2 during lipopolysaccharide-induced acute lung injury in mice. Steroids. 2016;112:81–87.
    1. Vuong LD, Nguyen QN, Truong VL. Anti-inflammatory and anti-oxidant effects of combination between sulforaphane and acetaminophen in LPS-stimulated RAW 264.7 macrophage cells. Immunopharmacol Immunotoxicol. 2019;41:413–419.
    1. Wang CH, Rong MY, Wang L, Ren Z, Chen LN, Jia JF, Li XY, Wu ZB, Chen ZN, Zhu P. CD147 up-regulates calcium-induced chemotaxis, adhesion ability and invasiveness of human neutrophils via a TRPM-7-mediated mechanism. Rheumatology (Oxford) 2014;53:2288–2296.
    1. Wang H, Cui B, Li Q, Ding X, Li P, Zhang T, Yang X, Ji G, Zhang F. The safety of fecal microbiota transplantation for Crohn’s disease: findings from a long-term study. Adv Ther. 2018;35:1935–1944.
    1. Wang JW, Wang YK, Zhang F, Su YC, Wang JY, Wu DC, Hsu WH. Initial experience of fecal microbiota transplantation in gastrointestinal disease: a case series. Kaohsiung J Med Sci. 2019;35:566–571.
    1. Wu X, Dai M, Buch H, Bai J, Long W, Long C, Tang X, Tu H, Zhang R, Zhu C, et al. The recognition and attitudes of postgraduate medical students toward fecal microbiota transplantation: a questionnaire study. Therap Adv Gastroenterol. 2019;12:1756284819869144.
    1. Yates DT, Loest CA, Ross TT, Hallford DM, Carter BH, Limesand SW. Effects of bacterial lipopolysaccharide injection on white blood cell counts, hematological variables, and serum glucose, insulin, and cortisol concentrations in ewes fed low- or high-protein diets. J Anim Sci. 2011;89:4286–4293.
    1. Zhang F, Luo W, Shi Y, Fan Z, Ji G. Should we standardize the 1,700-year-old fecal microbiota transplantation? Am J Gastroenterol. 2012;107:1755–1756.
    1. Zhang F, Cui B, He X, Nie Y, Wu K, Fan D, FMT-standardization Study Group Microbiota transplantation: concept, methodology and strategy for its modernization. Protein Cell. 2018;9:462–473.
    1. Zhang F, Zhang T, Zhu H, Borody TJ. Evolution of fecal microbiota transplantation in methodology and ethical issues. Curr Opin Pharmacol. 2019;49:11–16.
    1. Zipursky JS, Sidorsky TI, Freedman CA, Sidorsky MN, Kirkland KB. Patient attitudes toward the use of fecal microbiota transplantation in the treatment of recurrent Clostridium difficile infection. Clin Infect Dis. 2012;55:1652–1658.
    1. Zipursky JS, Sidorsky TI, Freedman CA, Sidorsky MN, Kirkland KB. Physician attitudes toward the use of fecal microbiota transplantation for the treatment of recurrent Clostridium difficile infection. Can J Gastroenterol Hepatol. 2014;28:319–324.

Source: PubMed

Подписаться