Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alexey V. Melnik is active.

Publication


Featured researches published by Alexey V. Melnik.


Science Translational Medicine | 2017

Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis

Teruaki Nakatsuji; Tiffany H. Chen; Saisindhu Narala; K.A. Chun; Aimee Two; T. Yun; Faiza Shafiq; Paul Kotol; Amina Bouslimani; Alexey V. Melnik; Haythem Latif; Kim Jn; Lockhart A; Artis K; Gloria David; Patricia A. Taylor; Joanne E. Streib; Pieter C. Dorrestein; Grier A; Gill; Karsten Zengler; Tissa Hata; Donald Y.M. Leung; Richard L. Gallo

Commensal skin bacteria produce previously unknown antimicrobial peptides that can inhibit Staphylococcus aureus colonization of atopic dermatitis subjects. Bacterial biological warfare in atopic dermatitis Normal human skin is colonized by a variety of bacteria, which typically do not perturb the host. However, Staphylococcus aureus is known to aggravate symptoms of atopic dermatitis. Nakatsuji et al. report that other strains of Staphylococcus residing on the skin of healthy individuals produce a novel antimicrobial peptide that can inhibit S. aureus growth. Colonization of pigskin or mice with these protective commensals reduced S. aureus replication. Autologous bacterial transplant in a small number of atopic dermatitis patients drastically reduced S. aureus skin burden. This commensal skin transplant is already approved by the U.S. Food and Drug Administration, with a clinical trial underway. The microbiome can promote or disrupt human health by influencing both adaptive and innate immune functions. We tested whether bacteria that normally reside on human skin participate in host defense by killing Staphylococcus aureus, a pathogen commonly found in patients with atopic dermatitis (AD) and an important factor that exacerbates this disease. High-throughput screening for antimicrobial activity against S. aureus was performed on isolates of coagulase-negative Staphylococcus (CoNS) collected from the skin of healthy and AD subjects. CoNS strains with antimicrobial activity were common on the normal population but rare on AD subjects. A low frequency of strains with antimicrobial activity correlated with colonization by S. aureus. The antimicrobial activity was identified as previously unknown antimicrobial peptides (AMPs) produced by CoNS species including Staphylococcus epidermidis and Staphylococcus hominis. These AMPs were strain-specific, highly potent, selectively killed S. aureus, and synergized with the human AMP LL-37. Application of these CoNS strains to mice confirmed their defense function in vivo relative to application of nonactive strains. Strikingly, reintroduction of antimicrobial CoNS strains to human subjects with AD decreased colonization by S. aureus. These findings show how commensal skin bacteria protect against pathogens and demonstrate how dysbiosis of the skin microbiome can lead to disease.


PLOS Computational Biology | 2014

Pep2Path: automated mass spectrometry-guided genome mining of peptidic natural products

Marnix H. Medema; Yared Paalvast; Don D. Nguyen; Alexey V. Melnik; Pieter C. Dorrestein; Eriko Takano; Rainer Breitling

Nonribosomally and ribosomally synthesized bioactive peptides constitute a source of molecules of great biomedical importance, including antibiotics such as penicillin, immunosuppressants such as cyclosporine, and cytostatics such as bleomycin. Recently, an innovative mass-spectrometry-based strategy, peptidogenomics, has been pioneered to effectively mine microbial strains for novel peptidic metabolites. Even though mass-spectrometric peptide detection can be performed quite fast, true high-throughput natural product discovery approaches have still been limited by the inability to rapidly match the identified tandem mass spectra to the gene clusters responsible for the biosynthesis of the corresponding compounds. With Pep2Path, we introduce a software package to fully automate the peptidogenomics approach through the rapid Bayesian probabilistic matching of mass spectra to their corresponding biosynthetic gene clusters. Detailed benchmarking of the method shows that the approach is powerful enough to correctly identify gene clusters even in data sets that consist of hundreds of genomes, which also makes it possible to match compounds from unsequenced organisms to closely related biosynthetic gene clusters in other genomes. Applying Pep2Path to a data set of compounds without known biosynthesis routes, we were able to identify candidate gene clusters for the biosynthesis of five important compounds. Notably, one of these clusters was detected in a genome from a different subphylum of Proteobacteria than that in which the molecule had first been identified. All in all, our approach paves the way towards high-throughput discovery of novel peptidic natural products. Pep2Path is freely available from http://pep2path.sourceforge.net/, implemented in Python, licensed under the GNU General Public License v3 and supported on MS Windows, Linux and Mac OS X.


Nature microbiology | 2017

Indexing the Pseudomonas specialized metabolome enabled the discovery of poaeamide B and the bananamides

Don D. Nguyen; Alexey V. Melnik; Nobuhiro Koyama; Xiaowen Lu; Michelle Schorn; Jinshu Fang; Kristen Aguinaldo; Tommie Lincecum; Maarten G. K. Ghequire; Víctor J. Carrión; Tina L. Cheng; Brendan M. Duggan; Jacob G. Malone; Tim H. Mauchline; Laura M. Sanchez; A. Marm Kilpatrick; Jos M. Raaijmakers; René De Mot; Bradley S. Moore; Marnix H. Medema; Pieter C. Dorrestein

Pseudomonads are cosmopolitan microorganisms able to produce a wide array of specialized metabolites. These molecules allow Pseudomonas to scavenge nutrients, sense population density and enhance or inhibit growth of competing microorganisms. However, these valuable metabolites are typically characterized one-molecule–one-microbe at a time, instead of being inventoried in large numbers. To index and map the diversity of molecules detected from these organisms, 260 strains of ecologically diverse origins were subjected to mass-spectrometry-based molecular networking. Molecular networking not only enables dereplication of molecules, but also sheds light on their structural relationships. Moreover, it accelerates the discovery of new molecules. Here, by indexing the Pseudomonas specialized metabolome, we report the molecular-networking-based discovery of four molecules and their evolutionary relationships: a poaeamide analogue and a molecular subfamily of cyclic lipopeptides, bananamides 1, 2 and 3. Analysis of their biosynthetic gene cluster shows that it constitutes a distinct evolutionary branch of the Pseudomonas cyclic lipopeptides. Through analysis of an additional 370 extracts of wheat-associated Pseudomonas, we demonstrate how the detailed knowledge from our reference index can be efficiently propagated to annotate complex metabolomic data from other studies, akin to the way in which newly generated genomic information can be compared to data from public databases.


mSystems | 2016

From Sample to Multi-Omics Conclusions in under 48 Hours

Robert A. Quinn; Jose A. Navas-Molina; Embriette R. Hyde; Se Jin Song; Yoshiki Vázquez-Baeza; Greg Humphrey; James Gaffney; Jeremiah J. Minich; Alexey V. Melnik; Jakob Herschend; Jeff DeReus; Austin Durant; Rachel J. Dutton; Mahdieh Khosroheidari; Clifford Green; Ricardo Azevedo da Silva; Pieter C. Dorrestein; Rob Knight

Polymicrobial infections are difficult to diagnose due to the challenge in comprehensively cultivating the microbes present. Omics methods, such as 16S rRNA sequencing, metagenomics, and metabolomics, can provide a more complete picture of a microbial community and its metabolite production, without the biases and selectivity of microbial culture. However, these advanced methods have not been applied to clinical or industrial microbiology or other areas where complex microbial dysbioses require immediate intervention. The reason for this is the length of time required to generate and analyze omics data. Here, we describe the development and application of a pipeline for multi-omics data analysis in time frames matching those of the culture-based approaches often used for these applications. This study applied multi-omics methods effectively in clinically relevant time frames and sets a precedent toward their implementation in clinical medicine and industrial microbiology. ABSTRACT Multi-omics methods have greatly advanced our understanding of the biological organism and its microbial associates. However, they are not routinely used in clinical or industrial applications, due to the length of time required to generate and analyze omics data. Here, we applied a novel integrated omics pipeline for the analysis of human and environmental samples in under 48 h. Human subjects that ferment their own foods provided swab samples from skin, feces, oral cavity, fermented foods, and household surfaces to assess the impact of home food fermentation on their microbial and chemical ecology. These samples were analyzed with 16S rRNA gene sequencing, inferred gene function profiles, and liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomics through the Qiita, PICRUSt, and GNPS pipelines, respectively. The human sample microbiomes clustered with the corresponding sample types in the American Gut Project (http://www.americangut.org ), and the fermented food samples produced a separate cluster. The microbial communities of the household surfaces were primarily sourced from the fermented foods, and their consumption was associated with increased gut microbial diversity. Untargeted metabolomics revealed that human skin and fermented food samples had separate chemical ecologies and that stool was more similar to fermented foods than to other sample types. Metabolites from the fermented foods, including plant products such as procyanidin and pheophytin, were present in the skin and stool samples of the individuals consuming the foods. Some food metabolites were modified during digestion, and others were detected in stool intact. This study represents a first-of-its-kind analysis of multi-omics data that achieved time intervals matching those of classic microbiological culturing. IMPORTANCE Polymicrobial infections are difficult to diagnose due to the challenge in comprehensively cultivating the microbes present. Omics methods, such as 16S rRNA sequencing, metagenomics, and metabolomics, can provide a more complete picture of a microbial community and its metabolite production, without the biases and selectivity of microbial culture. However, these advanced methods have not been applied to clinical or industrial microbiology or other areas where complex microbial dysbioses require immediate intervention. The reason for this is the length of time required to generate and analyze omics data. Here, we describe the development and application of a pipeline for multi-omics data analysis in time frames matching those of the culture-based approaches often used for these applications. This study applied multi-omics methods effectively in clinically relevant time frames and sets a precedent toward their implementation in clinical medicine and industrial microbiology.


Journal of Natural Products | 2017

Prioritizing Natural Product Diversity in a Collection of 146 Bacterial Strains Based on Growth and Extraction Protocols

Max Crüsemann; Ellis C. O’Neill; Charles B. Larson; Alexey V. Melnik; Dimitrios J. Floros; Ricardo R. da Silva; Paul R. Jensen; Pieter C. Dorrestein; Bradley S. Moore

In order to expedite the rapid and efficient discovery and isolation of novel specialized metabolites, while minimizing the waste of resources on rediscovery of known compounds, it is crucial to develop efficient approaches for strain prioritization, rapid dereplication, and the assessment of favored cultivation and extraction conditions. Herein we interrogated bacterial strains by systematically evaluating cultivation and extraction parameters with LC-MS/MS analysis and subsequent dereplication through the Global Natural Product Social Molecular Networking (GNPS) platform. The developed method is fast, requiring minimal time and sample material, and is compatible with high-throughput extract analysis, thereby streamlining strain prioritization and evaluation of culturing parameters. With this approach, we analyzed 146 marine Salinispora and Streptomyces strains that were grown and extracted using multiple different protocols. In total, 603 samples were analyzed, generating approximately 1.8 million mass spectra. We constructed a comprehensive molecular network and identified 15 molecular families of diverse natural products and their analogues. The size and breadth of this network shows statistically supported trends in molecular diversity when comparing growth and extraction conditions. The network provides an extensive survey of the biosynthetic capacity of the strain collection and a method to compare strains based on the variety and novelty of their metabolites. This approach allows us to quickly identify patterns in metabolite production that can be linked to taxonomy, culture conditions, and extraction methods, as well as informing the most valuable growth and extraction conditions.


Cell Host & Microbe | 2017

Three-Dimensional Microbiome and Metabolome Cartography of a Diseased Human Lung

Neha Garg; Mingxun Wang; Embriette R. Hyde; Ricardo R. da Silva; Alexey V. Melnik; Ivan Protsyuk; Amina Bouslimani; Yan Wei Lim; Richard Wong; Greg Humphrey; Gail Ackermann; Timothy Spivey; Sharon Brouha; Nuno Bandeira; Grace Y. Lin; Forest Rohwer; Douglas Conrad; Theodore Alexandrov; Rob Knight; Pieter C. Dorrestein

Our understanding of the spatial variation in the chemical and microbial makeup of an entire human organ remains limited, in part due to the size and heterogeneity of human organs and the complexity of the associated metabolome and microbiome. To address this challenge, we developed a workflow to enable the cartography of metabolomic and microbiome data onto a three-dimensional (3D) organ reconstruction built off radiological images. This enabled the direct visualization of the microbial and chemical makeup of a human lung from a cystic fibrosis patient. We detected host-derived molecules, microbial metabolites, medications, and region-specific metabolism of medications and placed it in the context of microbial distributions in the lung. Our tool further created browsable maps of a 3D microbiome/metabolome reconstruction map on a radiological image of a human lung and forms an interactive resource for the scientific community.


Nature Reviews Microbiology | 2018

Best practices for analysing microbiomes

Rob Knight; Alison Vrbanac; Bryn C. Taylor; Alexander A. Aksenov; Chris Callewaert; Justine W. Debelius; Antonio González; Tomasz Kosciolek; Laura-Isobel McCall; Daniel McDonald; Alexey V. Melnik; James T. Morton; Jose Navas; Robert A. Quinn; Jon G. Sanders; Austin D. Swafford; Luke R. Thompson; Anupriya Tripathi; Zhenjiang Zech Xu; Jesse Zaneveld; Qiyun Zhu; J. Gregory Caporaso; Pieter C. Dorrestein

Complex microbial communities shape the dynamics of various environments, ranging from the mammalian gastrointestinal tract to the soil. Advances in DNA sequencing technologies and data analysis have provided drastic improvements in microbiome analyses, for example, in taxonomic resolution, false discovery rate control and other properties, over earlier methods. In this Review, we discuss the best practices for performing a microbiome study, including experimental design, choice of molecular analysis technology, methods for data analysis and the integration of multiple omics data sets. We focus on recent findings that suggest that operational taxonomic unit-based analyses should be replaced with new methods that are based on exact sequence variants, methods for integrating metagenomic and metabolomic data, and issues surrounding compositional data analysis, where advances have been particularly rapid. We note that although some of these approaches are new, it is important to keep sight of the classic issues that arise during experimental design and relate to research reproducibility. We describe how keeping these issues in mind allows researchers to obtain more insight from their microbiome data sets.Complex microbial communities shape the dynamics of various environments. In this Review, Knight and colleagues discuss the best practices for performing a microbiome study, including experimental design, choice of molecular analysis technology, methods for data analysis and the integration of multiple omics data sets.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Lifestyle chemistries from phones for individual profiling

Amina Bouslimani; Alexey V. Melnik; Zhenjiang Xu; Amnon Amir; Ricardo R. da Silva; Mingxun Wang; Nuno Bandeira; Theodore Alexandrov; Rob Knight; Pieter C. Dorrestein

Significance This paper introduces the concept of skin-associated lifestyle chemistries found on personal belongings as a form of trace evidence. We propose a mass spectrometry-based approach to illuminate chemical traces recovered from personal objects. Using a chemical composite recovered from a swab of a phone, as a representative personal belonging, we can provide insights into personal lifestyle profile by predicting the kind of beauty product the individual uses, the food he/she eats, the medications he/she takes, or the places he/she has been. Therefore, the chemical interpretation of traces recovered from objects found on a crime scene can help a criminal investigator to learn about the lifestyle of the individual who used or touched these objects. Imagine a scenario where personal belongings such as pens, keys, phones, or handbags are found at an investigative site. It is often valuable to the investigative team that is trying to trace back the belongings to an individual to understand their personal habits, even when DNA evidence is also available. Here, we develop an approach to translate chemistries recovered from personal objects such as phones into a lifestyle sketch of the owner, using mass spectrometry and informatics approaches. Our results show that phones’ chemistries reflect a personalized lifestyle profile. The collective repertoire of molecules found on these objects provides a sketch of the lifestyle of an individual by highlighting the type of hygiene/beauty products the person uses, diet, medical status, and even the location where this person may have been. These findings introduce an additional form of trace evidence from skin-associated lifestyle chemicals found on personal belongings. Such information could help a criminal investigator narrowing down the owner of an object found at a crime scene, such as a suspect or missing person.


Frontiers in Plant Science | 2017

Mass Spectrometry Based Molecular 3D-Cartography of Plant Metabolites

Dimitrios J. Floros; Daniel Petras; Clifford A. Kapono; Alexey V. Melnik; Tie-Jun Ling; Rob Knight; Pieter C. Dorrestein

Plants play an essential part in global carbon fixing through photosynthesis and are the primary food and energy source for humans. Understanding them thoroughly is therefore of highest interest for humanity. Advances in DNA and RNA sequencing and in protein and metabolite analysis allow the systematic description of plant composition at the molecular level. With imaging mass spectrometry, we can now add a spatial level, typically in the micrometer-to-centimeter range, to their compositions, essential for a detailed molecular understanding. Here we present an LC-MS based approach for 3D plant imaging, which is scalable and allows the analysis of entire plants. We applied this approach in a case study to pepper and tomato plants. Together with MS/MS spectra library matching and spectral networking, this non-targeted workflow provides the highest sensitivity and selectivity for the molecular annotations and imaging of plants, laying the foundation for studies of plant metabolism and plant-environment interactions.


Analytical Chemistry | 2017

Coupling Targeted and Untargeted Mass Spectrometry for Metabolome-Microbiome-Wide Association Studies of Human Fecal Samples

Alexey V. Melnik; Ricardo R. da Silva; Embriette R. Hyde; Alexander A. Aksenov; Fernando Vargas; Amina Bouslimani; Ivan Protsyuk; Alan K. Jarmusch; Anupriya Tripathi; Theodore Alexandrov; Rob Knight; Pieter C. Dorrestein

Increasing appreciation of the gut microbiomes role in health motivates understanding the molecular composition of human feces. To analyze such complex samples, we developed a platform coupling targeted and untargeted metabolomics. The approach is facilitated through split flow from one UPLC, joint timing triggered by contact closure relays, and a script to retrieve the data. It is designed to detect specific metabolites of interest with high sensitivity, allows for correction of targeted information, enables better quantitation thus providing an advanced analytical tool for exploratory studies. Procrustes analysis revealed that untargeted approach provides a better correlation to microbiome data, associating specific metabolites with microbes that produce or process them. With the subset of over one hundred human fecal samples from the American Gut project, the implementation of the described coupled workflow revealed that targeted analysis using combination of single transition per compound with retention time misidentifies 30% of the targeted data and could lead to incorrect interpretations. At the same time, the targeted analysis extends detection limits and dynamic range, depending on the compounds, by orders of magnitude. A software application has been developed as a part of the workflow to allows for quantitative assessments based on calibration curves. Using this approach, we detect expected microbially modified molecules such as secondary bile acids and unexpected microbial molecules including Pseudomonas-associated quinolones and rhamnolipids in feces, setting the stage for metabolome-microbiome-wide association studies (MMWAS).

Collaboration


Dive into the Alexey V. Melnik's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rob Knight

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Neha Garg

University of Montana

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ivan Protsyuk

European Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gail Ackermann

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge