Celiac disease is an immune-mediated disorder with significant metabolic implications. Several factors have been proposed to explain the association between celiac disease in patients following a gluten-free diet and metabolic disorders, including metabolic syndrome. Growing evidence suggests a pivotal role of gut microbiome dysbiosis in the onset of celiac disease and its associated metabolic disturbances. The present narrative review examines (i) the connections between celiac disease and metabolism-related comorbidities, including metabolic syndrome and metabolic dysfunction-associated steatotic liver disease; (ii) the role of the gut microbiome in celiac disease, including the outcomes of gut microbiome dysbiosis in celiac children and adults; and (iii) the potential of microbial therapeutic strategies within the context of personalized medicine for patients with celiac disease and comorbid metabolic conditions. A synthesis of existing studies highlights several protective factors and interventions for future celiac disease prevention research. Adopting plant-based, health-promoting dietary patterns such as the Mediterranean or vegetarian diet within the first two years of life reduces celiac disease risk. These fiber- and phytochemical-rich diets support beneficial gut microbiota growth and short-chain fatty acid production, which maintain intestinal barrier integrity by enhancing mucus and tight junction proteins. Short-chain fatty acids also modulate immunity by inducing Tregs that secrete IL-10, suppressing pro-inflammatory Th1 responses and autoantibody production. Precision probiotics offer diverse therapeutic benefits in celiac disease by reducing inflammation, restoring beneficial microbes, and degrading immunogenic gliadin peptides. Postbiotics complement these effects by reinforcing barrier integrity and counteracting gliadin-induced inflammation. Thus, integrating clinical models with microbial biomarkers promises to improve celiac disease diagnosis and monitoring, enabling better risk stratification, earlier detection, and personalized management of this heterogeneous disease.
Celiac disease is an immune-mediated disorder with significant metabolic implications. Several factors have been proposed to explain the association between celiac disease in patients following a gluten-free diet and metabolic disorders, including metabolic syndrome. Growing evidence suggests a pivotal role of gut microbiome dysbiosis in the onset of celiac disease and its associated metabolic disturbances. The present narrative review examines (i) the connections between celiac disease and metabolism-related comorbidities, including metabolic syndrome and metabolic dysfunction-associated steatotic liver disease; (ii) the role of the gut microbiome in celiac disease, including the outcomes of gut microbiome dysbiosis in celiac children and adults; and (iii) the potential of microbial therapeutic strategies within the context of personalized medicine for patients with celiac disease and comorbid metabolic conditions. A synthesis of existing studies highlights several protective factors and interventions for future celiac disease prevention research. Adopting plant-based, health-promoting dietary patterns such as the Mediterranean or vegetarian diet within the first two years of life reduces celiac disease risk. These fiber- and phytochemical-rich diets support beneficial gut microbiota growth and short-chain fatty acid production, which maintain intestinal barrier integrity by enhancing mucus and tight junction proteins. Short-chain fatty acids also modulate immunity by inducing Tregs that secrete IL-10, suppressing pro-inflammatory Th1 responses and autoantibody production. Precision probiotics offer diverse therapeutic benefits in celiac disease by reducing inflammation, restoring beneficial microbes, and degrading immunogenic gliadin peptides. Postbiotics complement these effects by reinforcing barrier integrity and counteracting gliadin-induced inflammation. Thus, integrating clinical models with microbial biomarkers promises to improve celiac disease diagnosis and monitoring, enabling better risk stratification, earlier detection, and personalized management of this heterogeneous disease.
Fibromyalgia syndrome (FMS) is a chronic condition characterized by widespread musculoskeletal pain, fatigue, cognitive impairments, and sleep disturbances. Although traditionally considered psychogenic, recent research supports a multifactorial etiology involving central nervous system (CNS) dysregulation and significant immune involvement. This narrative review synthesizes current evidence regarding the role of immune mechanisms in FMS, with comparative insights into chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) and irritable bowel syndrome (IBS)—previously grouped under functional somatic syndromes (FSS). In FMS, immune dysregulation is evidenced by elevated levels of pro-inflammatory cytokines (e.g., IL-6, IL-8, TNF-α) and decreased anti-inflammatory mediators such as IL-10, contributing to symptomatology including pain amplification and fatigue. Neuroinflammation, as indicated by microglial activation in pain-processing CNS regions, further supports the role of immune signaling in central sensitization. Other contributing factors include oxidative stress, mitochondrial dysfunction, and immune cell alterations, particularly involving regulatory T cells and natural killer (NK) cells. Compared to FMS, CFS/ME exhibits greater systemic immune activation and more severe mitochondrial impairment, correlating with profound fatigue and cognitive decline. IBS, on the other hand, shows immune activation localized to the gastrointestinal tract, emphasizing the gut-brain axis. These findings highlight both shared and syndrome-specific immune features. To better reflect their systemic and immunological complexity, this review refers to these conditions collectively as chronic multisystem immune-related disorders (CMIRDs). The evidence supports the development of biomarker-based diagnostics and personalized immunomodulatory therapies. A multidisciplinary approach that integrates immunology and neurology is essential to improve outcomes for patients with FMS and related disorders.
Fibromyalgia syndrome (FMS) is a chronic condition characterized by widespread musculoskeletal pain, fatigue, cognitive impairments, and sleep disturbances. Although traditionally considered psychogenic, recent research supports a multifactorial etiology involving central nervous system (CNS) dysregulation and significant immune involvement. This narrative review synthesizes current evidence regarding the role of immune mechanisms in FMS, with comparative insights into chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) and irritable bowel syndrome (IBS)—previously grouped under functional somatic syndromes (FSS). In FMS, immune dysregulation is evidenced by elevated levels of pro-inflammatory cytokines (e.g., IL-6, IL-8, TNF-α) and decreased anti-inflammatory mediators such as IL-10, contributing to symptomatology including pain amplification and fatigue. Neuroinflammation, as indicated by microglial activation in pain-processing CNS regions, further supports the role of immune signaling in central sensitization. Other contributing factors include oxidative stress, mitochondrial dysfunction, and immune cell alterations, particularly involving regulatory T cells and natural killer (NK) cells. Compared to FMS, CFS/ME exhibits greater systemic immune activation and more severe mitochondrial impairment, correlating with profound fatigue and cognitive decline. IBS, on the other hand, shows immune activation localized to the gastrointestinal tract, emphasizing the gut-brain axis. These findings highlight both shared and syndrome-specific immune features. To better reflect their systemic and immunological complexity, this review refers to these conditions collectively as chronic multisystem immune-related disorders (CMIRDs). The evidence supports the development of biomarker-based diagnostics and personalized immunomodulatory therapies. A multidisciplinary approach that integrates immunology and neurology is essential to improve outcomes for patients with FMS and related disorders.
Soya (Glycine max L.) is a legume rich in nutrients (proteins, lipids, carbohydrates, and minerals) but also in anti-nutrients such as oxalate, which hampers the bioavailability of nutrients. The various processes used to treat it reduce the anti-nutrient content while affecting the protein content to a greater or lesser extent. This study aimed to evaluate the impact of a soaking period followed by cooking on the reduction of oxalates and enhancement of protein availability in soya flour.
To achieve this, the response surface methodology with the centred composite design was used to reduce the oxalate content and increase the protein content of cooked soya flour. The factors chosen were soaking time (5–24 h), cooking time (15–50 min), and cooking temperature (70–100°C). The soya flours obtained were characterised for protein and oxalate content using standard methods. The optimal samples were also characterised.
The results obtained showed that cooking and soaking times, followed by the quadratic effect of soaking time, significantly (p < 0.05) increase the protein content and decrease the oxalate content. In terms of optimal conditions, a soaking time of 25.44 h, a cooking temperature of 101.05°C, and a cooking time of 61.93 min reduced the oxalate content by 87.43% and also increased the protein content from 35.98 g/100 g DM to 49.16 g/100 g DM. Optimal conditions of the different treatments also increase lipids, reducing sugar, and the main minerals like Ca, P, Mg, and Fe.
The application of such conditions would help to combat protein deficiencies.
Soya (Glycine max L.) is a legume rich in nutrients (proteins, lipids, carbohydrates, and minerals) but also in anti-nutrients such as oxalate, which hampers the bioavailability of nutrients. The various processes used to treat it reduce the anti-nutrient content while affecting the protein content to a greater or lesser extent. This study aimed to evaluate the impact of a soaking period followed by cooking on the reduction of oxalates and enhancement of protein availability in soya flour.
To achieve this, the response surface methodology with the centred composite design was used to reduce the oxalate content and increase the protein content of cooked soya flour. The factors chosen were soaking time (5–24 h), cooking time (15–50 min), and cooking temperature (70–100°C). The soya flours obtained were characterised for protein and oxalate content using standard methods. The optimal samples were also characterised.
The results obtained showed that cooking and soaking times, followed by the quadratic effect of soaking time, significantly (p < 0.05) increase the protein content and decrease the oxalate content. In terms of optimal conditions, a soaking time of 25.44 h, a cooking temperature of 101.05°C, and a cooking time of 61.93 min reduced the oxalate content by 87.43% and also increased the protein content from 35.98 g/100 g DM to 49.16 g/100 g DM. Optimal conditions of the different treatments also increase lipids, reducing sugar, and the main minerals like Ca, P, Mg, and Fe.
The application of such conditions would help to combat protein deficiencies.
Despite being a fruit rich in resistant starch, acorns remain undervalued. Resistant starch is known to improve food acceptability when compared to traditional insoluble fibers, and recent research suggests the usage of acorn starch as an additive in fermented yogurt and milk products. Furthermore, non-thermal technologies such as high hydrostatic pressure and pulsed electric field can produce clean-labelled starches. Milk puddings are widely consumed all over the world and are usually produced using representative amounts of starch, making them an enticing food matrix for incorporating acorn starches. Hence, the effects of replacing commercial corn starch with acorn starch extracted by high hydrostatic pressure and pulsed electric field on the nutritional composition, functional and sensorial properties, and shelf-life of puddings were studied.
Extraction of starch from Quercus robur acorns was performed using high hydrostatic pressure or pulsed electric field. Extracted starch was used in chocolate puddings, replacing commercial corn starch. Shelf-life storage and microbiological analysis were conducted over 28 days, along with texture, color, pH, and nutritional composition assessments. Rheological properties, scanning electron microscopy, in vitro digestion, and soluble sugar, fatty acid, and salt content analyses were performed. Sensorial analysis was conducted with 71 volunteer panelists to evaluate the acceptability, preference, and similarity of puddings.
Replacing the commercial corn starch with acorn starch improves the rheological properties of puddings and has no negative impact on the nutritional composition, internal structure, or in vitro digestibility. Sensory analysis revealed that panelists preferred the acorn starch puddings over the control. After 28 days of storage at 4°C, there was a greater stabilization of the color parameters and an improvement in textural parameters of puddings without compromising microbial safety.
This study demonstrates the potential usage of starch from acorns in food applications, a fruit that is so undervalued.
Despite being a fruit rich in resistant starch, acorns remain undervalued. Resistant starch is known to improve food acceptability when compared to traditional insoluble fibers, and recent research suggests the usage of acorn starch as an additive in fermented yogurt and milk products. Furthermore, non-thermal technologies such as high hydrostatic pressure and pulsed electric field can produce clean-labelled starches. Milk puddings are widely consumed all over the world and are usually produced using representative amounts of starch, making them an enticing food matrix for incorporating acorn starches. Hence, the effects of replacing commercial corn starch with acorn starch extracted by high hydrostatic pressure and pulsed electric field on the nutritional composition, functional and sensorial properties, and shelf-life of puddings were studied.
Extraction of starch from Quercus robur acorns was performed using high hydrostatic pressure or pulsed electric field. Extracted starch was used in chocolate puddings, replacing commercial corn starch. Shelf-life storage and microbiological analysis were conducted over 28 days, along with texture, color, pH, and nutritional composition assessments. Rheological properties, scanning electron microscopy, in vitro digestion, and soluble sugar, fatty acid, and salt content analyses were performed. Sensorial analysis was conducted with 71 volunteer panelists to evaluate the acceptability, preference, and similarity of puddings.
Replacing the commercial corn starch with acorn starch improves the rheological properties of puddings and has no negative impact on the nutritional composition, internal structure, or in vitro digestibility. Sensory analysis revealed that panelists preferred the acorn starch puddings over the control. After 28 days of storage at 4°C, there was a greater stabilization of the color parameters and an improvement in textural parameters of puddings without compromising microbial safety.
This study demonstrates the potential usage of starch from acorns in food applications, a fruit that is so undervalued.
Plants possess tremendous medicinal properties which have been supposed to be promising candidates for biomedical applications, especially in the field of nanobiotechnology. To analyze one such view, the current study was adopted to synthesize gold nanoparticles (Au*nps) by employing the extract of Murraya koenigii (EMk) for the evaluation of phenolics, antioxidant, antimicrobial, hemolytic, and biocompatible activities.
The synthesis process was carried out in a single step by mixing EMk and gold salt (Au salt) solution and monitored using UV/Visible spectroscopy. The process was optimized via variation in environmental variables. Characterization techniques such as Fourier transform infrared (FTIR) spectroscopy, transmission electron microscopy (TEM), scanning electron microscopy (SEM), X-ray diffractometer (XRD), and energy dispersive X-rays (EDX) were employed. In vitro biological activities (total phenolic, antioxidant, antimicrobial, and hemolytic) using different concentrations of Au*nps along with EMk were assessed. An in vivo histopathology study on Wistar rats was analyzed.
The band of Au*nps was observed at 540 nm, which showed successful synthesis. The FTIR spectra of Au*nps indicated the role of different functional groups (alkane, aromatic ester, thiol, nitro, and aldehyde) of EMk during synthesis. The TEM analysis illustrated a 50 nm size of Au*nps; SEM showed the presence of some aggregates; EDX represented elemental nature, and XRD proved the crystalline nature of these Au*nps. The Au*nps possessed significant phenolic content and displayed prominent antioxidant activities by quenching free radicals. Similarly, momentous inhibitory action was observed against microbial strains of Escherichia coli and Bacillus subtilis. The hemolytic study showed the least to non-toxic effect of these nanoparticles on red blood cells (RBCs) even at enhanced concentration. Histopathology study showed fair compatibility without inducing any apparent pathological lesions on the liver tissues of Wistar rats.
Plausibly, all the above investigations strongly emphasized the use of medicinal plant-based Au*nps for biological applications.
Plants possess tremendous medicinal properties which have been supposed to be promising candidates for biomedical applications, especially in the field of nanobiotechnology. To analyze one such view, the current study was adopted to synthesize gold nanoparticles (Au*nps) by employing the extract of Murraya koenigii (EMk) for the evaluation of phenolics, antioxidant, antimicrobial, hemolytic, and biocompatible activities.
The synthesis process was carried out in a single step by mixing EMk and gold salt (Au salt) solution and monitored using UV/Visible spectroscopy. The process was optimized via variation in environmental variables. Characterization techniques such as Fourier transform infrared (FTIR) spectroscopy, transmission electron microscopy (TEM), scanning electron microscopy (SEM), X-ray diffractometer (XRD), and energy dispersive X-rays (EDX) were employed. In vitro biological activities (total phenolic, antioxidant, antimicrobial, and hemolytic) using different concentrations of Au*nps along with EMk were assessed. An in vivo histopathology study on Wistar rats was analyzed.
The band of Au*nps was observed at 540 nm, which showed successful synthesis. The FTIR spectra of Au*nps indicated the role of different functional groups (alkane, aromatic ester, thiol, nitro, and aldehyde) of EMk during synthesis. The TEM analysis illustrated a 50 nm size of Au*nps; SEM showed the presence of some aggregates; EDX represented elemental nature, and XRD proved the crystalline nature of these Au*nps. The Au*nps possessed significant phenolic content and displayed prominent antioxidant activities by quenching free radicals. Similarly, momentous inhibitory action was observed against microbial strains of Escherichia coli and Bacillus subtilis. The hemolytic study showed the least to non-toxic effect of these nanoparticles on red blood cells (RBCs) even at enhanced concentration. Histopathology study showed fair compatibility without inducing any apparent pathological lesions on the liver tissues of Wistar rats.
Plausibly, all the above investigations strongly emphasized the use of medicinal plant-based Au*nps for biological applications.
Down syndrome (DS), caused by trisomy 21, is strongly associated with an increased risk of early-onset Alzheimer’s disease (AD). This work explores the cellular, genetic, epigenetic, and neuropsychological mechanisms that underlie the accelerated development of AD in individuals with DS. We review key contributors such as amyloid-β accumulation, mitochondrial dysfunction, oxidative stress, tau pathology, neuroinflammation, and chromosomal and epigenetic instability in the neuropathology of AD in DS. Particular attention is given to genes, microRNAs, and chromatin remodeling factors encoded by human chromosome 21 (Hsa21) that regulate these pathological processes. We also highlight the roles of non-coding RNAs and altered DNA methylation patterns in modulating gene expression and neuronal vulnerability. Additionally, the writing evaluates current pharmacological and non-pharmacological interventions and addresses the critical need for inclusive, person-centered health services. Integrating molecular biology with clinical perspectives, the review emphasizes the importance of early diagnosis and coordinated care strategies for individuals with DS at risk for AD.
Down syndrome (DS), caused by trisomy 21, is strongly associated with an increased risk of early-onset Alzheimer’s disease (AD). This work explores the cellular, genetic, epigenetic, and neuropsychological mechanisms that underlie the accelerated development of AD in individuals with DS. We review key contributors such as amyloid-β accumulation, mitochondrial dysfunction, oxidative stress, tau pathology, neuroinflammation, and chromosomal and epigenetic instability in the neuropathology of AD in DS. Particular attention is given to genes, microRNAs, and chromatin remodeling factors encoded by human chromosome 21 (Hsa21) that regulate these pathological processes. We also highlight the roles of non-coding RNAs and altered DNA methylation patterns in modulating gene expression and neuronal vulnerability. Additionally, the writing evaluates current pharmacological and non-pharmacological interventions and addresses the critical need for inclusive, person-centered health services. Integrating molecular biology with clinical perspectives, the review emphasizes the importance of early diagnosis and coordinated care strategies for individuals with DS at risk for AD.
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a leading cause of chronic liver disease worldwide. Its prevalence is increasing due to its close relationship with obesity, insulin resistance, and other metabolic disorders. In this context, the gut-liver axis has been identified as a fundamental regulator in the progression of MASLD, integrating metabolic, immunological, and inflammatory signals that influence hepatic homeostasis. This article reviews the interconnection between the intestine and the liver in the onset and progression of MASLD, highlighting the roles of cholesterol and its metabolism, intestinal barrier permeability, microbiota, and hepatic signaling pathways. We analyze how intestinal dysbiosis and alterations in the enterohepatic circulation of bile acids affect cholesterol absorption and metabolism. Furthermore, we address the influence of endotoxin translocation, activation of the innate immune system, and the interaction of key transcription factors on disease progression from steatosis to advanced fibrosis and hepatocellular carcinoma (HCC). Finally, therapeutic strategies, including pharmacological, dietary, and immunomodulation-based approaches, are discussed to regulate cholesterol metabolism, modulate the intestinal microbiota, and restore gut-liver axis homeostasis. Integrating this knowledge could open new perspectives for treating and preventing MASLD, addressing the disease from a broader and multidisciplinary viewpoint.
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a leading cause of chronic liver disease worldwide. Its prevalence is increasing due to its close relationship with obesity, insulin resistance, and other metabolic disorders. In this context, the gut-liver axis has been identified as a fundamental regulator in the progression of MASLD, integrating metabolic, immunological, and inflammatory signals that influence hepatic homeostasis. This article reviews the interconnection between the intestine and the liver in the onset and progression of MASLD, highlighting the roles of cholesterol and its metabolism, intestinal barrier permeability, microbiota, and hepatic signaling pathways. We analyze how intestinal dysbiosis and alterations in the enterohepatic circulation of bile acids affect cholesterol absorption and metabolism. Furthermore, we address the influence of endotoxin translocation, activation of the innate immune system, and the interaction of key transcription factors on disease progression from steatosis to advanced fibrosis and hepatocellular carcinoma (HCC). Finally, therapeutic strategies, including pharmacological, dietary, and immunomodulation-based approaches, are discussed to regulate cholesterol metabolism, modulate the intestinal microbiota, and restore gut-liver axis homeostasis. Integrating this knowledge could open new perspectives for treating and preventing MASLD, addressing the disease from a broader and multidisciplinary viewpoint.
The science of manipulating matter at almost atomic scales to create new structures and devices that function at nanoscale dimensions is known as nanotechnology, which is essential to many sciences, such as medicine and environment. This field of study has been reported to investigate better alternatives for the advancement of medicine; one such alternative is the use of plants, which contain substantial amounts of essential phytochemicals. This study aims to utilize such a plant species, Canna indica (C. indica) leaves, known as traditional medicinal plants or commonly grown plants, to synthesize silver nanoparticles (AgNPs) and evaluate their potential in green medicine.
The synthesis was carried out using five varieties of leaf water extracts: Pink red, Yellow, Pink, Yellow red, and Red, under different conditions, to which scanning electron microscopy was performed. The antioxidant capacity was evaluated by total flavonoid content, total phenolic content, total antioxidant capacity, and 2,2-diphenyl-1-picrylhydrazyl radical scavenging assay. The antibacterial activity of AgNPs and water extracts was evaluated against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli). Finally, the cytotoxicity of AgNP is evaluated using the brine shrimp lethality assay.
The optimum condition for AgNP synthesis was determined to be room temperature, and Pink_AgNPs were observed as spherical with a size of 27–48 nm in scanning electron microscopy. The antioxidant assays concluded that AgNPs show significantly higher antioxidant capacity and exhibit higher scavenging activity. This study’s findings showed the efficiency of AgNPs against both strains, and higher efficiency against S. aureus. It was observed that with 240 ppm of AgNPs, 100% viability is obtained.
These novel findings emphasize the significance of C. indica AgNPs, their promise in the medical field, and their application in manufacturing green medicine for environmentally friendly healthcare.
The science of manipulating matter at almost atomic scales to create new structures and devices that function at nanoscale dimensions is known as nanotechnology, which is essential to many sciences, such as medicine and environment. This field of study has been reported to investigate better alternatives for the advancement of medicine; one such alternative is the use of plants, which contain substantial amounts of essential phytochemicals. This study aims to utilize such a plant species, Canna indica (C. indica) leaves, known as traditional medicinal plants or commonly grown plants, to synthesize silver nanoparticles (AgNPs) and evaluate their potential in green medicine.
The synthesis was carried out using five varieties of leaf water extracts: Pink red, Yellow, Pink, Yellow red, and Red, under different conditions, to which scanning electron microscopy was performed. The antioxidant capacity was evaluated by total flavonoid content, total phenolic content, total antioxidant capacity, and 2,2-diphenyl-1-picrylhydrazyl radical scavenging assay. The antibacterial activity of AgNPs and water extracts was evaluated against Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli). Finally, the cytotoxicity of AgNP is evaluated using the brine shrimp lethality assay.
The optimum condition for AgNP synthesis was determined to be room temperature, and Pink_AgNPs were observed as spherical with a size of 27–48 nm in scanning electron microscopy. The antioxidant assays concluded that AgNPs show significantly higher antioxidant capacity and exhibit higher scavenging activity. This study’s findings showed the efficiency of AgNPs against both strains, and higher efficiency against S. aureus. It was observed that with 240 ppm of AgNPs, 100% viability is obtained.
These novel findings emphasize the significance of C. indica AgNPs, their promise in the medical field, and their application in manufacturing green medicine for environmentally friendly healthcare.
Cancer remains one of the leading causes of morbidity and mortality globally, driven by genetic alterations, uncontrolled cell proliferation, and metabolic reprogramming. The tumor microenvironment (TME) is a highly dynamic and heterogeneous system composed of tumor cells, immune cells, stromal cells, and extracellular matrix (ECM) components, which influence cancer progression. Tumor-associated macrophages (TAMs), especially those polarized into the M2 phenotype, play a critical role in modulating this environment. M2 macrophages promote tumor progression through mechanisms such as immune suppression, angiogenesis, and metastasis. This polarization is heavily influenced by the altered metabolic landscape of tumors, where the Warburg effect leads to excessive lactate production, which in turn drives M2 polarization through G protein-coupled receptor 132 (GPR132). M2 macrophages secrete cytokines like IL-10, transforming growth factor β (TGF-β), and vascular endothelial growth factor (VEGF), which contribute to immune escape, tumor growth, and metastasis. The metabolic shifts within TAMs, especially the transition from oxidative phosphorylation to glycolysis, further support the pro-tumoral functions of these cells. This review explores the intricate relationship between M2 macrophage polarization bias, tumor metabolism, and the resulting impact on cancer progression, highlighting the potential of targeting these pathways for therapeutic strategies. The findings suggest that M2 macrophage polarization could serve as a key prognostic factor for cancer outcomes and provide a basis for future research into therapeutic interventions that target macrophage polarization and the tumor metabolic milieu.
Cancer remains one of the leading causes of morbidity and mortality globally, driven by genetic alterations, uncontrolled cell proliferation, and metabolic reprogramming. The tumor microenvironment (TME) is a highly dynamic and heterogeneous system composed of tumor cells, immune cells, stromal cells, and extracellular matrix (ECM) components, which influence cancer progression. Tumor-associated macrophages (TAMs), especially those polarized into the M2 phenotype, play a critical role in modulating this environment. M2 macrophages promote tumor progression through mechanisms such as immune suppression, angiogenesis, and metastasis. This polarization is heavily influenced by the altered metabolic landscape of tumors, where the Warburg effect leads to excessive lactate production, which in turn drives M2 polarization through G protein-coupled receptor 132 (GPR132). M2 macrophages secrete cytokines like IL-10, transforming growth factor β (TGF-β), and vascular endothelial growth factor (VEGF), which contribute to immune escape, tumor growth, and metastasis. The metabolic shifts within TAMs, especially the transition from oxidative phosphorylation to glycolysis, further support the pro-tumoral functions of these cells. This review explores the intricate relationship between M2 macrophage polarization bias, tumor metabolism, and the resulting impact on cancer progression, highlighting the potential of targeting these pathways for therapeutic strategies. The findings suggest that M2 macrophage polarization could serve as a key prognostic factor for cancer outcomes and provide a basis for future research into therapeutic interventions that target macrophage polarization and the tumor metabolic milieu.
Hypersensitivity reactions (HSRs) to paclitaxel, particularly those mediated by the solubilizer Cremophor® EL, are common, occurring in approximately 10% of patients despite premedication. Nab-paclitaxel, a newer formulation using human serum albumin as the vehicle, is generally considered a safer alternative due to a lower rate of HSRs. We present the case of a 44-year-old woman with breast cancer who developed severe HSRs following multiple doses of paclitaxel and carboplatin. Despite standard premedication, she experienced fever, erythematous skin eruptions, arthralgias, and systemic symptoms following her fourth and fifth cycles of treatment. Subsequent administration of nab-paclitaxel also elicited a similar severe reaction. Skin testing revealed a positive reaction to paclitaxel, but not to carboplatin, suggesting sensitization to paclitaxel. In the context of the similar reaction to nab-paclitaxel, this suggests sensitization to the taxane moiety itself rather than to the solubilizer. The combination of features consistent with both type IV hypersensitivity and cytokine release syndrome further complicates the presentation as well. To our knowledge, this is the first reported case of cross-reactivity between paclitaxel and nab-paclitaxel, challenging the assumption that nab-paclitaxel is always a safe alternative. This emphasizes the need for vigilance and thorough evaluation in patients experiencing atypical chemotherapy reactions, as cytokine release reactions may play a role even in the absence of immunotherapy. It also raises the concern that alternative formulations like nab-paclitaxel may not always be safe in patients with atypical or severe reactions, as they could possibly be sensitized to the taxane moiety itself.
Hypersensitivity reactions (HSRs) to paclitaxel, particularly those mediated by the solubilizer Cremophor® EL, are common, occurring in approximately 10% of patients despite premedication. Nab-paclitaxel, a newer formulation using human serum albumin as the vehicle, is generally considered a safer alternative due to a lower rate of HSRs. We present the case of a 44-year-old woman with breast cancer who developed severe HSRs following multiple doses of paclitaxel and carboplatin. Despite standard premedication, she experienced fever, erythematous skin eruptions, arthralgias, and systemic symptoms following her fourth and fifth cycles of treatment. Subsequent administration of nab-paclitaxel also elicited a similar severe reaction. Skin testing revealed a positive reaction to paclitaxel, but not to carboplatin, suggesting sensitization to paclitaxel. In the context of the similar reaction to nab-paclitaxel, this suggests sensitization to the taxane moiety itself rather than to the solubilizer. The combination of features consistent with both type IV hypersensitivity and cytokine release syndrome further complicates the presentation as well. To our knowledge, this is the first reported case of cross-reactivity between paclitaxel and nab-paclitaxel, challenging the assumption that nab-paclitaxel is always a safe alternative. This emphasizes the need for vigilance and thorough evaluation in patients experiencing atypical chemotherapy reactions, as cytokine release reactions may play a role even in the absence of immunotherapy. It also raises the concern that alternative formulations like nab-paclitaxel may not always be safe in patients with atypical or severe reactions, as they could possibly be sensitized to the taxane moiety itself.
This study attempts to offer a viable and sustainable solution related to the tomato value chain, which plays an active role in human diets but deteriorates very fast due to its short shelf life.
Fresh lemons (Citrus limon) and tomatoes (Solanum lycopersicum L.) were purchased from the local market. Previously, varying percentages of lemon juice (0%, 1%, 3%, 5%, and 10%) were added to clear jars containing peeled and pasted tomatoes, which were then sterilized. The physicochemical, antioxidant, nutritional, and microbiological characteristics of appertized tomato samples were assessed through the use of standardized techniques.
The addition of lemon juice significantly (P ˂ 0.05) reduced the total phenolic content of appertized tomatoes, while increasing the titratable acidity (P ˂ 0.05) and decreasing the hydrogen potential (pH) content (P ˂ 0.05). However, the addition of 10 g of lemon juice recorded the high flavonoid content (0.01 mg CE/g) and carotenoid content (16.52 mg/100 g) of samples. In terms of nutritional value, adding lemon juice to appertized tomatoes considerably reduced (P ˂ 0.05) their protein content while increasing their carbohydrate content. Regarding the mineral composition, the addition of lemon juice considerably (P ˂ 0.05) raised the amounts of calcium (Ca), phosphorus (P), and magnesium (Mg) in the appertized tomato samples. The results of this investigation fall within the ranges of the daily allowances that are advised. Pathogens including Salmonella, Clostridium, and Escherichia coli are inhibited, and yeasts and molds are destroyed, ensuring the product’s microbiological quality [476.57 to 0 colony-forming unit (CFU)].
Lemon juice helps to preserve consumer health and improve the preservation of appertized tomatoes.
This study attempts to offer a viable and sustainable solution related to the tomato value chain, which plays an active role in human diets but deteriorates very fast due to its short shelf life.
Fresh lemons (Citrus limon) and tomatoes (Solanum lycopersicum L.) were purchased from the local market. Previously, varying percentages of lemon juice (0%, 1%, 3%, 5%, and 10%) were added to clear jars containing peeled and pasted tomatoes, which were then sterilized. The physicochemical, antioxidant, nutritional, and microbiological characteristics of appertized tomato samples were assessed through the use of standardized techniques.
The addition of lemon juice significantly (P ˂ 0.05) reduced the total phenolic content of appertized tomatoes, while increasing the titratable acidity (P ˂ 0.05) and decreasing the hydrogen potential (pH) content (P ˂ 0.05). However, the addition of 10 g of lemon juice recorded the high flavonoid content (0.01 mg CE/g) and carotenoid content (16.52 mg/100 g) of samples. In terms of nutritional value, adding lemon juice to appertized tomatoes considerably reduced (P ˂ 0.05) their protein content while increasing their carbohydrate content. Regarding the mineral composition, the addition of lemon juice considerably (P ˂ 0.05) raised the amounts of calcium (Ca), phosphorus (P), and magnesium (Mg) in the appertized tomato samples. The results of this investigation fall within the ranges of the daily allowances that are advised. Pathogens including Salmonella, Clostridium, and Escherichia coli are inhibited, and yeasts and molds are destroyed, ensuring the product’s microbiological quality [476.57 to 0 colony-forming unit (CFU)].
Lemon juice helps to preserve consumer health and improve the preservation of appertized tomatoes.
Scientific evidence seems to indicate that, in males, intense and prolonged endurance sport can favor the onset of atrial fibrillation. A plausible explanation may be the impact that intense endurance sports produce on the three vertices of Coumel’s triangle. However, genetics is probably also involved in translating this impact into an arrhythmic phenotype. On a management level, the first task of the cardiologist is to exclude the presence of structural heart disease, channelopathy, endocrine and/or electrolyte disorders, and substance use. As for the treatment of arrhythmia, the “CARE” paradigm proposed by the latest ESC guidelines should probably be accompanied by detraining, although this suggestion is often rejected by the athlete. Anticoagulant therapy, where indicated, must take into account the risk of trauma that the sport entails, even if the particular pharmacodynamics/pharmacokinetics of DOACs should allow training/competition to take place when the anticoagulant effect of the previous administration has completely or almost completely worn off.
Scientific evidence seems to indicate that, in males, intense and prolonged endurance sport can favor the onset of atrial fibrillation. A plausible explanation may be the impact that intense endurance sports produce on the three vertices of Coumel’s triangle. However, genetics is probably also involved in translating this impact into an arrhythmic phenotype. On a management level, the first task of the cardiologist is to exclude the presence of structural heart disease, channelopathy, endocrine and/or electrolyte disorders, and substance use. As for the treatment of arrhythmia, the “CARE” paradigm proposed by the latest ESC guidelines should probably be accompanied by detraining, although this suggestion is often rejected by the athlete. Anticoagulant therapy, where indicated, must take into account the risk of trauma that the sport entails, even if the particular pharmacodynamics/pharmacokinetics of DOACs should allow training/competition to take place when the anticoagulant effect of the previous administration has completely or almost completely worn off.
Polymer-based nanoparticles have emerged as powerful multifunctional platforms in cancer theranostics, offering the ability to integrate diagnostic imaging and targeted therapy within a single system. These nanocarriers enable improved tumor localization, enhanced contrast agent delivery, and controlled therapeutic release, addressing limitations associated with conventional contrast agents such as poor specificity, rapid clearance, and systemic toxicity. Advances in polymer chemistry and nanoparticle fabrication methods, including solvent evaporation, nanoprecipitation, emulsion-diffusion, and emulsion polymerization, have allowed precise control over particle size, surface charge, and drug-loading efficiency, optimizing biodistribution and imaging performance. Hybrid polymer-inorganic nanoparticles further expand functionality by incorporating magnetic, optical, or radiopaque components, enabling multimodal imaging and stimuli-responsive drug release while maintaining biocompatibility. Key factors influencing the efficiency of polymer nanoparticle-based contrast agents include physicochemical properties such as particle size, morphology, surface functionalization, and responsiveness to tumor microenvironmental stimuli. These attributes collectively govern circulation time, cellular uptake, and accumulation in tumor tissues via passive and active targeting strategies. While promising, the clinical translation of these systems faces challenges including immunogenicity, pharmacokinetic variability, long-term safety concerns, and manufacturing scalability. Recent innovations in ligand functionalization, biomimetic coatings, and multifunctional nanoparticle design continue to advance therapeutic specificity and imaging precision, positioning polymer nanoparticles as versatile candidates for personalized oncologic care. This review provides a comprehensive synthesis of current methods for contrast agent integration, the role of physicochemical properties in performance, biological interactions, safety considerations, recent design innovations, translational barriers, and future research directions for polymer nanoparticle-based cancer theranostics.
Polymer-based nanoparticles have emerged as powerful multifunctional platforms in cancer theranostics, offering the ability to integrate diagnostic imaging and targeted therapy within a single system. These nanocarriers enable improved tumor localization, enhanced contrast agent delivery, and controlled therapeutic release, addressing limitations associated with conventional contrast agents such as poor specificity, rapid clearance, and systemic toxicity. Advances in polymer chemistry and nanoparticle fabrication methods, including solvent evaporation, nanoprecipitation, emulsion-diffusion, and emulsion polymerization, have allowed precise control over particle size, surface charge, and drug-loading efficiency, optimizing biodistribution and imaging performance. Hybrid polymer-inorganic nanoparticles further expand functionality by incorporating magnetic, optical, or radiopaque components, enabling multimodal imaging and stimuli-responsive drug release while maintaining biocompatibility. Key factors influencing the efficiency of polymer nanoparticle-based contrast agents include physicochemical properties such as particle size, morphology, surface functionalization, and responsiveness to tumor microenvironmental stimuli. These attributes collectively govern circulation time, cellular uptake, and accumulation in tumor tissues via passive and active targeting strategies. While promising, the clinical translation of these systems faces challenges including immunogenicity, pharmacokinetic variability, long-term safety concerns, and manufacturing scalability. Recent innovations in ligand functionalization, biomimetic coatings, and multifunctional nanoparticle design continue to advance therapeutic specificity and imaging precision, positioning polymer nanoparticles as versatile candidates for personalized oncologic care. This review provides a comprehensive synthesis of current methods for contrast agent integration, the role of physicochemical properties in performance, biological interactions, safety considerations, recent design innovations, translational barriers, and future research directions for polymer nanoparticle-based cancer theranostics.
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection (LRTI) burden among infants. Maternal vaccination is a promising preventive strategy, conferring passive immunity through transplacental antibody transfer. The current narrative review was conducted to summarize the current evidence of efficacy and safety of maternal RSV vaccination and assess the practical barriers to its implementation. This review was based on a structured literature search of PubMed/MEDLINE and Google Scholar to identify peer-reviewed studies published between January 2022 and March 2025 using terms such as “maternal RSV vaccine”, “efficacy”, “safety”, “pregnancy”, “Abrysvo”, and “hesitancy”. The review included 5 clinical trials evaluating maternal RSV vaccines and 17 observational and survey studies assessing vaccine acceptance across diverse settings. The bivalent RSVpreF vaccine (Abrysvo) is the only licensed maternal RSV vaccine as of May 2025. In the MATISSE phase 3 trial (n = 7,358), the vaccine demonstrated 81.8% efficacy against medically attended RSV-LRTI at 90 days and 69.4% at 180 days, with 57.1% efficacy against severe RSV-LRTI. No major safety concerns were identified; adverse events and preterm birth rates were comparable between groups. In contrast, trials of GSK’s RSVPreF3-Mat vaccine revealed higher rates of preterm birth (6.8% vs. 4.9%) and a numerical imbalance in infant deaths (0.4% vs. 0.2%), prompting early termination. Across 17 studies (n = 14,959), RSV vaccine acceptance ranged from 39% (France) to 87% (Netherlands), with safety concerns and cultural context influencing attitudes. This review highlights that maternal RSV vaccination with RSVpreF offers effective infant protection with an acceptable safety profile. Future research should focus on long-term infant outcomes, comparative effectiveness in diverse settings, and next-generation vaccines. Implementation will require public trust, cultural sensitivity, and equitable global access.
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection (LRTI) burden among infants. Maternal vaccination is a promising preventive strategy, conferring passive immunity through transplacental antibody transfer. The current narrative review was conducted to summarize the current evidence of efficacy and safety of maternal RSV vaccination and assess the practical barriers to its implementation. This review was based on a structured literature search of PubMed/MEDLINE and Google Scholar to identify peer-reviewed studies published between January 2022 and March 2025 using terms such as “maternal RSV vaccine”, “efficacy”, “safety”, “pregnancy”, “Abrysvo”, and “hesitancy”. The review included 5 clinical trials evaluating maternal RSV vaccines and 17 observational and survey studies assessing vaccine acceptance across diverse settings. The bivalent RSVpreF vaccine (Abrysvo) is the only licensed maternal RSV vaccine as of May 2025. In the MATISSE phase 3 trial (n = 7,358), the vaccine demonstrated 81.8% efficacy against medically attended RSV-LRTI at 90 days and 69.4% at 180 days, with 57.1% efficacy against severe RSV-LRTI. No major safety concerns were identified; adverse events and preterm birth rates were comparable between groups. In contrast, trials of GSK’s RSVPreF3-Mat vaccine revealed higher rates of preterm birth (6.8% vs. 4.9%) and a numerical imbalance in infant deaths (0.4% vs. 0.2%), prompting early termination. Across 17 studies (n = 14,959), RSV vaccine acceptance ranged from 39% (France) to 87% (Netherlands), with safety concerns and cultural context influencing attitudes. This review highlights that maternal RSV vaccination with RSVpreF offers effective infant protection with an acceptable safety profile. Future research should focus on long-term infant outcomes, comparative effectiveness in diverse settings, and next-generation vaccines. Implementation will require public trust, cultural sensitivity, and equitable global access.
Oncolytic virotherapy (OVT) employs genetically engineered or naturally occurring viruses to selectively replicate within tumor cells, leading to direct lysis and induction of systemic anti-tumor immune responses. This dual mechanism distinguishes OVT from conventional therapies and positions it as a promising candidate in precision oncology. This review synthesizes recent advancements in understanding the molecular mechanisms underlying OVT efficacy, including viral entry, replication kinetics, immunogenic cell death, and modulation of the tumor microenvironment. We highlight innovations in viral engineering, such as promoter targeting, microRNA control, and immune-modulatory gene insertions that enhance tumor specificity and therapeutic safety. Clinically, OVT has shown measurable benefits in various solid tumors, with several viruses, such as talimogene laherparepvec, entering regulatory approval and others progressing through late-phase clinical trials. When combined with immune checkpoint inhibitors, OVT has demonstrated synergistic effects by improving antigen presentation and reversing immunosuppressive signaling. Integration with targeted therapies and nanotechnology-based delivery systems has further refined viral biodistribution and pharmacodynamics. However, therapeutic resistance, immune clearance, stromal barriers, and heterogeneous tumor responses remain key limitations. Overcoming these challenges requires optimized delivery routes, predictive biomarkers, and combination strategies tailored to immune and genetic tumor profiles. As OVT evolves from proof-of-concept to a platform-based therapeutic strategy, its integration into multimodal cancer treatment protocols will depend on refined bridge oncolytic activity with durable immunotherapy effects.
Oncolytic virotherapy (OVT) employs genetically engineered or naturally occurring viruses to selectively replicate within tumor cells, leading to direct lysis and induction of systemic anti-tumor immune responses. This dual mechanism distinguishes OVT from conventional therapies and positions it as a promising candidate in precision oncology. This review synthesizes recent advancements in understanding the molecular mechanisms underlying OVT efficacy, including viral entry, replication kinetics, immunogenic cell death, and modulation of the tumor microenvironment. We highlight innovations in viral engineering, such as promoter targeting, microRNA control, and immune-modulatory gene insertions that enhance tumor specificity and therapeutic safety. Clinically, OVT has shown measurable benefits in various solid tumors, with several viruses, such as talimogene laherparepvec, entering regulatory approval and others progressing through late-phase clinical trials. When combined with immune checkpoint inhibitors, OVT has demonstrated synergistic effects by improving antigen presentation and reversing immunosuppressive signaling. Integration with targeted therapies and nanotechnology-based delivery systems has further refined viral biodistribution and pharmacodynamics. However, therapeutic resistance, immune clearance, stromal barriers, and heterogeneous tumor responses remain key limitations. Overcoming these challenges requires optimized delivery routes, predictive biomarkers, and combination strategies tailored to immune and genetic tumor profiles. As OVT evolves from proof-of-concept to a platform-based therapeutic strategy, its integration into multimodal cancer treatment protocols will depend on refined bridge oncolytic activity with durable immunotherapy effects.
This study evaluates the in vitro and in vivo antiplasmodial, hemolytic, and antioxidant activities of a combined extract of Ageratum conyzoides (A. conyzoides) and Bidens pilosa (B. pilosa), a traditionally used but scientifically unvalidated combination.
Plant leaves were extracted via aqueous decoction and cold maceration, combining equal parts to mimic traditional preparation. In vitro antiplasmodial activity against the chloroquine-sensitive Plasmodium falciparum 3D7 (Pf3D7) strain was assessed using the SYBR Green I assay. Cytotoxicity was evaluated via hemolysis test, and antioxidant potential using DPPH (2,2-diphenyl-1-picrylhydrazyl), ABTS [2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)], and FRAP (ferric ion reducing antioxidant potential) assays. The most potent combination was tested for acute toxicity and curative antimalarial activity in a rodent model.
Extract yields ranged from 6.6% (cold maceration extract of B. pilosa) to 29.2% [aqueous decoction extract of combination (Cd)]. Extracts showed moderate to mild in vitro antiplasmodial activity [IC50 (median inhibitory concentration): 24.8–96.6 µg/mL], with the aqueous Cd showing potential synergism [CI (combination index) < 1]. No significant cytotoxicity was observed (< 10% hemolysis). Moderate to good antioxidant activity was found in DPPH [SC50 (median scavenging concentration): 134.65–307.55 µg/mL] and ABTS assays (SC50: 92.23–183.45 µg/mL), with Cd showing the highest activity. FRAP values were low. The Cd extract demonstrated no significant acute toxicity up to 5,000 mg/kg and significant in vivo antimalarial activity, achieving 65% parasite inhibition at 200 mg/kg/day. It also prolonged survival time, with a maximum survival of 28 days at 200 mg/kg/day.
This preliminary investigation suggests that combined extracts of A. conyzoides and B. pilosa exhibit noteworthy in vitro and in vivo antiplasmodial activity against the tested strains. Further studies are warranted to validate these findings and develop optimized formulations as potential antimalarials.
This study evaluates the in vitro and in vivo antiplasmodial, hemolytic, and antioxidant activities of a combined extract of Ageratum conyzoides (A. conyzoides) and Bidens pilosa (B. pilosa), a traditionally used but scientifically unvalidated combination.
Plant leaves were extracted via aqueous decoction and cold maceration, combining equal parts to mimic traditional preparation. In vitro antiplasmodial activity against the chloroquine-sensitive Plasmodium falciparum 3D7 (Pf3D7) strain was assessed using the SYBR Green I assay. Cytotoxicity was evaluated via hemolysis test, and antioxidant potential using DPPH (2,2-diphenyl-1-picrylhydrazyl), ABTS [2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)], and FRAP (ferric ion reducing antioxidant potential) assays. The most potent combination was tested for acute toxicity and curative antimalarial activity in a rodent model.
Extract yields ranged from 6.6% (cold maceration extract of B. pilosa) to 29.2% [aqueous decoction extract of combination (Cd)]. Extracts showed moderate to mild in vitro antiplasmodial activity [IC50 (median inhibitory concentration): 24.8–96.6 µg/mL], with the aqueous Cd showing potential synergism [CI (combination index) < 1]. No significant cytotoxicity was observed (< 10% hemolysis). Moderate to good antioxidant activity was found in DPPH [SC50 (median scavenging concentration): 134.65–307.55 µg/mL] and ABTS assays (SC50: 92.23–183.45 µg/mL), with Cd showing the highest activity. FRAP values were low. The Cd extract demonstrated no significant acute toxicity up to 5,000 mg/kg and significant in vivo antimalarial activity, achieving 65% parasite inhibition at 200 mg/kg/day. It also prolonged survival time, with a maximum survival of 28 days at 200 mg/kg/day.
This preliminary investigation suggests that combined extracts of A. conyzoides and B. pilosa exhibit noteworthy in vitro and in vivo antiplasmodial activity against the tested strains. Further studies are warranted to validate these findings and develop optimized formulations as potential antimalarials.
Mucopolysaccharidosis type IIIB (MPS IIIB), or Sanfilippo Syndrome type B, is a lysosomal storage disorder caused by mutations in the NAGLU gene, which encodes the enzyme alpha-N-acetylglucosaminidase, responsible for the degradation of heparan sulfate. Progressive accumulation of undegraded glycosaminoglycans primarily affects the central nervous system, resulting in severe neurodegeneration. Cellular findings reveal impaired intracellular trafficking, especially within the Golgi apparatus, linked to GM130 depletion and accumulation of GM2 and GM3 gangliosides. Endocytic vesicles fail to properly fuse with lysosomes due to genetic defects, disrupting lysosomal degradation. This contributes to oxidative stress, mitochondrial dysfunction, and mitophagy failure, which collectively drive neuronal apoptosis. MPS IIIB shares pathways with Alzheimer’s and Parkinson’s, suggesting cellular aging processes. Given the lack of specific treatment, modulation of inflammatory pathways such as TLR4 emerges as a potential therapeutic strategy.
Mucopolysaccharidosis type IIIB (MPS IIIB), or Sanfilippo Syndrome type B, is a lysosomal storage disorder caused by mutations in the NAGLU gene, which encodes the enzyme alpha-N-acetylglucosaminidase, responsible for the degradation of heparan sulfate. Progressive accumulation of undegraded glycosaminoglycans primarily affects the central nervous system, resulting in severe neurodegeneration. Cellular findings reveal impaired intracellular trafficking, especially within the Golgi apparatus, linked to GM130 depletion and accumulation of GM2 and GM3 gangliosides. Endocytic vesicles fail to properly fuse with lysosomes due to genetic defects, disrupting lysosomal degradation. This contributes to oxidative stress, mitochondrial dysfunction, and mitophagy failure, which collectively drive neuronal apoptosis. MPS IIIB shares pathways with Alzheimer’s and Parkinson’s, suggesting cellular aging processes. Given the lack of specific treatment, modulation of inflammatory pathways such as TLR4 emerges as a potential therapeutic strategy.
Angiogenesis, invasion, and tube formation are critical processes in tumor progression and metastasis. The use of nanoparticles derived from natural products presents a promising approach for targeted cancer therapy. This study evaluates the anti-angiogenic and anti-invasive effects of Moringa oleifera silver nanoparticles (MO-AgNPs) as a therapeutic strategy against these processes.
The anti-angiogenic and anti-invasive activities of MO-AgNPs were investigated using a series of in vitro and ex vivo models. These included the rat aortic ring assay, endothelial tube formation assay, cell invasion assay using endothelial cell lines (Ea.hy926), and a three-dimensional (3D) co-culture spheroid model to simulate tumor microenvironment behavior. Comparisons were made with known inhibitors: quercetin (15.11 μg/mL) and suramin (100 μg/mL).
MO-AgNPs at 12 μg/mL significantly inhibited Ea.hy926 cell invasion by 62.10% and significantly suppressed endothelial tube formation, comparable to the effect of quercetin. In the ex vivo aortic ring assay, MO-AgNPs reduced microvessel sprouting by 83.824 ± 0.081%, surpassing the inhibition achieved by suramin. Additionally, in the 3D spheroid model, MO-AgNPs at concentrations of 12 μg/mL and 6 μg/mL, as well as quercetin, significantly reduced spheroid diameter by day 14, indicating suppressed invasive potential and angiogenic support.
MO-AgNPs exhibit strong anti-angiogenic and anti-invasive effects across various tumor-relevant models, highlighting their potential as a therapeutic agent against tumor progression and angiogenesis-related diseases. These results support further investigation of MO-AgNPs as a novel nanotherapeutic for cancer treatment.
Angiogenesis, invasion, and tube formation are critical processes in tumor progression and metastasis. The use of nanoparticles derived from natural products presents a promising approach for targeted cancer therapy. This study evaluates the anti-angiogenic and anti-invasive effects of Moringa oleifera silver nanoparticles (MO-AgNPs) as a therapeutic strategy against these processes.
The anti-angiogenic and anti-invasive activities of MO-AgNPs were investigated using a series of in vitro and ex vivo models. These included the rat aortic ring assay, endothelial tube formation assay, cell invasion assay using endothelial cell lines (Ea.hy926), and a three-dimensional (3D) co-culture spheroid model to simulate tumor microenvironment behavior. Comparisons were made with known inhibitors: quercetin (15.11 μg/mL) and suramin (100 μg/mL).
MO-AgNPs at 12 μg/mL significantly inhibited Ea.hy926 cell invasion by 62.10% and significantly suppressed endothelial tube formation, comparable to the effect of quercetin. In the ex vivo aortic ring assay, MO-AgNPs reduced microvessel sprouting by 83.824 ± 0.081%, surpassing the inhibition achieved by suramin. Additionally, in the 3D spheroid model, MO-AgNPs at concentrations of 12 μg/mL and 6 μg/mL, as well as quercetin, significantly reduced spheroid diameter by day 14, indicating suppressed invasive potential and angiogenic support.
MO-AgNPs exhibit strong anti-angiogenic and anti-invasive effects across various tumor-relevant models, highlighting their potential as a therapeutic agent against tumor progression and angiogenesis-related diseases. These results support further investigation of MO-AgNPs as a novel nanotherapeutic for cancer treatment.
Neurofibromatosis type 1 (NF1) is a hereditary, autosomal dominant condition marked by the development of tumors along the nervous system due to uncontrolled cell proliferation. The current case reports a 31-year-old male patient diagnosed with NF1 with the involvement of bilateral pheochromocytomas and colonic inflammatory polyps/leiomyoma. A genetic profile was explored through whole-exome sequencing to identify pathogenic variants, and segregation analysis was subsequently performed in the patient’s family. Sequencing analysis revealed a novel heterozygous frameshift variant, NF1 c.7301dupA (p.S2435Efs*11), which was identified as the pathogenic variant in the patient. Additionally, two identified variants, PMS2 c.2T>C (p.M1T) and MUTYH c.850-2A>G, may be associated with colonic tumor conditions in the patient. These findings provide insights into the molecular etiology underlying this rare presentation of multiple tumors in a Vietnamese male and may contribute to improved treatment planning and patient management.
Neurofibromatosis type 1 (NF1) is a hereditary, autosomal dominant condition marked by the development of tumors along the nervous system due to uncontrolled cell proliferation. The current case reports a 31-year-old male patient diagnosed with NF1 with the involvement of bilateral pheochromocytomas and colonic inflammatory polyps/leiomyoma. A genetic profile was explored through whole-exome sequencing to identify pathogenic variants, and segregation analysis was subsequently performed in the patient’s family. Sequencing analysis revealed a novel heterozygous frameshift variant, NF1 c.7301dupA (p.S2435Efs*11), which was identified as the pathogenic variant in the patient. Additionally, two identified variants, PMS2 c.2T>C (p.M1T) and MUTYH c.850-2A>G, may be associated with colonic tumor conditions in the patient. These findings provide insights into the molecular etiology underlying this rare presentation of multiple tumors in a Vietnamese male and may contribute to improved treatment planning and patient management.
The study evaluated the influence of different wall materials on the bioactive compounds in encapsulated Justicia carnea leaves extract.
Combinations of gelatin with maltodextrin or starch, and gum arabic with maltodextrin or starch were prepared in ratios of 1:3 to create four types of wall matrices. Each combination was dissolved in water to obtain 20% w/v solutions. J. carnea leaves were extracted, concentrated, and the resulting extract added to the wall material at a ratio of 1:2 and homogenized. Encapsulation was achieved through homogenization at 12,000 rpm for 30 min, followed by freeze drying. The resulting microcapsules were characterized using a scanning electron microscope (SEM) and differential scanning calorimeter (DSC). Physicochemical properties, pigment concentrations, and micronutrient compositions of the microcapsules were also evaluated using standard methods.
Carotenoids, chlorophyll, and anthocyanin were significantly higher (P ≤ 0.05) in the sample containing starch and gelatin [gelatin + starch + core (DGES)] as the wall matrix compared to other samples. Vitamins E and D, calcium (Ca), and manganese (Mn) in sample gum arabic + starch + core (CGS; blend of gum arabic starch) and sample DGES were not significantly different from each other. DGES exhibited significantly lower (60.19%) solubility than others (60.48–70.86%) and the highest (76.72%) encapsulation efficiency. SEM analysis revealed smooth surfaces and mostly polyhedral shapes, with particle sizes ranging from 10.534–14.159 μm across all samples. DSC analysis revealed that the particles are endothermic and amorphous in nature, except for the CGS sample, which became semi-crystalline at about 203.2°C.
The study showed that a composite wall material comprising starch and gelatin demonstrates enhanced effectiveness in the encapsulation of J. carnea leaves bioactive compounds.
The study evaluated the influence of different wall materials on the bioactive compounds in encapsulated Justicia carnea leaves extract.
Combinations of gelatin with maltodextrin or starch, and gum arabic with maltodextrin or starch were prepared in ratios of 1:3 to create four types of wall matrices. Each combination was dissolved in water to obtain 20% w/v solutions. J. carnea leaves were extracted, concentrated, and the resulting extract added to the wall material at a ratio of 1:2 and homogenized. Encapsulation was achieved through homogenization at 12,000 rpm for 30 min, followed by freeze drying. The resulting microcapsules were characterized using a scanning electron microscope (SEM) and differential scanning calorimeter (DSC). Physicochemical properties, pigment concentrations, and micronutrient compositions of the microcapsules were also evaluated using standard methods.
Carotenoids, chlorophyll, and anthocyanin were significantly higher (P ≤ 0.05) in the sample containing starch and gelatin [gelatin + starch + core (DGES)] as the wall matrix compared to other samples. Vitamins E and D, calcium (Ca), and manganese (Mn) in sample gum arabic + starch + core (CGS; blend of gum arabic starch) and sample DGES were not significantly different from each other. DGES exhibited significantly lower (60.19%) solubility than others (60.48–70.86%) and the highest (76.72%) encapsulation efficiency. SEM analysis revealed smooth surfaces and mostly polyhedral shapes, with particle sizes ranging from 10.534–14.159 μm across all samples. DSC analysis revealed that the particles are endothermic and amorphous in nature, except for the CGS sample, which became semi-crystalline at about 203.2°C.
The study showed that a composite wall material comprising starch and gelatin demonstrates enhanced effectiveness in the encapsulation of J. carnea leaves bioactive compounds.