Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Cell Host Microbe ; 32(6): 925-944.e10, 2024 Jun 12.
Article En | MEDLINE | ID: mdl-38754417

Hormones and neurotransmitters are essential to homeostasis, and their disruptions are connected to diseases ranging from cancer to anxiety. The differential reactivation of endobiotic glucuronides by gut microbial ß-glucuronidase (GUS) enzymes may influence interindividual differences in the onset and treatment of disease. Using multi-omic, in vitro, and in vivo approaches, we show that germ-free mice have reduced levels of active endobiotics and that distinct gut microbial Loop 1 and FMN GUS enzymes drive hormone and neurotransmitter reactivation. We demonstrate that a range of FDA-approved drugs prevent this reactivation by intercepting the catalytic cycle of the enzymes in a conserved fashion. Finally, we find that inhibiting GUS in conventional mice reduces free serotonin and increases its inactive glucuronide in the serum and intestines. Our results illuminate the indispensability of gut microbial enzymes in sustaining endobiotic homeostasis and indicate that therapeutic disruptions of this metabolism promote interindividual response variabilities.


Gastrointestinal Microbiome , Glucuronidase , Homeostasis , Animals , Gastrointestinal Microbiome/drug effects , Mice , Glucuronidase/metabolism , Mice, Inbred C57BL , Serotonin/metabolism , Glucuronides/metabolism , Humans , Intestines/microbiology , Male , Germ-Free Life
2.
Cell Chem Biol ; 30(11): 1402-1413.e7, 2023 11 16.
Article En | MEDLINE | ID: mdl-37633277

Indoxyl sulfate is a microbially derived uremic toxin that accumulates in late-stage chronic kidney disease and contributes to both renal and cardiovascular toxicity. Indoxyl sulfate is generated by the metabolism of indole, a compound created solely by gut microbial tryptophanases. Here, we characterize the landscape of tryptophanase enzymes in the human gut microbiome and find remarkable structural and functional similarities across diverse taxa. We leverage this homology through a medicinal chemistry campaign to create a potent pan-inhibitor, (3S) ALG-05, and validate its action as a transition-state analog. (3S) ALG-05 successfully reduces indole production in microbial culture and displays minimal toxicity against microbial and mammalian cells. Mice treated with (3S) ALG-05 show reduced cecal indole and serum indoxyl sulfate levels with minimal changes in other tryptophan-metabolizing pathways. These studies present a non-bactericidal pan-inhibitor of gut microbial tryptophanases with potential promise for reducing indoxyl sulfate in chronic kidney disease.


Gastrointestinal Microbiome , Renal Insufficiency, Chronic , Humans , Mice , Animals , Indican/pharmacology , Indican/metabolism , Tryptophanase , Gastrointestinal Microbiome/physiology , Indoles/pharmacology , Indoles/metabolism , Renal Insufficiency, Chronic/drug therapy , Mammals/metabolism
3.
Gut Microbes ; 14(1): 2107289, 2022.
Article En | MEDLINE | ID: mdl-35953888

Mycophenolate mofetil (MMF) is an important immunosuppressant prodrug prescribed to prevent organ transplant rejection and to treat autoimmune diseases. MMF usage, however, is limited by severe gastrointestinal toxicity that is observed in approximately 45% of MMF recipients. The active form of the drug, mycophenolic acid (MPA), undergoes extensive enterohepatic recirculation by bacterial ß-glucuronidase (GUS) enzymes, which reactivate MPA from mycophenolate glucuronide (MPAG) within the gastrointestinal tract. GUS enzymes demonstrate distinct substrate preferences based on their structural features, and gut microbial GUS enzymes that reactivate MPA have not been identified. Here, we compare the fecal microbiomes of transplant recipients receiving MMF to healthy individuals using shotgun metagenomic sequencing. We find that neither microbial composition nor the presence of specific structural classes of GUS genes are sufficient to explain the differences in MPA reactivation measured between fecal samples from the two cohorts. We next employed a GUS-specific activity-based chemical probe and targeted metaproteomics to identify and quantify the GUS proteins present in the human fecal samples. The identification of specific GUS enzymes was improved by using the metagenomics data collected from the fecal samples. We found that the presence of GUS enzymes that bind the flavin mononucleotide (FMN) is significantly correlated with efficient MPA reactivation. Furthermore, structural analysis identified motifs unique to these FMN-binding GUS enzymes that provide molecular support for their ability to process this drug glucuronide. These results indicate that FMN-binding GUS enzymes may be responsible for reactivation of MPA and could be a driving force behind MPA-induced GI toxicity.


Gastrointestinal Microbiome , Flavin Mononucleotide , Gastrointestinal Microbiome/physiology , Glucuronides , Humans , Immunosuppressive Agents , Mycophenolic Acid/therapeutic use , Proteomics
4.
ACS Infect Dis ; 7(7): 1969-1984, 2021 07 09.
Article En | MEDLINE | ID: mdl-33765392

Neisseria gonorrhoeae is an urgent threat to public health in the United States and around the world. Many of the current classes of antibiotics to treat N. gonorrhoeae infection are quickly becoming obsolete due to increased rates of resistance. Thus, there is a critical need for alternative antimicrobial targets and new chemical entities. Our team has repurposed the FDA-approved carbonic anhydrase inhibitor scaffold of acetazolamide to target N. gonorrhoeae and the bacteria's essential carbonic anhydrase, NgCA. This study established both structure-activity and structure-property relationships that contribute to both antimicrobial activity and NgCA activity. This ultimately led to molecules 20 and 23, which displayed minimum inhibitory concentration values as low as 0.25 µg/mL equating to an 8- to 16-fold improvement in antigonococcal activity compared to acetazolamide. These analogues were determined to be bacteriostatic against the pathogen and likely on-target against NgCA. Additionally, they did not exhibit any detrimental effects in cellular toxicity assays against both a human endocervical (End1/E6E7) cell line or colorectal adenocarcinoma cell line (Caco-2) at concentrations up to 128 µg/mL. Taken together, this study presents a class of antigonococcal agents with the potential to be advanced for further evaluation in N. gonorrhoeae infection models.


Carbonic Anhydrase Inhibitors , Neisseria gonorrhoeae , Acetazolamide/pharmacology , Caco-2 Cells , Carbonic Anhydrase Inhibitors/pharmacology , Humans , Structure-Activity Relationship
5.
Toxins (Basel) ; 12(9)2020 09 11.
Article En | MEDLINE | ID: mdl-32932981

Chronic kidney disease (CKD) afflicts more than 500 million people worldwide and is one of the fastest growing global causes of mortality. When glomerular filtration rate begins to fall, uremic toxins accumulate in the serum and significantly increase the risk of death from cardiovascular disease and other causes. Several of the most harmful uremic toxins are produced by the gut microbiota. Furthermore, many such toxins are protein-bound and are therefore recalcitrant to removal by dialysis. We review the derivation and pathological mechanisms of gut-derived, protein-bound uremic toxins (PBUTs). We further outline the emerging relationship between kidney disease and gut dysbiosis, including the bacterial taxa altered, the regulation of microbial uremic toxin-producing genes, and their downstream physiological and neurological consequences. Finally, we discuss gut-targeted therapeutic strategies employed to reduce PBUTs. We conclude that targeting the gut microbiota is a promising approach for the treatment of CKD by blocking the serum accumulation of PBUTs that cannot be eliminated by dialysis.


Bacteria/metabolism , Bacterial Toxins/metabolism , Gastrointestinal Microbiome , Renal Insufficiency, Chronic/microbiology , Uremia/microbiology , Animals , Disease Progression , Dysbiosis , Humans , Protein Binding , Renal Dialysis , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/therapy , Uremia/metabolism , Uremia/therapy
6.
J Med Chem ; 63(17): 9540-9562, 2020 09 10.
Article En | MEDLINE | ID: mdl-32787141

Vancomycin-resistant enterococci (VRE) are the second leading cause of hospital-acquired infections (HAIs) attributed to a drug-resistant bacterium in the United States, and resistance to the frontline treatments is well documented. To combat VRE, we have repurposed the FDA-approved carbonic anhydrase drug acetazolamide to design potent antienterococcal agents. Through structure-activity relationship optimization we have arrived at two leads possessing improved potency against clinical VRE strains from MIC = 2 µg/mL (acetazolamide) to MIC = 0.007 µg/mL (22) and 1 µg/mL (26). Physicochemical properties were modified to design leads that have either high oral bioavailability to treat systemic infections or low intestinal permeability to treat VRE infections in the gastrointestinal tract. Our data suggest the intracellular targets for the molecules are putative α-carbonic and γ-carbonic anhydrases, and homology modeling and molecular dynamics simulations were performed. Together, this study presents potential anti-VRE therapeutic options to provide alternatives for problematic VRE infections.


Acetazolamide/chemistry , Acetazolamide/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Design , Vancomycin-Resistant Enterococci/drug effects , Acetazolamide/pharmacokinetics , Acetazolamide/toxicity , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/toxicity , Caco-2 Cells , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Humans , Mice , Microbial Sensitivity Tests , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Tissue Distribution
...