Abstract
Breast cancer is characterized by oncobiosis, the abnormal composition of the microbiome in neoplastic diseases. The biosynthetic capacity of the oncobiotic flora in breast cancer is suppressed, as suggested by metagenomic studies. The microbiome synthesizes a set of cytostatic and antimetastatic metabolites that are downregulated in breast cancer, including cadaverine, a microbiome metabolite with cytostatic properties. We set out to assess how the protein expression of constitutive lysine decarboxylase (LdcC), a key enzyme for cadaverine production, changes in the feces of human breast cancer patients (n = 35). We found that the fecal expression of Escherichia coli LdcC is downregulated in lobular cases as compared to invasive carcinoma of no special type (NST) cases. Lobular breast carcinoma is characterized by low or absent expression of E-cadherin. Fecal E. coli LdcC protein expression is downregulated in E-cadherin negative breast cancer cases as compared to positive ones. Receiver operating characteristic (ROC) analysis of LdcC expression in lobular and NST cases revealed that fecal E. coli LdcC protein expression might have predictive values. These data suggest that the oncobiotic transformation of the microbiome indeed leads to the downregulation of the production of cytostatic and antimetastatic metabolites. In E-cadherin negative lobular carcinoma that has a higher potential for metastasis formation, the protein levels of enzymes producing antimetastatic metabolites are downregulated. This finding represents a new route that renders lobular cases permissive for metastasis formation. Furthermore, our findings underline the role of oncobiosis in regulating metastasis formation in breast cancer.
Introduction
Breast cancer is characterized by an altered gut [1–7], breast [8], oral [9], and urinary [9] microbiome. The majority of studies reported suppressed microbiome diversity (reviewed in Ref. [10]). Animal [11, 12] and human studies [13] suggest that antibiotic administration that reduces the biomass and diversity of gut flora, increases the risk for breast cancer development. Probiotic treatment has an inverse effect by decreasing the incidence of breast cancer [14]. These observations suggest a pathophysiological role for the oncobiome in breast cancer.
Changes to the microbiome can affect the immune functions of the host [15] and the microbes can modulate the behavior of the host by secreting pleiotropic metabolites. These metabolites get to cancer cells through the systemic circulation [10]. The majority of the studies report lower diversity in the breast cancer oncobiome as compared to the healthy eubiome, resulting in a restricted biosynthetic capacity [3, 6, 7]. This lower biosynthetic capacity probably leads to lower production of bacterial metabolites in both the serum and the tumor. A number of antineoplastic bacterial metabolites were identified in breast cancer, including short chain fatty acids [10], lithocholic acid [7, 16] and cadaverine [6].
Cadaverine can be synthesized from lysine through decarboxylation by bacterial enzymes (LdcC and CadA) [17, 18], although the human body can also produce cadaverine. Shigella flexneri, Shigella sonnei, Escherichia coli, and Streptococcus possess enzymes for cadaverine biosynthesis [19]. The main role of cadaverine is to buffer the pH of the environment [20]. The receptors for cadaverine are members of the trace amino acid receptor (TAAR) family [6, 21]. High TAAR1 expression correlates with better survival in breast cancer [22]. Furthermore, TAAR1, TAAR2, TAAR3, TAAR5, TAAR8, and TAAR9 can repress breast cancer [6]. Cadaverine suppresses epithelial-to-mesenchymal transition, cellular movement, chemotaxis, diapedesis, and metastasis formation in breast cancer cells, but has no effect on non-transformed cells [6].
There is an ample set of metagenomic studies in breast cancer cases [10]. However, to our best knowledge, no protein studies are available due to difficult sample preparation and lack of antibodies for analysis. In the current study, we assessed the expression of a bacterial enzyme, LdcC, which produces the cytostatic bioactive metabolite, cadaverine [6]. The aim of this study was to assess the fecal protein expression of E. coli LdcC in breast cancer patients.
Materials and methods
Patient cohort
Fecal protein expression was assessed in a female breast cancer patient cohort composed of 35 participants with a mean age of 57 years. Samples from stage I–III and Nottingham grade 1–3 patients were used in the study. The average stage and grade were not statistically different amongst the groups. Fecal samples were obtained from patients with no special type (NST) and lobular tumors; tumors of other histological types were excluded.
The collection and biobanking of feces were authorized by the Hungarian national authority (ETT). Patients and healthy volunteers meeting the following criteria were excluded from the study: 1) has a previous history of breast cancer or had been operated due to neoplasia, 2) has a disease of unknown origin, 3) has a chronic contagious disease, 4) had contagious diarrhea 6 months prior to enrollment, 5) taken antibiotics within the 6 months prior to enrollment, 6) had chemotherapy, biological therapy, or immunosuppressive therapy 6 months prior to enrollment, 7) used intravenous drugs 12 months prior to enrollment, 8) had piercing, tattooing, acupuncture, or other endangering behavior or action 12 months prior to enrollment, 9) exposure to an allergen to which the enrolled individual had been sensitized to, or 10) underwent colonoscopy 12 months prior to enrollment. The first morning feces was sampled; samples were frozen and deposited in the biobank within 2 h after defecation. Samples were stored at −70 °C until subsequent use. We obtained informed consent from study participants. The patient's routine pathological data were assessed in the study and were compared to the fecal expression of E. coli LdcC.
Fecal protein sample preparation
Fecal proteins were isolated as described in Ref. [6]. Fecal samples (100 mg) were lysed in 500 µL RIPA buffer (50 mM Tris, 150 mM NaCl, 0.1% SDS, 1% Triton X-100, 0.5% sodium deoxycholate, 1 mM EDTA, 1 mM Na3VO4, 1 mM PMSF, 1 mM NaF, protease inhibitor cocktail) and sonicated (Qsonica Q125 Sonicator, Newtown, Connecticut) 3 times for 30 s with 50% amplitude. After centrifugation, 8 µL β-mercaptoethanol and 25 µL 5× SDS sample buffer (50% glycerol, 10% SDS, 310 mM Tris HCl, pH 6.8, 100 mM DTT, 0.01% bromophenol blue) were added to each 100 µL extract. Then, fecal protein samples were heated for 10 min at 96 °C and held on ice until loading. Protein extract (40 µL) was loaded on 8% SDS-PAGE gels and proteins were separated and transferred onto nitrocellulose membrane. Ponceau-red staining took place after transfer, but before blocking. Ponceau-stained membranes were photographed and used in subsequent analyses. Membranes were cut at the 70 kDa standard and the top part was blotted for LdcC (E. coli LdcC, 1:100, Abcam (ab193351)). Membranes were blocked in TBST containing 5% BSA for 1 h and incubated with anti-LdcC primary antibody overnight at 4 °C. After washing with 1× TBS-TWEEN solution, the membranes were probed with IgG HRP-conjugated peroxidase secondary antibodies (1:2000, Cell Signaling Technology, Inc, Beverly, MA, USA). Bands were visualized by enhanced chemiluminescence reaction (SuperSignal West Pico Solutions, Thermo Fisher Scientific Inc., Rockford, IL, USA). Blots were evaluated by densitometry using Image J software and antibody signals were normalized to total protein stained by Ponceau-red (Sigma-Aldrich). A sample LdcC blot is published in Ref. [6].
Statistical analysis
All data are represented as average ± SEM. Statistical tests are mentioned in figure captions. For statistical analysis, Graphpad 8.1 software was used. The receiver operating characteristic (ROC) curve was re-calculated using R (version v 3.6.3 (2020-02-29)) [23] and the pROC package [24].
Results
Fecal expression of LdcC is low in the E-cadherin negative, lobular subtype of breast cancer
First, we assessed the fecal protein expression of E. coli LdcC as a function of the histological subtype of breast cancer. We found that the fecal expression of E. coli LdcC enzyme was lower in the lobular subtype compared to the invasive carcinoma of no special type (NST) of breast cancer (Fig. 1A).
Lobular breast carcinoma is characterized by lower or absent E-cadherin expression [25]. Furthermore, the absence of E-cadherin is a predictive factor for metastasis formation, and therefore, predicts poorer clinical outcomes [25–27]. We compared the LdcC expression in E-cadherin positive and negative tumors. The fecal expression of E. coli LdcC was significantly lower in E-cadherin negative cases as compared to E-cadherin positive cases (Fig. 1B).
The striking difference in the fecal expression of E. coli LdcC in E-cadherin+ versus E-cadherin− cases prompted us to assess whether it would be possible to discriminate between E-cadherin+ and E-cadherin− cases using LdcC expression. To that end, we performed a ROC analysis. According to the ROC analysis, fecal E. coli LdcC expression is a good assay to classify E-cadherin+ and E-cadherin− cases (Fig. 2). The area under the curve for the ROC analysis was 0.8276 (Std error: 0.0864, 95% confidence interval [CI]: 0.6583–0.9969). The threshold of the curve was 0.642 (corresponding specificity 83.33% (CI: 50–100) and sensitivity was 72.41% (CI: 55.17–86.21)). The P value for the curve was P=0.0126.
Discussion
All previous work on the breast cancer oncobiome applied nucleic acid-based techniques [10]. The bacterial metabolome was not assessed in dedicated studies, only tangentially in general metabolomic studies [7, 28, 29]. General bacterial proteomic studies are missing. Due to these caveats, we do not have a comprehensive picture of the metabolic function of the oncobiome and its pathophysiological role in breast cancer. Our aim was to provide an initial, rudimentary study on fecal bacterial protein expression in breast cancer patients, as a proof of principle study. The major technical drawback we faced was the lack of antibodies against relevant bacterial enzymes in diverse species. In this study, we used an antibody specific to the LdcC enzyme that produces cadaverine, which has cytostatic properties in breast cancer [6]. The anti-LdcC antibody was specific for E. coli and there were no available antibodies against the same enzyme in other species. LdcC was downregulated at the DNA level, when comparing breast cancer patients to healthy controls [6]. In support of this observation, breast cancer is characterized by suppressed diversity [10] and suppressed biosynthetic capacity of the gut microbiome [3, 6, 7].
Fecal E. coli LdcC enzyme expression was downregulated in lobular E-cadherin negative breast cancer cases. E-cadherin is a cell adhesion molecule, which is crucial in the suppression of invasion [26]. Epigenetic reprogramming or mutations that downregulate E-cadherin expression support tissue invasion and metastasis formation [25–27, 30]. Re-expression of E-cadherin reprograms mesenchymal cells towards epithelial morphology [30]. Lobular breast cancer has a higher potential for metastasis formation, marked by a higher frequency of peritoneal metastases (e.g., Krukenberg tumors). Increased peritoneal metastases is probably linked to low or absent E-cadherin expression. The finding that fecal E. coli LdcC expression was lower in E-cadherin negative cases is in good agreement with the observation that oncobiosis supports metastasis formation in breast cancer [6, 7, 15]. Cadaverine, produced by LdcC, suppresses metastasis formation [6].
Earlier, metagenome-based reports have suggested that the breast cancer oncobiome changes as a function of stage [6, 7], estrogen, and HER2 receptor status [1, 2, 10]. Therefore, the association of oncobiome changes with lobular breast cancer or with E-cadherin status is not surprising. However, to the best of our knowledge, breast cancer oncobiome changes have not associated with E-cadherin expression in breast cancer.
What do these data suggest to us from a functional perspective? The literature suggests that the biosynthetic or “bioconversion” capacity of the oncobiome in breast cancer is drastically suppressed [10, 31–35]. Bacterial estrogen reactivation, through the bacterial estrobolome, is upregulated in breast cancer patients. Increased estrogen reactivation and reuptake supports the proliferation of breast cancer cells [3, 4, 33, 35]. Cytostatic bacterial metabolites were also identified, such as short chain fatty acids [10], lithocholic acid [7, 16, 36] and cadaverine [6]. LdcC plays role in cadaverine biosynthesis. LdcC is downregulated in E-cadherin negative cases that have a higher risk for metastasis formation. These observations support the idea that oncobiotic transformation in breast cancer downregulates the production of cytostatic and antimetastatic metabolites and, hence, supports metastasis formation in humans [6, 7, 15]. The bacterial metabolome-driven metastatic events are likely to be parallel mechanisms to the pathways regulated by the loss of E-cadherin or other adhesion proteins.
We are approaching a better understanding of the pathological role of the oncobiome and the oncobiotic transformation in breast cancer. This study provides evidence that the low bacterial production of cytostatic/antimetastatic metabolites in human breast cancer supports metastasis formation in breast cancer. Furthermore, the results of the DNA-based studies suggesting differences in the microbiome in subtypes of breast cancer [1, 2, 6, 7, 35] can be translated into changes in the proteome and, most probably, to changes in the bacterial metabolome as well. From a therapeutic perspective, these data support the applicability of bacterial metabolites in suppressing or preventing breast cancer metastasis.
Perspectives
Oncobiosis characterizes most cancers [37, 38]. Breast cancer oncobiosis affects the distal gut [1–7], oral [9], urinary [9] and the breast's own microbiome [8]. Apparently, changes to these microbiome compartments can increase the risk for developing breast cancer. Breast cancer cases in women peak around the menopause, hence, aging is a risk factor for breast cancer. Aging comes along with dysbiosys [39, 40] suggesting a likely causative relationship between changes to the microbiome and increased risk to breast cancer. Aging-associated, low grade inflammation is held responsible for the deterioration of multiple organs and organ systems [39–43]. Inflammation may in one hand damage cells and lead to mutations and cellular dysfunction, eventually to cell death [44]. Furthermore, constant low grade inflammation may be a pathway for the aging-associated simplification of the microbiome [39]. These changes are then translated to changes to the microbial metabolome [45] that may in turn give an age-related pathway for breast carcinogenesis.
Data availability
All primary data are available at https://figshare.com/s/bc183edf92224c293f46 (DOI: 10.6084/m9.figshare.12017550).
Conflict of interest
The authors declare no conflict of interest.
Acknowledgment
We are grateful for Ms. Emese Boda, Mr. László Finta, Mr. András Nagylaki and Mr. László Bancsi for the technical assistance.
Our work was supported by grants from the NKFIH (K123975 and GINOP-2.3.2-15-2016-00006 to PB, PD124110 and FK128387 to EM, EFOP-3.6.2-16-2017-0006 and K120669 to KU). EM is supported by the ÚNKP-19-4-DE-79 New National Excellence Program of the Ministry of Human Capacities. The research was financed by the Higher Education Institutional Excellence Programme (NKFIH-1150-6/2019) of the Ministry of Innovation and Technology in Hungary, within the framework of the Biotechnology thematic programme of the University of Debrecen. EM was supported by a Bolyai fellowship from the Hungarian Academy of Sciences.
References
- 1.
Banerjee S, Tian T, Wei Z, Shih N, Feldman MD, Peck KN, et al.. Distinct microbial signatures associated with different breast cancer types. Front Microbiol 2018; 9: 951.
- 2.
Banerjee S, Wei Z, Tan F, Peck KN, Shih N, Feldman M, et al.. Distinct microbiological signatures associated with triple negative breast cancer. Sci Rep 2015; 5: 15162.
- 3.
Goedert JJ, Hua X, Bielecka A, Okayasu I, Milne GL, Jones GS, et al.. Postmenopausal breast cancer and oestrogen associations with the IgA-coated and IgA-noncoated faecal microbiota. Br J Cancer 2018; 23: 435.
- 4.
Goedert JJ, Jones G, Hua X, Xu X, Yu G, Flores R, et al.. Investigation of the association between the fecal microbiota and breast cancer in postmenopausal women: a population-based case-control pilot study. J Natl Cancer Inst 2015; 107: djv147.
- 5.
Kovacs T, Miko E, Ujlaki G, Sari Z, Bai P. The microbiome as a component of the tumor microenvironment. Adv Exp Med Biol 2020; 1225: 137–53.
- 6.↑
Kovács T, Mikó E, Vida A, Sebő É, Toth J, Csonka T, et al.. Cadaverine, a metabolite of the microbiome, reduces breast cancer aggressiveness through trace amino acid receptors. Sci Rep 2019; 9: 1300.
- 7.
Miko E, Vida A, Kovacs T, Ujlaki G, Trencsenyi G, Marton J, et al.. Lithocholic acid, a bacterial metabolite reduces breast cancer cell proliferation and aggressiveness. Biochim Biophys Acta 2018; 1859: 958–74.
- 8.↑
Urbaniak C, Cummins J, Brackstone M, Macklaim JM, Gloor GB, Baban CK, et al.. Microbiota of human breast tissue. Appl Environ Microbiol 2014; 80: 3007–14.
- 9.↑
Wang H, Altemus J, Niazi F, Green H, Calhoun BC, Sturgis C, et al.. Breast tissue, oral and urinary microbiomes in breast cancer. Oncotarget 2017; 8: 88122–38.
- 10.↑
Miko E, Kovacs T, Sebo E, Toth J, Csonka T, Ujlaki G, et al.. Microbiome-microbial metabolome-cancer cell interactions in breast cancer-familiar, but unexplored. Cells 2019; 8(4): E293.
- 11.
Kirkup B, McKee A, Makin K, Paveley J, Caim S, Alcon-Giner C, et al.. Perturbation of the gut microbiota by antibiotics results in accelerated breast tumour growth and metabolic dysregulation. bioRxiv 553602 2019. https://doi.org/10.1101/553602.
- 12.
Kirkup BM, McKee AM, Madgwick M, Price CA, Dreger SA, Makin KA, et al.. Antibiotic-induced disturbances of the gut microbiota result in accelerated breast tumour growth via a mast cell-dependent pathway. bioRxiv 2020.2003.2007.982108 2020. https://doi.org/10.1101/2020.03.07.982108.
- 13.↑
Velicer CM, Heckbert SR, Lampe JW, Potter JD, Robertson CA, Taplin SH. Antibiotic use in relation to the risk of breast cancer. JAMA 2004; 291: 827–35.
- 14.↑
Aragon F, Carino S, Perdigon G, de Moreno de LeBlanc A. The administration of milk fermented by the probiotic Lactobacillus casei CRL 431 exerts an immunomodulatory effect against a breast tumour in a mouse model. Immunobiology 2014; 219: 457–64.
- 15.↑
Buchta Rosean C, Bostic RR, Ferey JCM, Feng TY, Azar FN, Tung KS, et al.. Preexisting commensal dysbiosis is a host-intrinsic regulator of tissue inflammation and tumor cell dissemination in hormone receptor-positive breast cancer. Cancer Res 2019; 79: 3662–75.
- 16.
Kovács P, Csonka T, Kovács T, Sári Z, Ujlaki G, Sipos A, et al.. Lithocholic acid, a metabolite of the microbiome, increases oxidative stress in breast cancer. Cancers (Basel) 2019; 11: 1255. https://doi.org/10.3390/cancers11091255.
- 17.
Miller-Fleming L, Olin-Sandoval V, Campbell K, Ralser M. Remaining mysteries of molecular biology: the role of polyamines in the cell. J Mol Biol 2015; 427: 3389–406.
- 18.
Seiler N. Catabolism of polyamines. Amino Acids 2004; 26: 217–33.
- 19.↑
de las Rivas B, Marcobal A, Carrascosa AV, Munoz R. PCR detection of foodborne bacteria producing the biogenic amines histamine, tyramine, putrescine, and cadaverine. J Food Prot 2006; 69: 2509–14.
- 21.
Liberles SD. Trace amine-associated receptors: ligands, neural circuits, and behaviors. Curr Opin Neurobiol 2015; 34: 1–7.
- 22.↑
Vattai A, Akyol E, Kuhn C, Hofmann S, Heidegger H, von Koch F, et al.. Increased trace amine-associated receptor 1 (TAAR1) expression is associated with a positive survival rate in patients with breast cancer. J Cancer Res Clin Oncol 2017; 13: 017-2420.
- 24.↑
Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez J-C, et al.. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics 2011; 12: 77.
- 25.↑
Gould Rothberg BE, Bracken MB. E-cadherin immunohistochemical expression as a prognostic factor in infiltrating ductal carcinoma of the breast: a systematic review and meta-analysis. Breast Cancer Res Treat 2006; 100: 139–48.
- 26.↑
Parker C, Rampaul RS, Pinder SE, Bell JA, Wencyk PM, Blamey RW, et al.. E-cadherin as a prognostic indicator in primary breast cancer. Br J Cancer 2001; 85: 1958–63.
- 27.
Rakha EA, Abd El Rehim D, Pinder SE, Lewis SA, Ellis IO. E-cadherin expression in invasive non-lobular carcinoma of the breast and its prognostic significance. Histopathology 2005; 46: 685–93.
- 28.
Auslander N, Yizhak K, Weinstock A, Budhu A, Tang W, Wang XW, et al.. A joint analysis of transcriptomic and metabolomic data uncovers enhanced enzyme-metabolite coupling in breast cancer. Sci Rep 2016; 6: 29662.
- 29.
Tang X, Lin CC, Spasojevic I, Iversen ES, Chi JT, Marks JR. A joint analysis of metabolomics and genetics of breast cancer. Breast Cancer Res 2014; 16: 415.
- 30.↑
Lombaerts M, van Wezel T, Philippo K, Dierssen JW, Zimmerman RM, Oosting J, et al.. E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines. Br J Cancer 2006; 94: 661–71.
- 31.
Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: physiological and clinical implications. Maturitas 2017; 103: 45–53.
- 32.
Ervin SM, Li H, Lim L, Roberts LR, Liang X, Mani S, et al.. Gut microbial beta-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. J Biol Chem 2019; 294: 18586–99.
- 33.
Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, et al.. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. J Transl Med 2012; 10: 253.
- 34.
Komorowski AS, Pezo RC. Untapped “-omics”: the microbial metagenome, estrobolome, and their influence on the development of breast cancer and response to treatment. Breast Cancer Res Treat 2020; 179: 287–300. https://doi.org/10.1007/s10549-10019-05472-w.
- 35.
Kwa M, Plottel CS, Blaser MJ, Adams S. The intestinal microbiome and estrogen receptor-positive female breast cancer. J Natl Cancer Inst 2016; 108(8): djw029.
- 36.
Goldberg AA, Beach A, Davies GF, Harkness TA, Leblanc A, Titorenko VI. Lithocholic bile acid selectively kills neuroblastoma cells, while sparing normal neuronal cells. Oncotarget 2011; 2: 761–82.
- 37.
Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA. The microbiome, cancer, and cancer therapy. Nat Med 2019; 25: 377–88.
- 38.
Kiss B, Mikó E, Sebő É, Toth J, Ujlaki G, Szabó J, et al.. Oncobiosis and microbial metabolite signaling in pancreatic adenocarcinoma. Cancers (Basel) 2020; 12: E1068. https://doi.org/10.3390/cancers12051068.
- 39.↑
Buford TW, Carter CS, VanDerPol WJ, Chen D, Lefkowitz EJ, Eipers P, et al.. Composition and richness of the serum microbiome differ by age and link to systemic inflammation. Geroscience 2018; 40: 257–68.
- 40.
Singh H, Torralba MG, Moncera KJ, DiLello L, Petrini J, Nelson KE, et al.. Gastro-intestinal and oral microbiome signatures associated with healthy aging. Geroscience 2019; 41: 907–21.
- 41.
An JY, Darveau R, Kaeberlein M. Oral health in geroscience: animal models and the aging oral cavity. Geroscience 2018; 40: 1–10.
- 42.
An JY, Quarles EK, Mekvanich S, Kang A, Liu A, Santos D, et al.. Rapamycin treatment attenuates age-associated periodontitis in mice. Geroscience 2017; 39: 457–63.
- 43.
Kavanagh K, Hsu FC, Davis AT, Kritchevsky SB, Rejeski WJ, Kim S. Biomarkers of leaky gut are related to inflammation and reduced physical function in older adults with cardiometabolic disease and mobility limitations. Geroscience 2019; 41: 923–33.
- 44.↑
Ungvari Z, Tarantini S, Nyul-Toth A, Kiss T, Yabluchanskiy A, Csipo T, et al.. Nrf2 dysfunction and impaired cellular resilience to oxidative stressors in the aged vasculature: from increased cellular senescence to the pathogenesis of age-related vascular diseases. Geroscience 2019; 41: 727–38.
- 45.↑
Johnson LC, Parker K, Aguirre BF, Nemkov TG, D'Alessandro A, Johnson SA, et al.. The plasma metabolome as a predictor of biological aging in humans. Geroscience 2019; 41: 895–906.