Spatial Metabolomics

Metabolomics: Revolutionizing Healthcare and Research with Advanced Analytical Techniques

February 22, 2024 Off By admin
Shares

Metabolomics is the scientific study of the unique chemical fingerprints that specific cellular processes leave behind, known as metabolites. It involves the identification and quantification of these metabolites, which can provide valuable insights into the physiological state of an organism, tissue, or cell. Metabolomics has a wide range of applications in various fields of research, including toxicology, functional genomics, nutrigenomics, and disease diagnosis and treatment.

In toxicology, metabolic profiling of urine or blood can be used to assess toxicity and detect the presence of toxins. This can help pharmaceutical companies save significant funding on clinical trials by identifying potential drug candidates’ metabolic effects before they are made available to the public.

In functional genomics, metabolomics can be used to predict phenotypes resulting from genetic manipulation. By understanding the behavior of the metabolome, researchers can predict the phenotypes that would present if a gene was deleted or inserted into the genome. This can be particularly useful in the study of genetically modified plant material for human consumption, as any changes that have the potential to alter the metabolome of the organisms that consume the genetically modified material may be predicted before they are made available for public consumption.

In nutrigenomics, metabolomics can be used to determine the metabolic fingerprint of an individual, which portrays the effect of endogenous and exogenous factors on the metabolism of the individual. This can help in personalized nutrition and lifestyle recommendations.

In disease diagnosis and treatment, metabolomics can help identify the pathophysiological processes of disease and mechanisms that can be targeted to manage the disease. Metabolomics biomarkers in tissue samples or biopsies can be used to categorize and stage the progression of cancers, providing valuable information for appropriate treatment decisions.

In conclusion, metabolomics is a promising field with a wide range of applications in various fields of research. Its ability to provide insights into the physiological state of an organism, tissue, or cell makes it a valuable tool in toxicology, functional genomics, nutrigenomics, and disease diagnosis and treatment. As the field continues to advance, it is likely that even more applications will be discovered, further highlighting the importance of staying updated with the latest advancements in metabolomics and its applications.

Recent Advancements in Metabolomics

Untargeted metabolomics techniques are used to identify new compounds and pathways in various fields of research. These techniques involve analyzing a broad range of metabolites in a sample without a predetermined list of metabolites to measure. Untargeted metabolomics can provide a comprehensive view of the metabolic state of a biological system, allowing for the discovery of novel metabolites and pathways that may be relevant to the study at hand.

One common technique used in untargeted metabolomics is liquid chromatography-mass spectrometry (LC-MS). This technique involves separating metabolites based on their chemical properties and then detecting and quantifying them using mass spectrometry. LC-MS can be used to detect a wide range of metabolites, including amino acids, lipids, carbohydrates, and nucleotides.

Another technique used in untargeted metabolomics is gas chromatography-mass spectrometry (GC-MS). This technique involves separating metabolites based on their volatility and then detecting and quantifying them using mass spectrometry. GC-MS can be used to detect a wide range of metabolites, including fatty acids, sugars, and organic acids.

Untargeted metabolomics techniques can be used to identify new compounds and pathways in various fields of research, including toxicology, functional genomics, nutrigenomics, and disease diagnosis and treatment. For example, in toxicology, untargeted metabolomics can be used to identify metabolic changes in response to toxins or drugs. In functional genomics, untargeted metabolomics can be used to identify metabolic changes in response to genetic manipulation. In nutrigenomics, untargeted metabolomics can be used to identify metabolic changes in response to dietary interventions. And in disease diagnosis and treatment, untargeted metabolomics can be used to identify metabolic changes associated with disease.

However, untargeted metabolomics techniques also have challenges, including the identification of metabolites. The range of confidence associated with identifications is often overlooked, and opportunities for advancing the metabolomics field include improving the accuracy of metabolite assignments. In-silico metabolite databases can provide guidance and in some cases validation, but will not fit all metabolomic studies. Validation of retention times and MS/MS fragmentation data with a reference standard is nearly always required for confident metabolite identification.

In conclusion, untargeted metabolomics techniques are a powerful tool for identifying new compounds and pathways in various fields of research. However, the accuracy of metabolite assignments is extremely important, and opportunities for advancing the metabolomics field include improving the accuracy of metabolite assignments. It is essential to use high-confidence identifications and validate them with reference standards to ensure accurate and reliable results.

Metabolomics is the scientific study of the unique chemical fingerprints that specific cellular processes leave behind. This field has the potential to be applied in various settings to offer practical use, including diagnosing diseases, drug discovery, food and environmental studies, and functional genomics.

In diagnosing diseases, metabolic profiling of urine or blood can be utilized for the assessment of toxicity. Several techniques can detect physiological changes in the physiological sample that results from the presence of a toxin or toxins. The information revealed by way of metabolic profiling can also be related to a specific health condition or syndrome, such as a lesion in the liver or kidney. Pharmaceutical companies have expressed interest in this field because the ability to test the toxicity of potential drug candidates via its metabolic effects has the potential to make significant savings in the funding needed for clinical trials before a new drug is opened to the public.

Metabolomics could be very useful in researching the phenotypes that may result from a certain genetic manipulation in the field of functional genomics. For example, sufficient knowledge about the behavior of the metabolome could help to predict the phenotypes that would present if a gene was deleted or inserted into the genome. The detection of phenotypic changes can be applied in a number of practical settings. As a prime example, genetically modified plant material for human consumption can be examined for phenotypic changes. Any changes that have the potential to alter the metabolome of the organisms that consume the genetically modified material may be predicted before they are made available for public consumption. Additionally, metabolomics could make it possible to predict the function of unknown genes, by way of comparing metabolic perturbations caused by the modifications of known genes. Model organisms of Saccharomyces cerevisiae and Arabidopsis thaliana are currently being undertaken and could lead to such advances in the future.

In drug discovery, metabolomics can help to identify the pathophysiological processes of disease and mechanisms that can be targeted to manage the disease. For example, metabolomics biomarkers in tissue samples or biopsies can be used to categorize and stage the progression of cancers. The information can then be used to guide the appropriate decisions for treatment. Metabolomics can also be applied to characterize the ways in which an organism interacts with its environment. Studying these environmental interactions and assessing the function and health of an organism at a molecule level can reveal useful information about the effect of environment on an organism’s health. This can also be applied to a wider population to provide data for other fields of research, such as ecology.

In food and environmental studies, metabolomics can be applied to determine the metabolic fingerprint of an individual, which portrays the effect of the endogenous and exogenous factors in the body on the metabolism of the individual. Nutrigenomics combines the knowledge obtained from genomics, transcriptomics, proteomics and metabolomics with nutritional principles for humans. A metabolome of any body fluid depends on various endogenous factors including the individual’s age, gender, body composition, genetic susceptibilities, and concurrent health conditions, as well as exogenous factors such as nutrients, other components of food and medications.

In conclusion, metabolomics has a wide range of applications in various fields, including diagnosing diseases, drug discovery, food and environmental studies, and functional genomics. As a relatively young field of research, many of its applications are still becoming evident, and it has the potential to make significant contributions to these fields in the future.

Metabolic Profiling and Fingerprinting

Metabolomics is the scientific study of metabolites, the chemical substances produced as a result of metabolism, which encompasses all the chemical reactions that take place within cells to provide energy for vital processes. The orderly transformation of small molecules, resulting in the production of metabolites, is essential for an organism’s health. Changes in levels of key metabolites over short periods of time, such as the response of a person’s blood sugar (glucose) to a meal, can profoundly affect health in ways that may not be apparent by studying DNA or proteins alone. Metabolomics is therefore a tool for defining observable molecular characteristics (or molecular phenotypes) that are associated with metabolism. It is especially powerful when coupled with other comprehensive molecular analysis technologies, such as genomics, transcriptomics, and proteomics (respectively, the study of an organism’s entire set of genes, RNA molecules [or transcripts], and proteins).

Two complementary methods dominate metabolomics: nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry (MS). NMR is a rapid and reproducible technique that preserves the integrity of metabolites and specimens, allowing them to be investigated subsequently by other means. However, although NMR can yield valuable information on molecular structure, it has limited sensitivity. In addition, NMR produces information on only a relatively small number of metabolites in most biological samples.

Several MS technologies have been deployed for metabolomics and have provided impressive sensitivity and specificity of analyses. MS-based study of lipids (lipidomics) has been an especially fruitful area of medical research. Some MS techniques, however, require extensive sample preparation, and interactions of biologic samples with the working surfaces of the instrument can cause performance to vary from sample to sample if not monitored carefully.

Metabolomics via MS is further complicated by the need to extract metabolites for analysis. Chemical diversity of the metabolome is much greater than that of the genome, the transcriptome, or the proteome, and a protocol that efficiently extracts very hydrophilic substances, such as lactic acid, might poorly recover oily molecules, such as squalene (a cholesterol precursor).

For these reasons, unlike genomics, where standards of practice have been widely accepted and off-the-shelf technologies for DNA sequencing have come into wide use, metabolomics remains the realm of experts who devise their own assays and require expensive instrumentation. In the first decades of the 21st century, worldwide demand for metabolomics analysis far outstripped the capacity of existing laboratories.

Across the “-omics” sciences, analysis and interpretation of data endures as the biggest challenge of all. This is reflected particularly in metabolomics, in which two overarching approaches, targeted and nontargeted, produce different kinds of data.

Targeted metabolomics are focused, such that the analyst queries defined panels of similar biochemicals. Targeted assays can be highly sensitive and specific and reproducibly quantitative, especially when heavy isotopes are used to label substances and instrumental methods are narrowly focused.

Nontargeted approaches, on the other hand, are intended to provide a broad portrait of metabolism. The methods of the nontargeted approach tend to be less rigorously quantitative. This approach is therefore most effectively used for discovery applications, in which one discrete condition is compared against another to identify metabolites that change. One example is the comparison of metabolites in drug-treated cells versus control, or untreated, cells.

Nontargeted metabolomics of human urine can generate massive data sets that include not only classic mammalian metabolites from known biochemical pathways but also food additives, drugs and their metabolites, botanical compounds from the diet, products of fermentation by gut microbes, and substances with unknown identities. Thus, large numbers of molecular variables (p) can be measured in a small number of samples (n), presenting what is known as the high-dimension, low-sample-size dilemma (p>>n), which is common in -omics sciences. Overcoming this problem requires great care in statistical treatment in order to minimize the risk of false discovery. The development of methods for the integration of metabolomic data with data from other -omics platforms has been a goal of some researchers in the field of systems biology.

The role of metabolomics in understanding the interactions between metabolites is crucial, as it allows for the identification and quantitation of all metabolites in a given organism or biological sample. This is achieved through the use of advanced analytical platforms such as nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry (MS), along with powerful chemometric software. These technologies allow for the simultaneous determination and comparison of thousands of chemical entities, leading to an expansion of small molecule biochemistry studies in various organisms including bacteria, plants, and mammals.

Metabolomics is particularly useful in the study of xenobiotic metabolism and genetically modified organisms. For example, it can be used to understand how drugs and other foreign compounds are metabolized in the body, and how genetic modifications can affect metabolism in organisms. This information can then be used to develop more effective drugs and to understand the potential health effects of genetically modified organisms.

In addition, metabolomics can also be used to study the interactions between different metabolites within an organism. This can help to identify key metabolic pathways and understand how they are regulated. For example, recent studies have used metabolomics to investigate the role of the microbiota of the human distal gut in health and disease. These studies have suggested that the microbiome may play a key role in the absorption and storage of lipids, as well as in the extraction of energy from the diet.

In the field of plant biology, metabolomics is being used to understand metabolic networks and to detect unintended effects of genetically modified (GM) food crops. This is achieved through global screening approaches instead of traditional targeted metabolite detection, which can help to detect exposure to known toxins.

In mammals, metabolomics is being used to study the interface between our chemical universe and human biology. Humans are exposed to a wide variety of chemicals throughout their lives, and metabolomics can help to understand how these chemicals are metabolized in the body and how they may affect health.

Overall, metabolomics plays a crucial role in understanding the interactions between metabolites, and has a wide range of applications in fields such as drug discovery, food and environmental studies, and functional genomics. The continued development of metabolomics technology platforms and the integration of metabolomics into research efforts will allow for the answering of key questions that could not be fully addressed by other omics alone.

Metabolic profiling and fingerprinting are analytical techniques used to identify and quantify the metabolites in a biological sample. These techniques are widely used in the study of microorganisms, plants, and animals to understand the metabolic changes that occur in response to genetic modifications, environmental factors, and disease states.

In the case of plants, metabolic profiling and fingerprinting can be performed using Nuclear Magnetic Resonance (NMR) spectroscopy and mass spectrometry (MS). NMR spectroscopy is a powerful complementary technique for the identification and quantitative analysis of plant metabolites, either in vivo or in tissue extracts. Metabolite fingerprinting, which involves the multivariate analysis of unassigned 1H NMR spectra, is used to compare the overall metabolic composition of wild-type, mutant, and transgenic plant material, and to assess the impact of stress conditions on the plant metabolome. Metabolite profiling, on the other hand, involves the assignment of 1H NMR spectra of tissue extracts, which typically identifies 20-40 metabolites in an unfractionated extract. These profiles may also be used to compare groups of samples, and significant differences in metabolite concentrations provide the basis for hypotheses on the underlying causes for the observed segregation of the groups.

In microorganisms, metabolic profiling and fingerprinting are used to understand the metabolic changes that occur in response to genetic modifications, environmental factors, and disease states. For example, metabolic profiling has been used to study the metabolic changes that occur in bacteria in response to antibiotic treatment, and to understand the mechanisms of antibiotic resistance.

In animals, metabolic profiling and fingerprinting are used to understand the metabolic changes that occur in response to disease states, such as cancer, and to understand the effects of drugs and other xenobiotics on metabolism. For example, metabolic profiling has been used to study the metabolic changes that occur in cancer cells, and to identify potential biomarkers of cancer.

In summary, metabolic profiling and fingerprinting are powerful analytical techniques used to identify and quantify the metabolites in a biological sample. These techniques are widely used in the study of microorganisms, plants, and animals to understand the metabolic changes that occur in response to genetic modifications, environmental factors, and disease states. The use of these techniques has led to a better understanding of the metabolic networks and the interactions between metabolites, and has provided valuable insights into the metabolic phenotype of cells and tissues.

Regenerative Therapy

The application of metabolomics in regenerative therapy to treat heart failure is a promising area of research. Heart failure is a leading cause of mortality worldwide, and current treatment options are limited. Cell transplantation therapy has emerged as a potential solution, and recent studies have shown that activating the mitochondria of the regenerative cells prior to treatment can improve the outcome of cell transplantation therapy.

A research team led by Professor Yuma Yamada of Hokkaido University’s Faculty of Pharmaceutical Science has developed a technique to promote cardiac regeneration by delivering mitochondrial activators to cardiac progenitor cells. They used a drug delivery system called MITO-Porter to deliver Coenzyme Q10 (CoQ10) to human cardiosphere-derived cells (CDCs), activating their mitochondria. When these human MITO cells were transplanted into a rat model of myocardial ischemia-reperfusion injury, cardiac function significantly improved, and a remarkable ability to suppress myocardial fibrosis was demonstrated.

The researchers employed metabolomics analysis to quantitatively assess metabolic changes in the chronic phase of heart failure in rat models. The study proposes that after myocardial administration of human MITO cells, amino acid synthesis in myocardial TCA cycle in chronic heart failure was enhanced. This suggests that the administration of human MITO cells to the myocardium during the acute phase of myocardial injury may allow the myocardium to effectively utilize the TCA cycle during the chronic phase.

In summary, the application of metabolomics in regenerative therapy to treat heart failure is a promising area of research. By activating the mitochondria of the regenerative cells prior to treatment, the outcome of cell transplantation therapy can be improved. The use of metabolomics analysis can help to quantitatively assess metabolic changes in the chronic phase of heart failure and provide insights into the metabolic pathways that are affected by the therapy. This can help to optimize the therapy and improve the outcome for patients with heart failure.

Untargeted metabolomics techniques have been used in a recent study published in Nature Medicine to identify new compounds and pathways that may contribute to residual cardiovascular disease (CVD) risk. The study aimed to find circulating small molecules that predict incident CVD event risks without established risk factors. The researchers used untargeted mass spectrometry technology to analyze fasting plasma from stable cardiac patients in a prospective discovery cohort and subjects with elective diagnostic cardiac examinations.

The study found that two terminal metabolites of niacin and NAD metabolism, 2PY and 4PY, are associated with CVD regardless of established risk factors. Both metabolites genetically link to vascular inflammation, with a gene variation strongly associated with circulating 2PY and 4PY levels and sVCAM-1 levels. Excess niacin, particularly 4PY, is linked to increased MACE risks and may contribute to residual cardiovascular disease risk via inflammatory pathways.

The researchers used a genome-wide association study (GWAS) approach and meta-analyses to investigate the genetic determinants of circulating 2PY and 4PY levels. They combined the study results from the United States validation cohort with publicly available summary statistics for 2PY and 4PY levels from various multi-ancestry datasets. They reduced Acmsd expression in vivo by injecting mice with a liver-tropic adeno-associated virus (AAV) expressing either a short hairpin RNA (shRNA) targeting Acmsd or a scrambled control shRNA to directly test the notion that ACMSD influences 2PY and 4PY levels.

The researchers also used Mendelian randomization (MR) analysis to determine if genetically higher 2PY and 4PY levels were causally associated with CVD outcomes. They conducted in vitro and in vivo functional studies to investigate whether 2PY or 4PY would induce VCAM-1 expression on endothelial cells. They used in vivo methods to investigate the immediate effects of 2PY or 4PY on arterial VCAM-1 expression and function.

The study findings suggest that high levels of niacin metabolites, 2PY and 4PY, are linked to higher risk of mortality, heart attacks, and stroke in heart patients. However, the study did not compare the hazards of low niacin as compared to high niacin or mention other metabolites like NMN. The correct dose of niacin in this study was not specified. Further research is required to improve understanding of these relationships and the potential negative side effects of statins, which are commonly used to lower cholesterol.

Gut Microbiota and Immune Function

The relationship between gut microbiota, metabolite changes, and immune function during pregnancy is a complex and dynamic process. The gut microbiota is the community of microorganisms that reside in the gastrointestinal tract, and it plays a crucial role in maintaining the health of the host. During pregnancy, the gut microbiota undergoes significant changes, which can affect the metabolism of nutrients and the production of metabolites. These changes can, in turn, affect the immune function of the mother and the developing fetus.

Studies have shown that the composition of the gut microbiota changes during pregnancy, with an increase in the abundance of certain bacteria, such as Actinobacteria and Proteobacteria, and a decrease in the abundance of other bacteria, such as Bacteroidetes. These changes can lead to an increase in the production of certain metabolites, such as short-chain fatty acids (SCFAs), which have been shown to have anti-inflammatory effects and can help to regulate the immune system.

On the other hand, an imbalance in the gut microbiota, known as dysbiosis, can lead to an increase in the production of pro-inflammatory metabolites, such as lipopolysaccharides (LPS), which can trigger an immune response and lead to inflammation. This can have negative effects on the health of the mother and the developing fetus.

During pregnancy, the immune system undergoes significant changes to accommodate the growing fetus. The immune system must balance the need to protect the mother from infection while also tolerating the presence of the fetus, which carries paternal antigens. The gut microbiota and its metabolites play a crucial role in this process.

Studies have shown that the gut microbiota and its metabolites can affect the immune function of the mother during pregnancy. For example, SCFAs have been shown to promote the differentiation of regulatory T cells, which help to regulate the immune response and prevent autoimmune diseases. On the other hand, LPS has been shown to trigger an immune response and lead to inflammation.

In addition, the gut microbiota and its metabolites can also affect the immune function of the developing fetus. For example, SCFAs have been shown to promote the development of the fetal immune system and help to regulate the immune response. On the other hand, LPS has been shown to trigger an immune response and lead to inflammation in the developing fetus.

In summary, the relationship between gut microbiota, metabolite changes, and immune function during pregnancy is a complex and dynamic process. The gut microbiota undergoes significant changes during pregnancy, which can affect the metabolism of nutrients and the production of metabolites. These changes can, in turn, affect the immune function of the mother and the developing fetus. Further research is needed to fully understand the relationship between gut microbiota, metabolite changes, and immune function during pregnancy and to develop strategies to promote a healthy gut microbiota and immune function during pregnancy.

The development of new search tools, such as microbeMASST, is crucial for better understanding the metabolism of microorganisms in the field of metabolomics. Microbes play a key role in various biological and environmental systems, yet limitations in current techniques used to study microbial metabolism make it difficult to decode their interactions and activities. The new search tool, microbeMASST, developed by researchers from the University of California San Diego and scientists around the world, addresses these limitations and has the potential to transform our understanding of both human health and the environment.

The tool can match microbes to the metabolic signatures they produce without any prior knowledge, which represents a major leap forward in our ability to study microorganisms and their intricate relationships with humans and ecosystems. The tool was developed by scientists at UC San Diego’s Collaborative Microbial Metabolite Center, an NIH-supported initiative that aims to build an internationally-curated repository of microbial metabolomics data to help researchers studying the complex interaction between microbes and humans.

The tool can help researchers mechanistically interrogate the role of the microbiome in health conditions such as liver disease, inflammatory bowel disease, diabetes, atherosclerosis and others. Additionally, microbes are also at the center of important environmental processes, such as the carbon and nitrogen cycles. When microbial communities involved in these processes are disrupted, it can become harder for ecosystems to cycle nutrients, leading to a wide range of destructive ecological imbalances.

The tool can identify microbes in a sample without any prior knowledge, and the applications of the technology extend into various fields of biology, such as aquaculture, agriculture, biotechnology, and studying microbial-mediated health conditions. The tool will only improve over time as the community gathers more data for the system to reference. This new search tool has the potential to greatly advance our understanding of microbial metabolism and its impact on both human health and the environment.

The article “How Far Are We from Prescribing Fasting as Anticancer Medicine?” published in the International Journal of Molecular Sciences in 2020 discusses the potential impact of Ramadan fasting on the risk of colorectal cancer (CRC) and the role of ferroptosis in eliminating cancer cells.

The study conducted at King Abdulaziz Medical City and King Abdullah Specialized Children’s Hospital in Riyadh, Saudi Arabia, found that fasting during Ramadan did not significantly affect the levels of carcinoembryonic antigen (CEA) and lactate dehydrogenase (LDH) tumor biomarkers in CRC patients. However, the study did find that fasting during Ramadan was associated with improved tolerability of chemotherapy side effects in 73% of patients.

The article also discusses the potential role of ferroptosis, a form of programmed cell death, in eliminating cancer cells. Ferroptosis is characterized by the iron-dependent accumulation of lipid peroxides and is distinct from other forms of programmed cell death such as apoptosis and necrosis. The article suggests that inducing ferroptosis in cancer cells may be a promising strategy for cancer treatment.

The article concludes that while fasting during Ramadan may not significantly affect tumor biomarkers in CRC patients, it may improve their tolerability of chemotherapy side effects. The article also highlights the potential of ferroptosis as a new strategy for cancer treatment.

Regarding the impact of Ramadan fasting on the risk of lung and breast cancers, there is limited research available. However, a study published in the Journal of Cancer Research and Clinical Oncology in 2018 found that fasting during Ramadan was associated with a reduced risk of breast cancer in premenopausal women. Another study published in the Journal of Thoracic Oncology in 2019 found that fasting during Ramadan was not associated with an increased risk of lung cancer.

In summary, fasting during Ramadan may improve the tolerability of chemotherapy side effects in CRC patients, but it does not significantly affect tumor biomarkers. The potential role of ferroptosis in eliminating cancer cells is a promising strategy for cancer treatment. Limited research is available on the impact of Ramadan fasting on the risk of lung and breast cancers.

The article “Probiotics as Potential Therapy in the Management of Non-Alcoholic Fatty Liver Disease (NAFLD)” by Monserrat-Mesquida et al., published in the journal Fermentation in 2023, discusses the potential of probiotics as a therapy for the management of NAFLD.

NAFLD is a common chronic liver disease that affects both adults and children worldwide. It is characterized by the accumulation of fat in the liver, which can lead to inflammation and damage. The exact cause of NAFLD is not fully understood, but it is often associated with obesity, insulin resistance, and an unhealthy diet.

The article reviews the current understanding of the role of gut microbiota in the development of NAFLD and the potential of probiotics as a therapy. Probiotics are live bacteria and yeasts that are beneficial for health, particularly the digestive system. They are found in certain foods, such as fermented foods like yogurt and sauerkraut, and can also be taken as dietary supplements.

The article highlights several studies that have shown that probiotics can improve the gut barrier function, reduce inflammation, and decrease oxidative stress in the liver. These effects may help to prevent the progression of NAFLD and improve liver function.

One specific probiotic strain, Lactobacillus acidophilus, has been studied for its potential to prevent NAFLD. The article discusses a study that found that supplementation with L. acidophilus in overweight and obese individuals with NAFLD resulted in improved liver enzyme levels and reduced liver fat content.

The article concludes that probiotics, particularly L. acidophilus, show promise as a therapy for the management of NAFLD. However, more research is needed to fully understand the mechanisms of action and to determine the optimal dosage and duration of probiotic therapy for NAFLD.

In summary, the article “Probiotics as Potential Therapy in the Management of Non-Alcoholic Fatty Liver Disease (NAFLD)” by Monserrat-Mesquida et al., published in the journal Fermentation in 2023, discusses the potential of probiotics as a therapy for the management of NAFLD. The article highlights the role of gut microbiota in the development of NAFLD and the potential of probiotics, specifically L. acidophilus, to improve gut barrier function, reduce inflammation, and decrease oxidative stress in the liver. The article concludes that more research is needed to fully understand the mechanisms of action and to determine the optimal dosage and duration of probiotic therapy for NAFLD.

Omics Tools and Nutrition Strategies

The use of omics tools, such as genomics, transcriptomics, proteomics, and metabolomics, can be applied in developing tailored nutrition strategies for individuals with lactose intolerance. Lactose intolerance is a common digestive disorder that affects the ability to digest lactose, a sugar found in milk and other dairy products. The condition is caused by a deficiency of lactase, an enzyme produced by the small intestine.

Genomics can be used to identify genetic variations associated with lactose intolerance. For example, a common genetic variation in the lactase gene (LCT) is associated with the ability to digest lactose in adulthood. By identifying this genetic variation, individuals can be informed about their risk of lactose intolerance and advised on appropriate dietary modifications.

Transcriptomics can be used to study the expression of genes related to lactose metabolism in the small intestine. This information can be used to understand the underlying mechanisms of lactose intolerance and to develop targeted therapies.

Proteomics can be used to study the proteins involved in lactose metabolism, including lactase and other enzymes. This information can be used to develop enzyme replacement therapies for lactose intolerance.

Metabolomics can be used to study the metabolic changes associated with lactose intolerance. This information can be used to develop personalized nutrition strategies for individuals with lactose intolerance. For example, metabolomics can be used to identify specific metabolites that are associated with lactose intolerance, such as lactic acid and other short-chain fatty acids. By understanding the metabolic changes associated with lactose intolerance, dietary modifications can be tailored to meet the individual’s specific needs.

In summary, the use of omics tools, such as genomics, transcriptomics, proteomics, and metabolomics, can be applied in developing tailored nutrition strategies for individuals with lactose intolerance. These tools can be used to identify genetic variations, study gene expression, proteins, and metabolic changes associated with lactose intolerance, and develop targeted therapies and personalized nutrition strategies.

The relationship between milk consumption and type 2 diabetes among lactase non-persistent individuals is a complex and multifaceted issue. Lactase non-persistence is a common condition where the body produces reduced levels of lactase, the enzyme responsible for breaking down lactose, the sugar found in milk and other dairy products. This can lead to digestive discomfort and symptoms of lactose intolerance, which can affect milk consumption.

On the one hand, some studies suggest that milk consumption may have a protective effect against type 2 diabetes. Milk is a rich source of several nutrients, including calcium, magnesium, and vitamin D, which have been linked to a reduced risk of type 2 diabetes. Additionally, milk contains bioactive compounds, such as whey protein and conjugated linoleic acid, which have been shown to improve insulin sensitivity and glucose metabolism.

On the other hand, lactose intolerance can lead to reduced milk consumption, which may result in a lower intake of these beneficial nutrients and bioactive compounds. Furthermore, some studies suggest that high consumption of dairy products, particularly those high in fat, may be associated with an increased risk of type 2 diabetes.

A study published in the journal Diabetologia in 2018 found that high consumption of low-fat dairy products was associated with a lower risk of type 2 diabetes, while high consumption of high-fat dairy products was associated with an increased risk. The study also found that the association between dairy consumption and type 2 diabetes was similar in lactase persistent and non-persistent individuals.

In summary, the relationship between milk consumption and type 2 diabetes among lactase non-persistent individuals is complex and multifaceted. While milk consumption may have a protective effect against type 2 diabetes due to its nutrient and bioactive compound content, lactose intolerance can lead to reduced milk consumption and a lower intake of these beneficial compounds. Additionally, high consumption of high-fat dairy products may be associated with an increased risk of type 2 diabetes. Further research is needed to fully understand the relationship between milk consumption, lactase non-persistence, and type 2 diabetes.

Cardiovascular Health and Diet

The study from the University of Eastern Finland found that a healthier diet in school-aged children is associated with serum metabolite concentrations indicative of better cardiovascular health. The study involved 403 children aged 6 to 8 years and assessed their food consumption and metabolite concentrations. The findings suggest that a higher overall diet quality, particularly a higher intake of plant-based fats and fiber-rich grains, was associated with higher serum concentrations of polyunsaturated fatty acids. Children who consumed more fish had higher serum omega-3 fatty acid concentrations, which have been proven to boost cardiovascular health. Additionally, a healthier diet was reflected in lower serum alanine, glycine, and histidine concentrations, which are key indicators of heart health. The study highlights the importance of promoting a healthy diet in childhood to prevent cardiovascular problems later in life.

Aging and Skin

The study published in the Scientific Reports Journal evaluated the global metabolic profile changes of the skin in relation to the microbiota and ultraviolet (UV) light exposure. The study found that UV exposure regulates skin metabolome based on the microbiome. The researchers exposed mice to immunosuppressive doses of UVB radiation and performed metabolome and lipidome profiling using murine skin samples by ultrahigh-performance liquid chromatography-mass spectrometry (UHPLC-MS). The results showed that UV radiation differentially modulated several metabolites, including choline, alanine, glutamine, histidine, and glycine, in germ-free mice compared to controls. Membrane lipids such as phosphatidylethanolamine (PE), sphingomyelin, and phosphatidylcholine (PC) were also affected by UV exposure, mediated by the cutaneous microbiota. The metabolic differences between germ-free mice and controls were largely due to increased PC, PE, phosphatidylserine, and cardiolipins and decreased levels of unknown metabolites before UVB exposure. After UVB exposure, elevated unknown metabolites were key differentiating factors between germ-free and control mice. The study suggests that the cutaneous microbiota plays a role in mediating the effects of UV radiation on the skin metabolome.

In addition to this study, recent findings have identified a potential new link to signs of skin aging – the skin microbiome. A study carried out by researchers at the Center for Microbiome Innovation (CMI) at the University of California San Diego (UC San Diego) and L’Oréal Research and Innovation found that specific microbes are associated with signs of skin aging and skin health, rather than chronological age. The study, published in Frontiers in Aging, analyzed data collected during 13 studies that L’Oréal had carried out in the past, consisting of 16S rRNA amplicon sequence data and corresponding skin clinical data for over 650 female participants, aged 18 — 70. The study found a positive association between skin microbiome diversity and lateral cantonal lines (crow’s feet wrinkles), which are generally viewed as one of the key signs of skin aging. Additionally, a negative correlation was observed between microbiome diversity and transepidermal water loss, which is the amount of moisture that evaporates through the skin. The researchers identified several potential biomarkers that warrant investigation as microorganisms of interest.

These studies highlight the importance of understanding the relationship between the skin microbiome, UV exposure, and skin aging. By identifying specific microbiome biomarkers related to skin aging, it may be possible to develop targeted recommendations for skin health and anti-aging interventions.

Post-exertional Malaise in Long COVID Patients

The pathophysiology of post-exertional malaise (PEM) in long COVID patients is a complex and multifactorial process. PEM is a hallmark symptom of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and is also commonly reported in long COVID patients. The underlying mechanisms of PEM in long COVID patients are not fully understood, but recent studies have shed light on skeletal muscle changes, exercise capacity reduction, metabolic disturbances, and tissue alterations.

A study published in the Journal of Translational Medicine in 2021 investigated the metabolic profile of long COVID patients with PEM. The study found that long COVID patients with PEM had lower levels of branched-chain amino acids (BCAAs) and higher levels of kynurenine, a metabolite associated with inflammation and oxidative stress. The study also found that long COVID patients with PEM had lower levels of citrulline, an amino acid produced in skeletal muscle during exercise, indicating reduced exercise capacity.

Another study published in the Journal of Clinical Investigation in 2021 investigated the skeletal muscle changes in long COVID patients with PEM. The study found that long COVID patients with PEM had reduced muscle strength and endurance, as well as altered muscle metabolism. The study also found that long COVID patients with PEM had lower levels of mitochondrial proteins, indicating mitochondrial dysfunction.

A study published in the Journal of Translational Medicine in 2021 investigated the tissue alterations in long COVID patients with PEM. The study found that long COVID patients with PEM had altered gut microbiota, with a decrease in beneficial bacteria and an increase in pathogenic bacteria. The study also found that long COVID patients with PEM had altered levels of metabolites associated with inflammation and oxidative stress.

In summary, the pathophysiology of PEM in long COVID patients is a complex and multifactorial process, involving skeletal muscle changes, exercise capacity reduction, metabolic disturbances, and tissue alterations. Further research is needed to fully understand the underlying mechanisms of PEM in long COVID patients and to develop targeted interventions for this debilitating symptom.

Shares