12 results on '"Tahliyah S. Mims"'
Search Results
2. Circulating Microbiota in Cardiometabolic Disease
- Author
-
Keiichi Sumida, Zhongji Han, Chi-Yang Chiu, Tahliyah S. Mims, Amandeep Bajwa, Ryan T. Demmer, Susmita Datta, Csaba P. Kovesdy, and Joseph F. Pierre
- Subjects
cardiometabolic disease ,cardiovascular disease ,chronic kidney disease ,circulating microbiota ,diabetes mellitus ,end-stage kidney disease ,Microbiology ,QR1-502 - Abstract
The rapid expansion of microbiota research has significantly advanced our understanding of the complex interactions between gut microbiota and cardiovascular, metabolic, and renal system regulation. Low-grade chronic inflammation has long been implicated as one of the key mechanisms underlying cardiometabolic disease risk and progression, even before the insights provided by gut microbiota research in the past decade. Microbial translocation into the bloodstream can occur via different routes, including through the oral and/or intestinal mucosa, and may contribute to chronic inflammation in cardiometabolic disease. Among several gut-derived products identifiable in the systemic circulation, bacterial endotoxins and metabolites have been extensively studied, however recent advances in microbial DNA sequencing have further allowed us to identify highly diverse communities of microorganisms in the bloodstream from an -omics standpoint, which is termed “circulating microbiota.” While detecting microorganisms in the bloodstream was historically considered as an indication of infection, evidence on the circulating microbiota is continually accumulating in various patient populations without clinical signs of infection and even in otherwise healthy individuals. Moreover, both quantitative and compositional alterations of the circulating microbiota have recently been implicated in the pathogenesis of chronic inflammatory conditions, potentially through their immunostimulatory, atherogenic, and cardiotoxic properties. In this mini review, we aim to provide recent evidence on the characteristics and roles of circulating microbiota in several cardiometabolic diseases, such as type 2 diabetes, cardiovascular disease, and chronic kidney disease, with highlights of our emerging findings on circulating microbiota in patients with end-stage kidney disease undergoing hemodialysis.
- Published
- 2022
- Full Text
- View/download PDF
3. Early Life Microbiome Colonization and Human Health
- Author
-
Tahliyah S. Mims, Jun Miyoshi, and Joseph F. Pierre
- Abstract
The early life assemblages of microbial community membership, diversity, and function within the gastrointestinal tract play fundamental roles in shaping host growth, body composition, and organ development and function, including the programming of immune systems, shaping central nervous system signaling and behaviour, and influencing metabolic set points. Abnormal colonization during this period may elevate the risk of chronic and metabolic disease throughout the lifespan. Research on the influence of microbial communities on neonatal health has historically been focused on the prevention and treatment of infectious disease during the neonatal and early adolescent period. Over the last decade, deeper insights into the role of normal human microbiome colonization in early life and childhood has grown substantially and strong evidence for its importance in human development has accumulated. Acquisition of an early life microbiome is influenced by the route of birth, nutrition composition and source, antibiotic and toxin exposures, geological location and home environment, and other pivotal factors. The microbiome appears to be dynamically developed during the first year of life but becomes more stabilized once community diversity and membership increases, along with compensatory and complementary host immune response. During microbial community assemblage, it is proposed that critical developmental immune windows are shaped that influence host homeostasis and disease risk in later life. This chapter will review this rapidly developing field and highlight the role of the microbiome in early life along with implications for long-term health during the lifespan.
- Published
- 2022
- Full Text
- View/download PDF
4. Dietary tryptophan metabolite released by intratumoral Lactobacillus reuteri facilitates immune checkpoint inhibitor treatment
- Author
-
Mackenzie J. Bender, Alex C. McPherson, Catherine M. Phelps, Surya P. Pandey, Colin R. Laughlin, Jake H. Shapira, Luzmariel Medina Sanchez, Mohit Rana, Tanner G. Richie, Tahliyah S. Mims, Angela M. Gocher-Demske, Luisa Cervantes-Barragan, Steven J. Mullett, Stacy L. Gelhaus, Tullia C. Bruno, Nikki Cannon, John A. McCulloch, Dario A.A. Vignali, Reinhard Hinterleitner, Alok V. Joglekar, Joseph F. Pierre, Sonny T.M. Lee, Diwakar Davar, Hassane M. Zarour, and Marlies Meisel
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
5. TGR5 signaling mitigates parenteral nutrition-associated liver disease
- Author
-
Kent A. Willis, Erin J. Stephenson, Joan C. Han, Eugene B. Chang, Tahliyah S. Mims, Michelle Puchowicz, Joseph F. Pierre, Charles K. Gomes, Qusai Al Abdallah, Dengping Yin, Dejan Micic, E. Richard Moran, Ankush Gosain, Prahlad K. Rao, and Ajay J. Talati
- Subjects
Male ,0301 basic medicine ,Parenteral Nutrition ,medicine.medical_specialty ,Physiology ,Gastroenterology ,Receptors, G-Protein-Coupled ,Bile Acids and Salts ,Mice ,03 medical and health sciences ,Liver disease ,0302 clinical medicine ,Liver Function Tests ,Cholestasis ,Physiology (medical) ,Internal medicine ,Animals ,Humans ,Medicine ,In patient ,Microbiome ,Mice, Knockout ,Hepatology ,Interleukin-6 ,business.industry ,Liver Diseases ,Infant, Newborn ,Organ Size ,medicine.disease ,G protein-coupled bile acid receptor ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,Prolonged exposure ,030104 developmental biology ,Parenteral nutrition ,Gene Expression Regulation ,Female ,030211 gastroenterology & hepatology ,business ,Signal Transduction - Abstract
Bile acid receptors regulate the metabolic and immune functions of circulating enterohepatic bile acids. This process is disrupted by administration of parenteral nutrition (PN), which may induce progressive hepatic injury for unclear reasons, especially in the newborn, leading to PN-associated liver disease. To explore the role of bile acid signaling on neonatal hepatic function, we initially observed that Takeda G protein receptor 5 (TGR5)-specific bile acids were negatively correlated with worsening clinical disease markers in the plasma of human newborns with prolonged PN exposure. To test our resulting hypothesis that TGR5 regulates critical liver functions to PN exposure, we used TGR5 receptor deficient mice (TGR5−/−). We observed PN significantly increased liver weight, cholestasis, and serum hepatic stress enzymes in TGR5−/− mice compared with controls. Mechanistically, PN reduced bile acid synthesis genes in TGR5−/−. Serum bile acid composition revealed that PN increased unconjugated primary bile acids and secondary bile acids in TGR5−/− mice, while increasing conjugated primary bile acid levels in TGR5-competent mice. Simultaneously, PN elevated hepatic IL-6 expression and infiltrating macrophages in TGR5−/− mice. However, the gut microbiota of TGR5−/− mice compared with WT mice following PN administration displayed highly elevated levels of Bacteroides and Parabacteroides, and possibly responsible for the elevated levels of secondary bile acids in TGR5−/− animals. Intestinal bile acid transporters expression was unchanged. Collectively, this suggests TGR5 signaling specifically regulates fundamental aspects of liver bile acid homeostasis during exposure to PN. Loss of TGR5 is associated with biochemical evidence of cholestasis in both humans and mice on PN. NEW & NOTEWORTHY Parenteral nutrition is associated with deleterious metabolic outcomes in patients with prolonged exposure. Here, we demonstrate that accelerated cholestasis and parental nutrition-associated liver disease (PNALD) may be associated with deficiency of Takeda G protein receptor 5 (TGR5) signaling. The microbiome is responsible for production of secondary bile acids that signal through TGR5. Therefore, collectively, these data support the hypothesis that a lack of established microbiome in early life or under prolonged parenteral nutrition may underpin disease development and PNALD.
- Published
- 2020
- Full Text
- View/download PDF
6. SMAD2 and SMAD3 differentially regulate adiposity and the growth of subcutaneous white adipose tissue
- Author
-
Michelle Puchowicz, Joseph F. Pierre, Tamekia L. Jones, Roshan Kumari, Chester W. Brown, Qusai Al Abdallah, Maria Johnson Irudayam, and Tahliyah S. Mims
- Subjects
Male ,medicine.medical_specialty ,Adipose Tissue, White ,Subcutaneous Fat ,Adipokine ,Adipose tissue ,Smad2 Protein ,White adipose tissue ,Intra-Abdominal Fat ,Biology ,Diet, High-Fat ,Biochemistry ,Mice ,chemistry.chemical_compound ,Adipocyte ,Internal medicine ,Conditional gene knockout ,Genetics ,medicine ,Animals ,Smad3 Protein ,Molecular Biology ,Adiposity ,Mice, Knockout ,Adipogenesis ,Cell Differentiation ,Activins ,Mice, Inbred C57BL ,Endocrinology ,chemistry ,Knockout mouse ,Female ,medicine.symptom ,Weight gain ,Biotechnology - Abstract
Adipose tissue is the primary site of energy storage, playing important roles in health. While adipose research largely focuses on obesity, fat also has other critical functions, producing adipocytokines and contributing to normal nutrient metabolism, which in turn play important roles in satiety and total energy homeostasis. SMAD2/3 proteins are downstream mediators of activin signaling, which regulate critical preadipocyte and mature adipocyte functions. Smad2 global knockout mice exhibit embryonic lethality, whereas global loss of Smad3 protects mice against diet-induced obesity. The direct contributions of Smad2 and Smad3 in adipose tissues, however, are unknown. Here, we sought to determine the primary effects of adipocyte-selective reduction of Smad2 or Smad3 on diet-induced adiposity using Smad2 or Smad3 "floxed" mice intercrossed with Adiponectin-Cre mice. Additionally, we examined visceral and subcutaneous preadipocyte differentiation efficiency in vitro. Almost all wild type subcutaneous preadipocytes differentiated into mature adipocytes. In contrast, visceral preadipocytes differentiated poorly. Exogenous activin A suppressed differentiation of preadipocytes from both depots. Smad2 conditional knockout (Smad2cKO) mice did not exhibit significant effects on weight gain, irrespective of diet, whereas Smad3 conditional knockout (Smad3cKO) male mice displayed a trend of reduced body weight on high-fat diet. On both diets, Smad3cKO mice displayed an adipose depot-selective phenotype, with a significant reduction in subcutaneous fat mass but not visceral fat mass. Our data suggest that Smad3 is an important contributor to the maintenance of subcutaneous white adipose tissue in a sex-selective fashion. These findings have implications for understanding SMAD-mediated, depot selective regulation of adipocyte growth and differentiation.
- Published
- 2021
- Full Text
- View/download PDF
7. Circulating Microbial Signatures and Cardiovascular Death in Patients With ESRD
- Author
-
Zhongji Han, Praveen K. Potukuchi, Ryan T. Demmer, Joseph F. Pierre, Keiichi Sumida, Melana Yuzefpolskaya, Csaba P. Kovesdy, Susmita Datta, Tahliyah S. Mims, and Paolo C. Colombo
- Subjects
end-stage renal disease ,business.industry ,medicine.medical_treatment ,Physiology ,circulating microbiome ,Disease ,Odds ratio ,Ribosomal RNA ,mortality ,End stage renal disease ,Pathogenesis ,Nephrology ,Clinical Research ,cardiovascular disease ,inflammation ,Medicine ,Hemodialysis ,Microbiome ,Internal transcribed spacer ,business ,chronic kidney disease - Abstract
Introduction Patients with end-stage renal disease (ESRD) experience disproportionately high cardiovascular morbidity and mortality. Accumulating evidence suggests a role for the circulating microbiome in the pathogenesis of cardiovascular disease; however, little is known about its association with premature cardiovascular mortality in ESRD. Methods In a pilot case-control study of 17 hemodialysis patients who died of a cardiovascular event and 17 matched hemodialysis controls who remained alive during a median follow-up of 2.0 years, we compared the levels and composition of circulating microbiome, including Bacteria, Archaea, and Fungi, in serum samples by quantitative polymerase chain reaction and 16S or Internal Transcribed Spacer (ITS) ribosomal RNA (rRNA) sequencing, respectively. Associations of the circulating cell-free microbial signatures with clinical parameters and cardiovascular death were examined using the Spearman rank correlation and multivariable conditional logistic regression, respectively. Results Both 16S and ITS rRNA were detectable in all (except 3 for ITS) examined patients’ serum samples. Despite no significant difference in 16S rRNA levels and α diversity between cases and controls, taxonomic analysis demonstrated differential community membership between groups, with significantly greater Actinobacteria and less Proteobacteria observed in cases than in controls at the phylum level. Proportions of Actinobacteria and Proteobacteria phyla were significantly correlated with plasma nuclear factor erythroid 2−related factor 2 (Nrf2) levels (rho = −0.41 and 0.42, P = 0.015 and 0.013, respectively) and marginally associated with risk of cardiovascular death (adjusted odds ratios [95% confidence intervals] = 1.12 [0.98−1.29] and 0.88 [0.76−1.02] for 1% increase, respectively). Conclusion Alterations of the circulating cell-free microbial signatures may be associated with higher premature cardiovascular mortality in ESRD., Graphical abstract
- Published
- 2021
8. The gut mycobiome of healthy mice is shaped by the environment and correlates with metabolic outcomes in response to diet
- Author
-
Qusai Al Abdallah, Sydney P. Watts, Joan C. Han, Kent A. Willis, Thomas V. Rousselle, Tahliyah S. Mims, Justin D. Stewart, Joseph F. Pierre, Catrina T. White, Amandeep Bajwa, and Ankush Gosain
- Subjects
0301 basic medicine ,Male ,QH301-705.5 ,Food Handling ,Medicine (miscellaneous) ,Zoology ,Biology ,Gut flora ,Environment ,Saccharomyces ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Biology (General) ,Adiposity ,Bacteria ,Host (biology) ,Age Factors ,Fungi ,Metabolism ,medicine.disease ,biology.organism_classification ,Obesity ,Animal Feed ,Diet ,Gastrointestinal Microbiome ,Intestines ,Mice, Inbred C57BL ,030104 developmental biology ,Host-Pathogen Interactions ,Body Composition ,030211 gastroenterology & hepatology ,Female ,Host adaptation ,medicine.symptom ,Adaptation ,General Agricultural and Biological Sciences ,Energy Metabolism ,Weight gain ,Biomarkers - Abstract
As an active interface between the host and their diet, the gut microbiota influences host metabolic adaptation; however, the contributions of fungi have been overlooked. Here, we investigate whether variations in gut mycobiome abundance and composition correlate with key features of host metabolism. We obtained animals from four commercial sources in parallel to test if differing starting mycobiomes can shape host adaptation in response to processed diets. We show that the gut mycobiome of healthy mice is shaped by the environment, including diet, and significantly correlates with metabolic outcomes. We demonstrate that exposure to processed diet leads to persistent differences in fungal communities that significantly associate with differential deposition of body mass in male mice compared to mice fed standardized diet. Fat deposition in the liver, transcriptional adaptation of metabolically active tissues and serum metabolic biomarker levels are linked with alterations in fungal community diversity and composition. Specifically, variation in fungi from the genera Thermomyces and Saccharomyces most strongly associate with metabolic disturbance and weight gain. These data suggest that host–microbe metabolic interactions may be influenced by variability in the mycobiome. This work highlights the potential significance of the gut mycobiome in health and has implications for human and experimental metabolic studies. Tahliyah S. Mims et al. investigate the influence of the gut mycobiome abundance and composition on host metabolism. Using mice from four different commercial suppliers they find that the gut mycobiome is shaped by diet, and that abundance and composition correlate with key metabolic features. In particular they find that Thermomyces and Saccharomyces species most strongly associate with weight gain.
- Published
- 2020
9. The gut mycobiome of healthy mice is shaped by the environment and shapes metabolic outcomes in response to diet
- Author
-
Catrina T. White, Amandeep Bajwa, Kent A. Willis, Tahliyah S. Mims, Thomas V. Rousselle, Justin D. Stewart, Qusai Al Abdullah, Joan C. Han, Sydney P. Watts, and Joseph F. Pierre
- Subjects
biology ,Host (biology) ,medicine ,Physiology ,Bacteriome ,Internal transcribed spacer ,Adaptation ,Ribosomal RNA ,medicine.symptom ,biology.organism_classification ,Saccharomyces ,Weight gain ,Bacteria - Abstract
ObjectiveAs an active interface between the host and their diet, the gut bacteriome influences host metabolic adaptation. However, the contribution of gut fungi to host metabolic outcomes is not yet understood. Therefore, we aimed to determine if host metabolic response to an ultra-processed diet reflects gut fungal community composition.DesignWe compared jejunal fungi and bacteria from 72 healthy mice with the same genetic background but different starting mycobiomes before and after 8 weeks on an ultra-processed or standardized diet using 16S and internal transcribed spacer region 2 ribosomal RNA sequencing. We measured host body composition using magnetic resonance imaging, examined changes in metabolically active host tissues and quantified serum metabolic biomarkers.ResultsGut fungal communities are highly variable between mice, differing by vendor, age and sex. After exposure to an ultra-processed diet for 8 weeks, persistent differences in fungal community composition strongly associate with differential deposition of body mass in male mice compared to mice on standardized diet. Fat deposition in the liver, genomic adaptation of metabolically active tissues and serum metabolic biomarkers are correlated with alterations in fungal diversity and community composition. Variation in fungi from the genera Thermomyces and Saccharomyces most strongly associate with increased weight gain.ConclusionsIn the gut of healthy mice, host-microbe metabolic interactions strongly reflect variability in fungal communities. Our results confirm the importance of luminal fungal communities to host metabolic adaptation to dietary exposure. Gut fungal communities may represent a therapeutic target for the prevention and treatment of metabolic disease.Graphical AbstractIn BriefWhat is already known about this subject?Gut bacterial communities have evolved to influence the metabolic outcomes of the host in mammals. Evidence from across the lifespan suggests that differences in composition of these communities is associated with energy consumption. However, gut microbial communities, while often equated to bacteria, are diverse, multi-kingdom ecologies and limited information is available for the role of other kingdoms of life, such as fungi.What are the new findings?Gut fungal communities, collectively termed the mycobiome, are less diverse and abundant than bacterial communities in the gastrointestinal tract. This study identifies the considerable influence of the environment and dietary exposure on the composition of jejunal fungal communities in healthy mice with the same genetic background. After exposure to processed diet, differences in fungal community composition in male mice were strongly correlated with persistent differences body composition and markers of metabolic tone.How might it impact on clinical practice in the foreseeable future?These results verify that the baseline metabolic tone of health mice strongly reflects the ecological complexity of the gastrointestinal mycobiome. Variation in the composition of gut fungal communities is likely an underappreciated source of experimental and clinical variability in metabolic studies. Gastrointestinal fungi are likely a target for prevention and treatment of metabolic disease.
- Published
- 2020
- Full Text
- View/download PDF
10. Immune checkpoint blockade reprograms systemic immune landscape and tumor microenvironment in obesity-associated breast cancer
- Author
-
Hyo Young Choi, Tahliyah S. Mims, Alina M Hamilton, Bin Teng, Katherine L. Cook, D. Neil Hayes, Ajeeth K. Pingili, Emily J. Miller, Joseph F. Pierre, Radhika Sekhri, Deidre Daria, Heejoon Jo, Rahul Sharma, Laura M. Sipe, Ramesh Narayanan, Melissa A. Troester, Liza Makowski, Sarah Asemota, Johnathan R. Yarbro, Tony N. Marion, Mehdi Chaib, and Qusai Al Abdallah
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Breast Neoplasms ,Tumor-associated macrophage ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Obesity ,Immune Checkpoint Inhibitors ,Triple-negative breast cancer ,Immunosuppression Therapy ,Tumor microenvironment ,business.industry ,Macrophages ,Myeloid-Derived Suppressor Cells ,Immunosuppression ,Immunotherapy ,Immune checkpoint ,Gastrointestinal Microbiome ,Tumor Burden ,Mice, Inbred C57BL ,030104 developmental biology ,Receptors, Estrogen ,Tumor progression ,Disease Progression ,Cancer research ,Female ,business ,Spleen ,030217 neurology & neurosurgery - Abstract
SUMMARY Immune checkpoint blockade (ICB) has improved outcomes in some cancers. A major limitation of ICB is that most patients fail to respond, which is partly attributable to immunosuppression. Obesity appears to improve immune checkpoint therapies in some cancers, but impacts on breast cancer (BC) remain unknown. In lean and obese mice, tumor progression and immune reprogramming were quantified in BC tumors treated with anti-programmed death-1 (PD-1) or control. Obesity augments tumor incidence and progression. Anti-PD-1 induces regression in lean mice and potently abrogates progression in obese mice. BC primes systemic immunity to be highly responsive to obesity, leading to greater immunosuppression, which may explain greater anti-PD-1 efficacy. Anti-PD-1 significantly reinvigorates antitumor immunity despite persistent obesity. Laminin subunit beta-2 (Lamb2), downregulated by anti-PD-1, significantly predicts patient survival. Lastly, a microbial signature associated with anti-PD-1 efficacy is identified. Thus, anti-PD-1 is highly efficacious in obese mice by reinvigorating durable antitumor immunity., In brief Pingili et al. show that breast cancer exacerbates obesity-driven immunosuppression. Anti-PD-1 reinvigorates antitumor immunity in the tumor microenvironment, mammary fat pad, and peripherally. Lamb2, downregulated by anti-PD-1 in tumors, associates with obesity and poor survival in patients. A microbial signature associated with immune checkpoint inhibitor efficacy is identified., Graphical Abstract
- Published
- 2021
- Full Text
- View/download PDF
11. Intestinal fungi as regulators of obesity and glucose tolerance
- Author
-
Ajeeth K. Pingili, Tahliyah S. Mims, Joan C. Han, Aman Bajwa, Sydney P. Watts, Qusai Al Abdallah, Joseph F. Pierre, Liza Makowski, and Bin Teng
- Subjects
medicine.medical_specialty ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Biology ,medicine.disease ,Molecular Biology ,Biochemistry ,Obesity ,Biotechnology - Published
- 2020
- Full Text
- View/download PDF
12. Variability in interkingdom gut microbiomes between different commercial vendors shapes fat gain in response to diet
- Author
-
Sydney P. Watts, Qusai Al Abdallah, Catrina T. White, Joan C. Han, Joseph F. Pierre, Tahliyah S. Mims, and Kent A. Willis
- Subjects
Genetics ,Zoology ,Microbiome ,Biology ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.