Journal Home > Volume 1 , Issue 1

Due to the limitations of existing approaches, a rapid, sensitive, accurate, comprehensive, and generally applicable strategy to diagnose and treat bacterial and fungal infections remains a major challenge. Here, based on the ramanome technology platform, we propose a culture‐free, one cell resolution, phenome‐genome‐combined strategy called single‐cell identification, viability and vitality tests and source tracking (SCIVVS). For each cell directly extracted from a clinical specimen, the fingerprint region of the D2O‐probed single cell Raman spectrum (SCRS) enables species‐level identification based on a reference SCRS database of pathogen species, whereas the C‐D band accurately quantifies viability, metabolic vitality, phenotypic susceptibility to antimicrobials, and their intercellular heterogeneity. Moreover, to source track a cell, Raman‐activated cell sorting followed by sequencing or cultivation proceeds, producinging an indexed, high coverage genome assembly or a pure culture from precisely one pathogenic cell. Finally, an integrated SCIVVS workflow that features automated profiling and sorting of metabolic and morphological phenomes can complete the entire process in only a few hours. Because it resolves heterogeneity for both the metabolic phenome and genome, targets functions, can be automated, and is orders‐of‐magnitude faster while cost‐effective, SCIVVS is a new technological and data framework to diagnose and treat bacterial and fungal infections in various clinical and disease control settings.


menu
Abstract
Full text
Outline
About this article

Single cell metabolic phenome and genome via the ramanome technology platform: Precision medicine of infectious diseases at the ultimate precision?

Show Author's information Jian Xu1 ( )Jianzhong Zhang2Yingchun Xu3Yi‐Wei Tang4Bo Ma1,5Yuzhang Wu6,( )
Single‐Cell Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, China
State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
Medical Affairs, Danaher Corporation/Cepheid, Shanghai, China
Qingdao Single‐Cell Biotech. Co., Ltd., Qingdao, Shandong, China
Chongqing International Institute of Immunology, Chongqing, Sichuan, China

[Corrections added on 24 May 2023, after first online publication: 1. Expansion of CAST‐R has been corrected in the Abbreviations section. 2. Microbial dingle‐cell has been corrected as Microbial single‐cell in the figure 1 caption.]

Abstract

Due to the limitations of existing approaches, a rapid, sensitive, accurate, comprehensive, and generally applicable strategy to diagnose and treat bacterial and fungal infections remains a major challenge. Here, based on the ramanome technology platform, we propose a culture‐free, one cell resolution, phenome‐genome‐combined strategy called single‐cell identification, viability and vitality tests and source tracking (SCIVVS). For each cell directly extracted from a clinical specimen, the fingerprint region of the D2O‐probed single cell Raman spectrum (SCRS) enables species‐level identification based on a reference SCRS database of pathogen species, whereas the C‐D band accurately quantifies viability, metabolic vitality, phenotypic susceptibility to antimicrobials, and their intercellular heterogeneity. Moreover, to source track a cell, Raman‐activated cell sorting followed by sequencing or cultivation proceeds, producinging an indexed, high coverage genome assembly or a pure culture from precisely one pathogenic cell. Finally, an integrated SCIVVS workflow that features automated profiling and sorting of metabolic and morphological phenomes can complete the entire process in only a few hours. Because it resolves heterogeneity for both the metabolic phenome and genome, targets functions, can be automated, and is orders‐of‐magnitude faster while cost‐effective, SCIVVS is a new technological and data framework to diagnose and treat bacterial and fungal infections in various clinical and disease control settings.

Keywords: diagnosis of microbial infections, phenome, ramanome, Raman‐activated cell sorting (RACS), single cell sequencing

References(53)

[1]

O’Neill J. Tackling drug‐resistant infections globally: final report and recommendations. Review on antimicrobial resistance. Wellcome trust and HM government; 2016. https://amr‐review.org/sites/default/files/160525_Final%20paper_with%20cover.pdf

[2]

Chen T, Wu Q, Li S, Xiong S, Jiang S, Tan Q, et al. Microbiological quality and characteristics of probiotic products in China. J Sci Food Agric. 2014;94(1):131–8. https://doi.org/10.1002/jsfa.6221

[3]

Warzée J.‐P, Elli M, Fall A, Cattivelli D, François J.‐Y. Supranational assessment of the quality of probiotics: collaborative initiative between independent accredited testing laboratories. Microorganisms. 2021;9(7):1456. https://doi.org/10.3390/microorganisms9071456

[4]

Wilkinson MG. Flow cytometry as a potential method of measuring bacterial viability in probiotic products: a review. Trends Food Sci Technol. 2018;78:1–10. https://doi.org/10.1016/j.tifs.2018.05.006

[5]

Davis C. Enumeration of probiotic strains: review of culture‐dependent and alternative techniques to quantify viable bacteria. J Microbiol Methods. 2014;103:9–17. https://doi.org/10.1016/j.mimet.2014.04.012

[6]

Masco L, Huys G, De Brandt E, Temmerman R, Swings J. Culture‐dependent and culture‐independent qualitative analysis of probiotic products claimed to contain bifidobacteria. Int J Food Microbiol. 2005;102(2):221–30. https://doi.org/10.1016/j.ijfoodmicro.2004.11.018

[7]

Rault A, Béal C, Ghorbal S, Ogier J.‐C, Bouix M. Multiparametric flow cytometry allows rapid assessment and comparison of lactic acid bacteria viability after freezing and during frozen storage. Cryobiology. 2007;55(1):35–43. https://doi.org/10.1016/j.cryobiol.2007.04.005

[8]

Martinon A, Cronin UP, Quealy J, Stapleton A, Wilkinson MG. Swab sample preparation and viable real‐time PCR methodologies for the recovery of Escherichia coli, Staphylococcus aureus or Listeria monocytogenes from artificially contaminated food processing surfaces. Food Control. 2012;24(1):86–94. https://doi.org/10.1016/j.foodcont.2011.09.007

[9]

Guevara B, Cogdill AG. Helicobacter pylori: a review of current diagnostic and management strategies. Dig Dis Sci. 2020;65(7):1917–31. https://doi.org/10.1007/s10620‐020‐06193‐7

[10]

Tshibangu‐Kabamba E, Yamaoka Y. Helicobacter pylori infection and antibiotic resistance—from biology to clinical implications. Nat Rev Gastroenterol Hepatol. 2021;18(9):613–29. https://doi.org/10.1038/s41575‐021‐00449‐x

[11]

Mégraud F, Lehours P. Helicobacter pylori detection and antimicrobial susceptibility testing. Clin Microbiol Rev. 2007;20(2):280–322. https://doi.org/10.1128/CMR.00033‐06

[12]

Rüssmann H, Adler K, Haas R, Gebert B, Koletzko S, Heesemann J. Rapid and accurate determination of genotypic clarithromycin resistance in cultured Helicobacter pylori by fluorescent in situ hybridization. J Clin Microbiol. 2001;39(11):4142–4. https://doi.org/10.1128/JCM.39.11.4142‐4144.2001

[13]

Zullo A, Hassan C, Lorenzetti R, Winn S, Morini S. A clinical practice viewpoint: to culture or not to culture Helicobacter pylori? Dig Liver Dis. 2003;35(5):357–61. https://doi.org/10.1016/S1590‐8658(03)00081‐1

[14]

Shao Y, Lu R, Yang Y, Xu Q, Wang B, Ye G. Antibiotic resistance of Helicobacter pylori to 16 antibiotics in clinical patients. J Clin Lab Anal. 2018;32(4):e22339. https://doi.org/10.1002/jcla.22339

[15]

Pohl D, Keller PM, Bordier V, Wagner K. Review of current diagnostic methods and advances in Helicobacter pylori diagnostics in the era of next generation sequencing. World J Gastroenterol. 2019;25(32):4629–60. https://doi.org/10.3748/wjg.v25.i32.4629

[16]

Lauener FN, Imkamp F, Lehours P, Buissonnière A, Benejat L, Zbinden R, et al. Genetic determinants and prediction of antibiotic resistance phenotypes in Helicobacter pylori. J Clin Med. 2019;8(1):53. https://doi.org/10.3390/jcm8010053

[17]

Tuan VP, Narith D, Tshibangu‐Kabamba E, Dung HDQ, Viet PT, Sokomoth S, et al. A next‐generation sequencing‐based approach to identify genetic determinants of antibiotic resistance in Cambodian Helicobacter pylori clinical isolates. J Clin Med. 2019;8(6):858. https://doi.org/10.3390/jcm8060858

[18]

Chen D, Cunningham SA, Cole NC, Kohner PC, Mandrekar JN, Patel R. Phenotypic and molecular antimicrobial susceptibility of Helicobacter pylori. Antimicrob Agents Chemother. 2017;61(4):e02530–16. https://doi.org/10.1128/AAC.02530‐16

[19]

Qureshi NNG, Schiller B, Neal L. Evolution of amoxicillin resistance of Helicobacter pylori in vitro: characterization of resistance mechanisms. Microb Drug Resist. 2014;20(6):509–16. https://doi.org/10.1089/mdr.2014.0019

[20]

Binh TT, Suzuki R, Trang TTH, Kwon DH, Yamaoka Y. Search for novel candidate mutations for metronidazole resistance in Helicobacter pylori using Next‐Generation Sequencing. Antimicrob Agents Chemother. 2015;59(4):2343–8. https://doi.org/10.1128/AAC.04852‐14

[21]

Brown CL, Keenum IM, Dai D, Zhang L, Vikesland PJ, Pruden A. Critical evaluation of short, long, and hybrid assembly for contextual analysis of antibiotic resistance genes in complex environmental metagenomes. Sci Rep. 2021;11(1):3753. https://doi.org/10.1038/s41598‐021‐83081‐8

[22]

Raghunathan A, Ferguson HR, Bornarth CJ, Song W, Driscoll M, Lasken RS. Genomic DNA amplification from a single bacterium. Appl Environ Microbiol. 2005;71(6):3342–7. https://doi.org/10.1128/AEM.71.6.3342‐3347.2005

[23]

Direito SOL, Zaura E, Little M, Ehrenfreund P, Röling WFM. Systematic evaluation of bias in microbial community profiles induced by whole genome amplification. Environ Microbiol. 2014;16(3):643–57. https://doi.org/10.1111/1462‐2920.12365

[24]

He Y, Wang X, Ma B, Xu J. Ramanome technology platform for label‐free screening and sorting of microbial cell factories at single‐cell resolution. Biotechnol Adv. 2019;37(6):107388. https://doi.org/10.1016/j.biotechadv.2019.04.010

[25]

He Y, Huang S, Zhang P, Ji Y, Xu J. Intra‐Ramanome Correlation Analysis unveils metabolite conversion network from an isogenic population of cells. mBio. 2021;12(4):e01470–21. https://doi.org/10.1128/mBio.01470‐21

[26]

Berry D, Mader E, Lee TK, Woebken D, Wang Y, Zhu D, et al. Tracking heavy water (D2O) incorporation for identifying and sorting active microbial cells. Proc Natl Acad Sci U S A. 2015;112(2):E194–203. https://doi.org/10.1073/pnas.1420406112

[27]

Jing X, Gou H, Gong Y, Su X, Xu L, Ji Y, et al. Raman‐activated cell sorting and metagenomic sequencing revealing carbon‐fixing bacteria in the ocean. Environ Microbiol. 2018;20(6):2241–55. https://doi.org/10.1111/1462‐2920.14268

[28]

Wang X, Ren L, Su Y, Ji Y, Liu Y, Li C, et al. Raman‐Activated Droplet Sorting (RADS) for label‐free high‐throughput screening of microalgal single‐cells. Anal Chem. 2017;89(22):12569–77. https://doi.org/10.1021/acs.analchem.7b03884

[29]

Wang T, Ji Y, Wang Y, Jia J, Li J, Huang S, et al. Quantitative dynamics of triacylglycerol accumulation in microalgae populations at single‐cell resolution revealed by Raman microspectroscopy. Biotechnol Biofuels. 2014;7(1):58. https://doi.org/10.1186/1754‐6834‐7‐58

[30]

Ji Y, He Y, Cui Y, Wang T, Wang Y, Li Y, et al. Raman spectroscopy provides a rapid, non‐invasive method for quantitation of starch in live, unicellular microalgae. Biotechnol J. 2014;9(12):1512–8. https://doi.org/10.1002/biot.201400165

[31]

He Y, Zhang P, Huang S, Wang T, Ji Y, Xu J. Label‐free, simultaneous quantification of starch, protein and triacylglycerol in single microalgal cells. Biotechnol Biofuels. 2017;10(1):275. https://doi.org/10.1186/s13068‐017‐0967‐x

[32]

Tao Y, Wang Y, Huang S, Zhu P, Huang WE, Ling J, et al. Metabolic‐activity‐based assessment of antimicrobial effects by D2O‐labeled single‐cell Raman microspectroscopy. Anal Chem. 2017;89(7):4108–15. https://doi.org/10.1021/acs.analchem.6b05051

[33]

Teng L, Wang X, Wang X, Gou H, Ren L, Wang T, et al. Label‐free, rapid and quantitative phenotyping of stress response in E. coli via ramanome. Sci Rep. 2016;6(1):34359. https://doi.org/10.1038/srep34359

[34]

Hekmatara M, Heidari Baladehi M, Ji Y, Xu J. D2O‐probed Raman microspectroscopy distinguishes the metabolic dynamics of macromolecules in organellar anticancer drug response. Anal Chem. 2021;93(4):2125–34. https://doi.org/10.1021/acs.analchem.0c03925

[35]

Wang Y, Song Y, Tao Y, Muhamadali H, Goodacre R, Zhou N.‐Y, et al. Reverse and multiple stable isotope probing to study bacterial metabolism and interactions at the single cell level. Anal Chem. 2016;88(19):9443–50. https://doi.org/10.1021/acs.analchem.6b01602

[36]

Ho C.‐S, Jean N, Hogan CA, Blackmon L, Jeffrey SS, Holodniy M, et al. Rapid identification of pathogenic bacteria using Raman spectroscopy and deep learning. Nat Commun. 2019;10(1):4927. https://doi.org/10.1038/s41467‐019‐12898‐9

[37]

Heidari Baladehi M, Hekmatara M, He Y, Bhaskar Y, Wang Z, Liu L, et al. Culture‐free identification and metabolic profiling of microalgal single cells via ensemble learning of ramanomes. Anal Chem. 2021;93(25):8872–80. https://doi.org/10.1021/acs.analchem.1c01015

[38]

Liu M, Zhu P, Zhang L, Gong Y, Wang C, Sun L, et al. Single‐cell identification, drug susceptibility test, and whole‐genome sequencing of Helicobacter pylori directly from gastric biopsy by CAntimicrobial Agents and Chemotherapylinical Antimicrobial Susceptibility Test Ramanometry. Clin Chem. 2022;68(8):1064–74. https://doi.org/10.1093/clinchem/hvac082

[39]

Jing X, Gong Y, Xu T, Meng Y, Han X, Su X, et al. One‐cell metabolic phenotyping and sequencing of soil microbiome by Raman‐Activated Gravity‐Driven Encapsulation (RAGE). mSystems. 2021;6(3):e00181–21. https://doi.org/10.1128/mSystems.00181‐21

[40]

Xu T, Gong Y, Su X, Zhu P, Dai J, Xu J, et al. Phenome–genome profiling of single bacterial cell by Raman‐Activated Gravity‐driven Encapsulation and sequencing. Small. 2020;16(30):2001172. https://doi.org/10.1002/smll.202001172

[41]

Zhu P, Ren L, Zhu Y, Dai J, Liu H, Mao Y, et al. Rapid, automated, and reliable antimicrobial susceptibility test from positive blood culture by CAST‐R. mLife. 2022;1(3):329–40. https://doi.org/10.1002/mlf2.12019

[42]

Wang C, Chen R, Xu J, Jin L. Single‐cell Raman spectroscopy identifies Escherichia coli persisters and reveals their enhanced metabolic activities. Front Microbiol. 2022;13:936726. https://doi.org/10.3389/fmicb.2022.936726

[43]

Wang X, Xin Y, Ren L, Sun Z, Zhu P, Ji Y, et al. Positive dielectrophoresis‐based Raman‐activated droplet sorting for culture‐free and label‐free screening of enzyme function in vivo. Sci Adv. 2020;6(32):eabb3521. https://doi.org/10.1126/sciadv.abb3521

[44]

Wang X, Ren L, Diao Z, He Y, Zhang J, Liu M, et al. Robust spontaneous Raman flow cytometry for single‐cell metabolic phenome profiling via pDEP‐DLD‐RFC. Adv Sci. 2023;10(16):e2207497. https://doi.org/10.1002/advs.202207497

[45]

Jing X, Gong Y, Xu T, Davison PA, MacGregor‐Chatwin C, Hunter CN, et al. Revealing CO2‐fixing SAR11 bacteria in the ocean by Raman‐based single‐cell metabolic profiling and genomics. BioDesign Res. 2022;2022:9782712. https://doi.org/10.34133/2022/9782712

[46]

Jing X, Gong Y, Pan H, Meng Y, Ren Y, Diao Z, et al. Single‐cell Raman‐activated sorting and cultivation (scRACS‐Culture) for assessing and mining in situ phosphate‐solubilizing microbes from nature. ISME Commun. 2022;2(1):106. https://doi.org/10.1038/s43705‐022‐00188‐3

[47]

Zheng W, Zhao S, Yin Y, Zhang H, Needham DM, Evans ED, et al. High‐throughput, single‐microbe genomics with strain resolution, applied to a human gut microbiome. Science. 2022;376(6597):eabm1483. https://doi.org/10.1126/science.abm1483

[48]

Zhang M, Hong W, Abutaleb NS, Li J, Dong P.‐T, Zong C, et al. Rapid determination of antimicrobial susceptibility by stimulated Raman scattering imaging of D2O metabolic incorporation in a single bacterium. Adv Sci. 2020;7(19):2001452. https://doi.org/10.1002/advs.202001452

[49]

Li H, Hsieh K, Wong PK, Mach KE, Liao JC, Wang T.‐H. Single‐cell pathogen diagnostics for combating antibiotic resistance. Nat Rev Methods Primers. 2023;3(1):6. https://doi.org/10.1038/s43586‐022‐00190‐y

[50]

Xu T, Li Y, Han X, Kan L, Ren J, Sun L, et al. Versatile, facile and low‐cost single‐cell isolation, culture and sequencing by optical tweezer‐assisted pool‐screening. Lab Chip. 2023;23(1):125–35. https://doi.org/10.1039/D2LC00888B

[51]

Diao Z, Kan L, Zhao Y, Yang H, Song J, Wang C, et al. Artificial intelligence‐assisted automatic and index‐based microbial single‐cell sorting system for One‐Cell‐One‐Tube. mLife. 2022;1(4):448–59. https://doi.org/10.1002/mlf2.12047

[52]

Kolaček S, Hojsak I, Berni Canani R, Guarino A, Indrio F, Orel R, et al. Commercial probiotic products: a call for improved quality control. A position paper by the ESPGHAN working group for probiotics and prebiotics. J Pediatr Gastroenterol Nutr. 2017;65(1):117–24. https://doi.org/10.1097/mpg.0000000000001603

[53]

Adefisoye MA, Green E, Njobeh PB, Oyedeji AB, Adebo OA. Quality control of probiotic beverages and organisms. Probiotic Beverages. 2021:389–402. https://doi.org/10.1016/B978‐0‐12‐818588‐9.00006‐1

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Published: 09 May 2023
Issue date: June 2023

Copyright

© 2023 The Authors. Tsinghua University Press.

Acknowledgements

ACKNOWLEDGMENTS

We thank Yang Liu for graphics support. This study was funded by the National Key R&D Program of China (2022YFA1304101), CAS (XDB29050400), the National Natural Science Foundation of China (32030003), and Shenzhen ‐ Hong Kong Innovation Circle Plan (SGDX2019081623060946).

Rights and permissions

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

Return