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.
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.
The objective of this study is to ascertain the antimicrobial and anticancer properties of dry Morinda citrifolia L. (noni) pulp hydroalcoholic extracts.
In this study, dry noni samples were immersed in hydro-alcoholic solvents, ethanol (EtOH) and methanol (MeOH). Using the intelligent-flash extractor (KBE-I5) and freeze-vacuum dryer, noni ethanol (NE) and noni methanol (NM) extracts were obtained for antimicrobial testing against bacterial and fungal strains via disc diffusion assay. Cell viability was assessed using the cell counting kit-8 (CCK-8) assay, acridine orange (AO) staining, and western blotting to evaluate anticancer effects on human cancer cells. Novel phytoconstituents were identified using dual-mode ultra-performance liquid chromatography quadrupole exactive orbitrap-tandem mass spectrometer (UPLC-Q-exactive orbitrap-MS/MS) analysis.
Extraction yielded 16.8% for NE and 25.8% for NM. NE minimum inhibitory concentrations (MICs) against Escherichia coli (EC), Saccharomyces cerevisiae (SC), Staphylococcus aureus (SA), and Streptococcus thermophilus (ST) being 177, 52, 388, and 283 mg/mL. NM MICs values were 105, 47, 312, and 135 mg/mL, respectively. Anticancer half inhibitory concentrations (IC50s) for NE against human colon adenocarcinoma cell (HT-29) and human bladder cancer cell lines (UMUC-3) were 758 and 899 µg/mL. For NM, IC50s were 1,231 (HT-29) and 1,173 (UMUC-3) µg/mL. Cell death indicators include organelle deformities, AO fluorescence, and autophagy protein expression. In dual ion-scan mode UPLC analysis, 17 distinct phytoconstituents were identified, including 2-Hydroxycinnamic acid, 4-Hydroxycinnamic acid, and riboflavin, known for treating cancer, metabolic dysfunctions, and COVID-19. The 14 constituents were discovered in noni fruit for the first time.
Noni fruit extracts show antimicrobial/anticancer activity and therapeutic potential, establishing noni fruit as a promising food and medicinal source based on composition.
The objective of this study is to ascertain the antimicrobial and anticancer properties of dry Morinda citrifolia L. (noni) pulp hydroalcoholic extracts.
In this study, dry noni samples were immersed in hydro-alcoholic solvents, ethanol (EtOH) and methanol (MeOH). Using the intelligent-flash extractor (KBE-I5) and freeze-vacuum dryer, noni ethanol (NE) and noni methanol (NM) extracts were obtained for antimicrobial testing against bacterial and fungal strains via disc diffusion assay. Cell viability was assessed using the cell counting kit-8 (CCK-8) assay, acridine orange (AO) staining, and western blotting to evaluate anticancer effects on human cancer cells. Novel phytoconstituents were identified using dual-mode ultra-performance liquid chromatography quadrupole exactive orbitrap-tandem mass spectrometer (UPLC-Q-exactive orbitrap-MS/MS) analysis.
Extraction yielded 16.8% for NE and 25.8% for NM. NE minimum inhibitory concentrations (MICs) against Escherichia coli (EC), Saccharomyces cerevisiae (SC), Staphylococcus aureus (SA), and Streptococcus thermophilus (ST) being 177, 52, 388, and 283 mg/mL. NM MICs values were 105, 47, 312, and 135 mg/mL, respectively. Anticancer half inhibitory concentrations (IC50s) for NE against human colon adenocarcinoma cell (HT-29) and human bladder cancer cell lines (UMUC-3) were 758 and 899 µg/mL. For NM, IC50s were 1,231 (HT-29) and 1,173 (UMUC-3) µg/mL. Cell death indicators include organelle deformities, AO fluorescence, and autophagy protein expression. In dual ion-scan mode UPLC analysis, 17 distinct phytoconstituents were identified, including 2-Hydroxycinnamic acid, 4-Hydroxycinnamic acid, and riboflavin, known for treating cancer, metabolic dysfunctions, and COVID-19. The 14 constituents were discovered in noni fruit for the first time.
Noni fruit extracts show antimicrobial/anticancer activity and therapeutic potential, establishing noni fruit as a promising food and medicinal source based on composition.
Depression is one of the most important mental diseases. Different pharmacological and non-pharmacological methods are used to treat depression. Traditional and complementary medicine also have a special role in the treatment of depression. Among the specific medicinal formulations mentioned in Traditional Persian Medicine (TPM), an important and widely used form is “Mufarrah” (exhilarating), which indirectly refers to the mood-stabilizing group. In this work, a related traditional formulation has been reformulated and standardized as a conventional tablet.
A simple and famous example among this group is “Mufarrah-e-Bared-e-Saghir”, containing Rosa × damascena Herrm., Coriandrum sativum L., Melissa Officinalis L. Following tablet preparation of the mentioned remedy, total phenolic and flavonoid content was determined using the spectrophotometric method. Volatile constituent analysis and quantification of linalool as the main component were carried out via gas chromatography (GC) [GC/MS (mass spectrometry) and GC/FID (flame ionization detector)].
According to the results, the main compound of the final product was linalool (54.6%). Linalool, total phenol, and total flavonoid amounts have been calculated, respectively, 2,379.65 ± 262.13 µg/mL of the extracted essential oil, 163.23 ± 0.61, and 41.41 ± 2.3 mg/g extract.
Prepared tablets as a reformulated traditional medicine product with rich total phenols and flavonoids, as well as the presence of linalool as a considerable icon with antidepressant activities, can be introduced to the Persian medicinal plants market to control depression.
Depression is one of the most important mental diseases. Different pharmacological and non-pharmacological methods are used to treat depression. Traditional and complementary medicine also have a special role in the treatment of depression. Among the specific medicinal formulations mentioned in Traditional Persian Medicine (TPM), an important and widely used form is “Mufarrah” (exhilarating), which indirectly refers to the mood-stabilizing group. In this work, a related traditional formulation has been reformulated and standardized as a conventional tablet.
A simple and famous example among this group is “Mufarrah-e-Bared-e-Saghir”, containing Rosa × damascena Herrm., Coriandrum sativum L., Melissa Officinalis L. Following tablet preparation of the mentioned remedy, total phenolic and flavonoid content was determined using the spectrophotometric method. Volatile constituent analysis and quantification of linalool as the main component were carried out via gas chromatography (GC) [GC/MS (mass spectrometry) and GC/FID (flame ionization detector)].
According to the results, the main compound of the final product was linalool (54.6%). Linalool, total phenol, and total flavonoid amounts have been calculated, respectively, 2,379.65 ± 262.13 µg/mL of the extracted essential oil, 163.23 ± 0.61, and 41.41 ± 2.3 mg/g extract.
Prepared tablets as a reformulated traditional medicine product with rich total phenols and flavonoids, as well as the presence of linalool as a considerable icon with antidepressant activities, can be introduced to the Persian medicinal plants market to control depression.
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a pivotal regulator of cellular redox balance and detoxification, critical for maintaining hepatocyte homeostasis. However, its dysregulation has emerged as a key driver in hepatocellular carcinoma (HCC), the most prevalent form of liver cancer. This review synthesizes recent advancements (2023–2025) to elucidate Nrf2’s context-dependent dual functions: tumor suppressive roles during early carcinogenesis through oxidative stress mitigation, versus oncogenic effects in advanced stages via promoting proliferation, survival, and treatment resistance. We systematically analyze molecular mechanisms of Nrf2 activation, including Kelch-like ECH-associated protein 1 (KEAP1)-dependent/independent pathways and epigenetic regulation, supported by clinical data linking Nrf2 expression to patient prognosis. Preclinical and translational research on Nrf2-targeted therapies is evaluated, with a focus on combinatorial strategies overcoming resistance. Despite challenges in developing selective modulators, integrating multi-omics biomarkers and context-specific interventions offers promise for precision medicine in HCC.
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a pivotal regulator of cellular redox balance and detoxification, critical for maintaining hepatocyte homeostasis. However, its dysregulation has emerged as a key driver in hepatocellular carcinoma (HCC), the most prevalent form of liver cancer. This review synthesizes recent advancements (2023–2025) to elucidate Nrf2’s context-dependent dual functions: tumor suppressive roles during early carcinogenesis through oxidative stress mitigation, versus oncogenic effects in advanced stages via promoting proliferation, survival, and treatment resistance. We systematically analyze molecular mechanisms of Nrf2 activation, including Kelch-like ECH-associated protein 1 (KEAP1)-dependent/independent pathways and epigenetic regulation, supported by clinical data linking Nrf2 expression to patient prognosis. Preclinical and translational research on Nrf2-targeted therapies is evaluated, with a focus on combinatorial strategies overcoming resistance. Despite challenges in developing selective modulators, integrating multi-omics biomarkers and context-specific interventions offers promise for precision medicine in HCC.
The tropics are abundant in both animals and plants, but also in pathogenic agents. There, the world’s greatest burden of diseases and mortality is concentrated. Co-infections are the rule, making laboratory diagnosis complex. Simultaneous multidiagnostic methods are desirable; however, they are mostly expensive and inaccessible to the populations of the region. The aim of our research was to produce synthetic peptides of the most important pathogens that can be used in a simultaneous multidiagnostic technique. Thus, we designed a low-cost method to detect antibodies, the multiple antigen blot assay (MABA), using synthetic peptides as the main source of antigens from endemic tropical diseases. This method allows the simultaneous detection of antibodies against 26 different agents with only a few microliters of sera, plasma, or saliva. The development of this system is the result of a long process, and the pipeline of our approach from then to nowadays is presented. Specific epitopes with the greatest antigenic potential using immunoinformatic algorithms have been selected from worldwide and tropical pathogens and then assayed by a successive chain of immunological techniques [PEPSCAN®, enzyme-linked immunosorbent assay (ELISA), and MABA] to evaluate the sensitivity and specificity of those synthetic peptides for their usefulness in diagnosis. Years of work have been required for this complex process, with the recent incorporation of new immunoinformatic predictive tools, methodologies, and cost advantages. It can be concluded that synthetic peptides are a promising approach for diagnostic processes based either on the detection of antigens or antibodies.
The tropics are abundant in both animals and plants, but also in pathogenic agents. There, the world’s greatest burden of diseases and mortality is concentrated. Co-infections are the rule, making laboratory diagnosis complex. Simultaneous multidiagnostic methods are desirable; however, they are mostly expensive and inaccessible to the populations of the region. The aim of our research was to produce synthetic peptides of the most important pathogens that can be used in a simultaneous multidiagnostic technique. Thus, we designed a low-cost method to detect antibodies, the multiple antigen blot assay (MABA), using synthetic peptides as the main source of antigens from endemic tropical diseases. This method allows the simultaneous detection of antibodies against 26 different agents with only a few microliters of sera, plasma, or saliva. The development of this system is the result of a long process, and the pipeline of our approach from then to nowadays is presented. Specific epitopes with the greatest antigenic potential using immunoinformatic algorithms have been selected from worldwide and tropical pathogens and then assayed by a successive chain of immunological techniques [PEPSCAN®, enzyme-linked immunosorbent assay (ELISA), and MABA] to evaluate the sensitivity and specificity of those synthetic peptides for their usefulness in diagnosis. Years of work have been required for this complex process, with the recent incorporation of new immunoinformatic predictive tools, methodologies, and cost advantages. It can be concluded that synthetic peptides are a promising approach for diagnostic processes based either on the detection of antigens or antibodies.
Inflammatory bowel disease (IBD), which includes ulcerative colitis and Crohn’s disease, is becoming a major public health concern in Africa, particularly in cities, due to urbanization, dietary changes, and improved diagnostic tools. The present study discusses the benefits, disadvantages, and practical limitations of the pharmacological treatments for IBD patients that are currently accessible in Africa. Given the limits of conventional treatments, such as their potential for considerable side effects, high cost, and often limited accessibility, this review explores emerging new treatment approaches such as nanomedicine, personalized medicine, and the use of traditional African medicines. Highlighting the urgency for potential alternative treatments, this review explores new and developing therapeutic innovations to enhance IBD management and improve the quality of life for African patients.
Inflammatory bowel disease (IBD), which includes ulcerative colitis and Crohn’s disease, is becoming a major public health concern in Africa, particularly in cities, due to urbanization, dietary changes, and improved diagnostic tools. The present study discusses the benefits, disadvantages, and practical limitations of the pharmacological treatments for IBD patients that are currently accessible in Africa. Given the limits of conventional treatments, such as their potential for considerable side effects, high cost, and often limited accessibility, this review explores emerging new treatment approaches such as nanomedicine, personalized medicine, and the use of traditional African medicines. Highlighting the urgency for potential alternative treatments, this review explores new and developing therapeutic innovations to enhance IBD management and improve the quality of life for African patients.
Since our previous summary of the 74 FDA-approved kinase inhibitors in clinical and preclinical trials for non-cancerous neurological treatment, the US FDA has approved 13 additional kinase inhibitors since early 2022. This update incorporates new evidence for the now 87 FDA-approved kinase inhibitors in clinical and preclinical trials for the treatment of non-cancerous neurological disorders. By the end of October 2024, nearly all 87 FDA-approved kinase inhibitors have been tested in various animal models of non-cancerous neurological disorders, with twenty entered into clinical trials and six used for off-label treatments of neurological conditions in humans. Considering the challenges posed by intellectual property (IP), legal considerations, and limited blood-brain barrier (BBB) permeability, which may restrict some FDA-approved kinase inhibitors from effectively targeting the central nervous system (CNS), we further discuss the feasibility of designing novel proprietary analogs with enhanced BBB penetration to improve their therapeutic potential in neurological disorders. The new drugs typically retain full IP rights and remain costly; while repurposing kinase inhibitors may provide effective and affordable treatments for non-cancerous neurological disorders.
Since our previous summary of the 74 FDA-approved kinase inhibitors in clinical and preclinical trials for non-cancerous neurological treatment, the US FDA has approved 13 additional kinase inhibitors since early 2022. This update incorporates new evidence for the now 87 FDA-approved kinase inhibitors in clinical and preclinical trials for the treatment of non-cancerous neurological disorders. By the end of October 2024, nearly all 87 FDA-approved kinase inhibitors have been tested in various animal models of non-cancerous neurological disorders, with twenty entered into clinical trials and six used for off-label treatments of neurological conditions in humans. Considering the challenges posed by intellectual property (IP), legal considerations, and limited blood-brain barrier (BBB) permeability, which may restrict some FDA-approved kinase inhibitors from effectively targeting the central nervous system (CNS), we further discuss the feasibility of designing novel proprietary analogs with enhanced BBB penetration to improve their therapeutic potential in neurological disorders. The new drugs typically retain full IP rights and remain costly; while repurposing kinase inhibitors may provide effective and affordable treatments for non-cancerous neurological disorders.
The objective of this study was to develop a simple quantitative model (SQM) to predict maximum plasma concentration (Cmax) and the area under the curve (AUC) of renally excreted drugs (n = 16) in pregnant women from non-pregnant women.
The SQM was developed using 6 physiological parameters and the fraction unbound protein in plasma (fup) as the product characteristic. The six physiological parameters used in this study were total body water, blood volume, cardiac output, glomerular filtration rate (GFR), volume of the fetoplacental unit and blood flow of the fetoplacental unit. A factor was derived based on the average values of the physiological parameters and fup for different gestational ages to predict Cmax and AUC values in pregnant women from non-pregnant women. The predicted values from SQM were then compared with the dedicated clinical studies as well as predicted values by a physiologically-based pharmacokinetic (PBPK) model.
Out of 17 Cmax data points, 15 (88.2%), 15 (88.2%), and 12 (70.6%) data points were within 0.5–2.0-fold, 0.5–1.5-fold and 0.7–1.30-fold prediction error, respectively, by SQM, whereas, 17 (100%), 15 (88.2%), and 13 (76.5%) data points were within 0.5–2.0-fold, 0.5–1.5-fold and 0.7–1.30 fold prediction error, respectively, by PBPK. Out of 36 AUC data points, 36 (100%), 34 (94.4%), and 30 (83.3%) data points were within 0.5–2.0-fold, 0.5–1.5-fold and 0.7–1.30-fold prediction error, respectively, by SQM, whereas, 35 (97.2%), 33 (91.7%), and 27 (75%) data points were within 0.5–2.0-fold, 0.5–1.5-fold and 0.7–1.30-fold prediction error, respectively, by PBPK. The results of the study indicated that the predictive power of both models was very good.
The results of the study indicate that the SQM in its predictive performance is as robust and accurate as whole body PBPK.
The objective of this study was to develop a simple quantitative model (SQM) to predict maximum plasma concentration (Cmax) and the area under the curve (AUC) of renally excreted drugs (n = 16) in pregnant women from non-pregnant women.
The SQM was developed using 6 physiological parameters and the fraction unbound protein in plasma (fup) as the product characteristic. The six physiological parameters used in this study were total body water, blood volume, cardiac output, glomerular filtration rate (GFR), volume of the fetoplacental unit and blood flow of the fetoplacental unit. A factor was derived based on the average values of the physiological parameters and fup for different gestational ages to predict Cmax and AUC values in pregnant women from non-pregnant women. The predicted values from SQM were then compared with the dedicated clinical studies as well as predicted values by a physiologically-based pharmacokinetic (PBPK) model.
Out of 17 Cmax data points, 15 (88.2%), 15 (88.2%), and 12 (70.6%) data points were within 0.5–2.0-fold, 0.5–1.5-fold and 0.7–1.30-fold prediction error, respectively, by SQM, whereas, 17 (100%), 15 (88.2%), and 13 (76.5%) data points were within 0.5–2.0-fold, 0.5–1.5-fold and 0.7–1.30 fold prediction error, respectively, by PBPK. Out of 36 AUC data points, 36 (100%), 34 (94.4%), and 30 (83.3%) data points were within 0.5–2.0-fold, 0.5–1.5-fold and 0.7–1.30-fold prediction error, respectively, by SQM, whereas, 35 (97.2%), 33 (91.7%), and 27 (75%) data points were within 0.5–2.0-fold, 0.5–1.5-fold and 0.7–1.30-fold prediction error, respectively, by PBPK. The results of the study indicated that the predictive power of both models was very good.
The results of the study indicate that the SQM in its predictive performance is as robust and accurate as whole body PBPK.
Merozoite invasion of erythrocytes relies on molecular interactions between parasite and host proteins, making these proteins potential therapeutic targets. This review summarizes research on Plasmodium falciparum merozoite invasion conducted at the Instituto de Inmunología, San Juan de Dios Hospital, between 1990 and 2000. Erythrocyte-binding analyses of P. falciparum proteins merozoite surface protein 1 (MSP-1), MSP-2, acid basic repeat antigen (ABRA) (MSP-9), apical membrane antigen-1 (AMA-1), rhoptry-associated protein 1 (RAP-1), glycophorin-binding protein 130 (GBP-130), serine repeat antigen 5 (SERA-5), erythrocyte-binding antigen 140 (EBA-140), and EBA-175 identified 50 high-activity binding peptides (HABPs) with nanomolar-range dissociation constants. Most of these peptides inhibit merozoite invasion and belong to erythrocyte-binding regions of their respective proteins. Several HABPs overlap with epitopes recognized by inhibitory monoclonal antibodies (mAbs). MSP-1 HABP-5501 contains epitopes for mAbs 12.8, 12.10, MaliM03 fragment antigen binding (Fab), and 42D6 Fab, all of which block invasion. MSP-2 HABPs include epitopes targeted by opsonizing antibodies, while MSP-9 HABPs (2148–2153) interact with Band 3 during invasion. AMA-1 HABPs (4315, 4316, and 4325) contribute to the 1F9 epitope, a key target of immune recognition. RAP-1 HABP-26188 elicits inhibitory antibodies, including cross-reactive mAbs SP5.2 and SP8.18, the latter displaying parasite growth inhibition. GBP-130 HABP-2220 binds glycophorin and induces invasion-blocking antibodies. SERA-5 HABP-6725 contains sequences targeted by native SERA-5 antibodies, while HABPs 6727 and 6733 are recognized by antibodies from natural infection and vaccination. EBA-140 HABPs (26135, 26144, and 26147) are located in Region II (RII) and are recognized by mAbs with moderate to strong neutralizing activity. EBA-175 HABPs (1779, 1783, 1814, 1815, and 1818) are in recombinant fragments recognized by antibodies eluted from immune complexes. HABPs 1779 and 1783, located in RII, bind erythrocytes independently of sialic acid, inhibit rRII-EBA binding, and interact with α(2,3)-sialyllactose. HABP-1783 also contains the target site of mAb R217, which potently blocks EBA-175 binding to glycophorin A and inhibits invasion.
Merozoite invasion of erythrocytes relies on molecular interactions between parasite and host proteins, making these proteins potential therapeutic targets. This review summarizes research on Plasmodium falciparum merozoite invasion conducted at the Instituto de Inmunología, San Juan de Dios Hospital, between 1990 and 2000. Erythrocyte-binding analyses of P. falciparum proteins merozoite surface protein 1 (MSP-1), MSP-2, acid basic repeat antigen (ABRA) (MSP-9), apical membrane antigen-1 (AMA-1), rhoptry-associated protein 1 (RAP-1), glycophorin-binding protein 130 (GBP-130), serine repeat antigen 5 (SERA-5), erythrocyte-binding antigen 140 (EBA-140), and EBA-175 identified 50 high-activity binding peptides (HABPs) with nanomolar-range dissociation constants. Most of these peptides inhibit merozoite invasion and belong to erythrocyte-binding regions of their respective proteins. Several HABPs overlap with epitopes recognized by inhibitory monoclonal antibodies (mAbs). MSP-1 HABP-5501 contains epitopes for mAbs 12.8, 12.10, MaliM03 fragment antigen binding (Fab), and 42D6 Fab, all of which block invasion. MSP-2 HABPs include epitopes targeted by opsonizing antibodies, while MSP-9 HABPs (2148–2153) interact with Band 3 during invasion. AMA-1 HABPs (4315, 4316, and 4325) contribute to the 1F9 epitope, a key target of immune recognition. RAP-1 HABP-26188 elicits inhibitory antibodies, including cross-reactive mAbs SP5.2 and SP8.18, the latter displaying parasite growth inhibition. GBP-130 HABP-2220 binds glycophorin and induces invasion-blocking antibodies. SERA-5 HABP-6725 contains sequences targeted by native SERA-5 antibodies, while HABPs 6727 and 6733 are recognized by antibodies from natural infection and vaccination. EBA-140 HABPs (26135, 26144, and 26147) are located in Region II (RII) and are recognized by mAbs with moderate to strong neutralizing activity. EBA-175 HABPs (1779, 1783, 1814, 1815, and 1818) are in recombinant fragments recognized by antibodies eluted from immune complexes. HABPs 1779 and 1783, located in RII, bind erythrocytes independently of sialic acid, inhibit rRII-EBA binding, and interact with α(2,3)-sialyllactose. HABP-1783 also contains the target site of mAb R217, which potently blocks EBA-175 binding to glycophorin A and inhibits invasion.
The immunosuppressive drug brequinar (BQR) is a potent inhibitor of dihydroorotate dehydrogenase (DHODH) active against autoimmune diseases and viral infections. This oral drug is currently evaluated for the treatment of cancers, notably acute myeloid leukemia to limit the suppressive function of myeloid cells. A combination of BQR and an anti-PD-1 (programmed death-1) antibody has revealed potent antitumor and antimetastatic activities. BQR induced a marked down-regulation of PD-L1 (programmed death-ligand 1) gene expression and a large decrease of PD-L1 protein expression in implanted tumors in mice.
The present study evaluated the capacity of BQR to interact directly with the PD-L1 protein dimer using molecular modeling.
Molecular docking experiments revealed a modest capacity of BQR to stabilize PD-L1 dimers. The PD-L1 binding capacities of four known BQR analogs were compared to establish structure-binding relationships. The protein binding was significantly enhanced when the acid function of BQR was replaced with a trifluoroethanol substituent. The interaction was further reinforced when BQR was coupled to a mitochondria-targeted triphenylphosphine (TPP) unit. Among three BQR-TPP hybrids, compound B2 with a short alkyl linker revealed a prominent capacity to interact with PD-L1, superior to that of the reference biphenyl ligand BMS-202.
Two PD-L1 binders derived from BQR have been identified and the protein interaction modeled. Our study underlines the possibility of designing novel small molecule ligands targeted to the PD-L1 dimer interface based on the BQR scaffold.
The immunosuppressive drug brequinar (BQR) is a potent inhibitor of dihydroorotate dehydrogenase (DHODH) active against autoimmune diseases and viral infections. This oral drug is currently evaluated for the treatment of cancers, notably acute myeloid leukemia to limit the suppressive function of myeloid cells. A combination of BQR and an anti-PD-1 (programmed death-1) antibody has revealed potent antitumor and antimetastatic activities. BQR induced a marked down-regulation of PD-L1 (programmed death-ligand 1) gene expression and a large decrease of PD-L1 protein expression in implanted tumors in mice.
The present study evaluated the capacity of BQR to interact directly with the PD-L1 protein dimer using molecular modeling.
Molecular docking experiments revealed a modest capacity of BQR to stabilize PD-L1 dimers. The PD-L1 binding capacities of four known BQR analogs were compared to establish structure-binding relationships. The protein binding was significantly enhanced when the acid function of BQR was replaced with a trifluoroethanol substituent. The interaction was further reinforced when BQR was coupled to a mitochondria-targeted triphenylphosphine (TPP) unit. Among three BQR-TPP hybrids, compound B2 with a short alkyl linker revealed a prominent capacity to interact with PD-L1, superior to that of the reference biphenyl ligand BMS-202.
Two PD-L1 binders derived from BQR have been identified and the protein interaction modeled. Our study underlines the possibility of designing novel small molecule ligands targeted to the PD-L1 dimer interface based on the BQR scaffold.
Pentafuranosylnucleos(t)ides represent a class of natural compounds regulating diverse cell functions being preferably components of biopolymers and also participating as cyclic regulatory low-molecular ligands. Disaccharide nucleosides and related analogs are considered as therapeutically potent compounds for the treatment of cancer, viral diseases, and a variety of metabolic disorders by mimicking a structure of biochemically occurring molecules participating in nicotinamide adenine dinucleotide (NAD+) transformation. Several approaches have been developed on the way to the chemical synthesis of poly(adenosine diphosphate ribose) (PAR), a unique biopolymer taking part in DNA repair and associated functions, that would allow extensive studies of molecular mechanisms of a variety of diseases. The present review consists of the following main parts, the first one including structural characterization, biochemical roles, and chemical synthesis of disaccharide nucleosides from different sources and biopolymers on their basis, the second one describing therapeutic applications of disaccharide nucleosides and their analogs. General conclusion and perspectives are summarized in the last part.
Pentafuranosylnucleos(t)ides represent a class of natural compounds regulating diverse cell functions being preferably components of biopolymers and also participating as cyclic regulatory low-molecular ligands. Disaccharide nucleosides and related analogs are considered as therapeutically potent compounds for the treatment of cancer, viral diseases, and a variety of metabolic disorders by mimicking a structure of biochemically occurring molecules participating in nicotinamide adenine dinucleotide (NAD+) transformation. Several approaches have been developed on the way to the chemical synthesis of poly(adenosine diphosphate ribose) (PAR), a unique biopolymer taking part in DNA repair and associated functions, that would allow extensive studies of molecular mechanisms of a variety of diseases. The present review consists of the following main parts, the first one including structural characterization, biochemical roles, and chemical synthesis of disaccharide nucleosides from different sources and biopolymers on their basis, the second one describing therapeutic applications of disaccharide nucleosides and their analogs. General conclusion and perspectives are summarized in the last part.
This study aimed to elucidate the structural basis for the interaction of five natural anti-arthritic compounds, diacerein, rhein, glucosamines [glucosamine 3-sulfate (G3S), and glucosamine 6-sulfate (G6S)], and chondroitin disaccharide Δdi-4S (C4S) with cytochrome P450 2C9 (CYP2C9).
The investigated compounds were docked individually to the defined binding site in CYP2C9 based on the published crystal structure (PDB code: 1R9O).
All investigated ligands bound deep in the active site pocket in close proximity to the heme. Except for chondroitin, all ligands are bonded to residues found in critical secondary structures that form the boundary of the active site cavity, including B-C loop, F helix, F-G loop, and I helix. A total of 12 amino acids were involved in the binding, and all were critical residues located in four out of six substrate recognition sites (SRSs) that have been identified as important substrate binding and catalysis regions in other CYP isoforms. The relatively more potent binding (lower CDOCKER interaction energy) observed for diacerein and rhein compared to glucosamines and C4S are likely due to two main factors: a higher number of bonds between the ligand molecule and CYP2C9 active site residues (14 versus 0–4), and direct interaction with the heme moiety. The binding residues identified in both diacerein and rhein were the residues that also bonded with sulfaphenazole, the specific and potent CYP2C9 inhibitor.
Collectively, this study has provided insights into structural features of CYP2C9 critical for inhibition and formed a basis for further exploration of structural determinants for potency and specificity of therapeutic compounds as CYP2C9 inhibitors.
This study aimed to elucidate the structural basis for the interaction of five natural anti-arthritic compounds, diacerein, rhein, glucosamines [glucosamine 3-sulfate (G3S), and glucosamine 6-sulfate (G6S)], and chondroitin disaccharide Δdi-4S (C4S) with cytochrome P450 2C9 (CYP2C9).
The investigated compounds were docked individually to the defined binding site in CYP2C9 based on the published crystal structure (PDB code: 1R9O).
All investigated ligands bound deep in the active site pocket in close proximity to the heme. Except for chondroitin, all ligands are bonded to residues found in critical secondary structures that form the boundary of the active site cavity, including B-C loop, F helix, F-G loop, and I helix. A total of 12 amino acids were involved in the binding, and all were critical residues located in four out of six substrate recognition sites (SRSs) that have been identified as important substrate binding and catalysis regions in other CYP isoforms. The relatively more potent binding (lower CDOCKER interaction energy) observed for diacerein and rhein compared to glucosamines and C4S are likely due to two main factors: a higher number of bonds between the ligand molecule and CYP2C9 active site residues (14 versus 0–4), and direct interaction with the heme moiety. The binding residues identified in both diacerein and rhein were the residues that also bonded with sulfaphenazole, the specific and potent CYP2C9 inhibitor.
Collectively, this study has provided insights into structural features of CYP2C9 critical for inhibition and formed a basis for further exploration of structural determinants for potency and specificity of therapeutic compounds as CYP2C9 inhibitors.
Microbial pathogens with antibiotic resistance have become challenging to manage in the last few decades. The situation is alarming and a threat to humankind. Despite emerging new drug candidates, there are numerous strategies for efficient antibiotic delivery for treating such infections; antibiotic-resistant bacteria are still not eradicated adequately from the infected hosts. Recently, antimicrobial peptides (AMPs) have emerged as saviors in overcoming antibiotic resistance against bacteria. AMPs are being produced naturally as well as synthetically. Irrespective of the source of production, AMPs have shown higher specificity and lower toxicity to the host. Such functions have been attributed to distinct structures, functions, and varied mechanisms of action. This review highlights sources, structural and physiological characteristics, action mechanisms, and biological potency towards clinical applications. Most recently, AMPs have also been explored in treating cancers and tumors. Despite the entry of a few AMPs into clinical trials, there are limitations associated with their usage, like shorter half-life, protease cleavage, and toxicity. There is an urgent need to produce AMPs with intensified activity, biocompatibility, and lesser toxicity. This review sheds light on these aspects and the future of AMPs for the betterment of the human race.
Microbial pathogens with antibiotic resistance have become challenging to manage in the last few decades. The situation is alarming and a threat to humankind. Despite emerging new drug candidates, there are numerous strategies for efficient antibiotic delivery for treating such infections; antibiotic-resistant bacteria are still not eradicated adequately from the infected hosts. Recently, antimicrobial peptides (AMPs) have emerged as saviors in overcoming antibiotic resistance against bacteria. AMPs are being produced naturally as well as synthetically. Irrespective of the source of production, AMPs have shown higher specificity and lower toxicity to the host. Such functions have been attributed to distinct structures, functions, and varied mechanisms of action. This review highlights sources, structural and physiological characteristics, action mechanisms, and biological potency towards clinical applications. Most recently, AMPs have also been explored in treating cancers and tumors. Despite the entry of a few AMPs into clinical trials, there are limitations associated with their usage, like shorter half-life, protease cleavage, and toxicity. There is an urgent need to produce AMPs with intensified activity, biocompatibility, and lesser toxicity. This review sheds light on these aspects and the future of AMPs for the betterment of the human race.
This paper investigates two possible treatment targets for neuroblastoma (NB) stage 4 (NBS4), c-Src kinase (Csk) and retinoic acid (RA) signalling pathways as potential candidates for a multi-target drug. Research has demonstrated that many cancer cells overexpress and/or hyperactivate c-Src, a tyrosine that is a member of the Src-family kinases. In the case of NBS4, there are indications that successful inhibition of c-Src could inhibit disease progression. Research into the altered signalling of RA, which preserves the differentiated state of adult neurons, neural stem cells, and NB cells (SH-SY5Y), is also investigated as a potential multi-target drug.
Using computer-aided technology, including OpenEye Scientific suite, Molegro Virtual docking, Samson suite, and Discovery Studio Visualiser, the results revealed that the receptors for both targets, Csk and RA, share similar amino acid sequencing that ranges from 80–100%, offering the possibility of further testing for multi-target drug use. Work was done to explore possible synthesis routes for each of the four compounds using the retrosynthesis program Spaya. Predictive toxicology was done using the Toxicity Estimation Software Tool (T.E.S.T.).
Four compounds (inhibitors) targeting the Csk tyrosine kinase and RA pathways were identified as potential inhibitors.
Currently, no effective therapeutic agents for NBS4 exist. Immunotherapy which has proven effective in treating various cancers, is currently used to treat NBS4 and has a 40% to 50% survival rate. This paper investigates two possible treatment targets for NBS4, Csk and RA signalling pathways as possible candidates for a multi-target drug. Four potential inhibitors have been identified.
This paper investigates two possible treatment targets for neuroblastoma (NB) stage 4 (NBS4), c-Src kinase (Csk) and retinoic acid (RA) signalling pathways as potential candidates for a multi-target drug. Research has demonstrated that many cancer cells overexpress and/or hyperactivate c-Src, a tyrosine that is a member of the Src-family kinases. In the case of NBS4, there are indications that successful inhibition of c-Src could inhibit disease progression. Research into the altered signalling of RA, which preserves the differentiated state of adult neurons, neural stem cells, and NB cells (SH-SY5Y), is also investigated as a potential multi-target drug.
Using computer-aided technology, including OpenEye Scientific suite, Molegro Virtual docking, Samson suite, and Discovery Studio Visualiser, the results revealed that the receptors for both targets, Csk and RA, share similar amino acid sequencing that ranges from 80–100%, offering the possibility of further testing for multi-target drug use. Work was done to explore possible synthesis routes for each of the four compounds using the retrosynthesis program Spaya. Predictive toxicology was done using the Toxicity Estimation Software Tool (T.E.S.T.).
Four compounds (inhibitors) targeting the Csk tyrosine kinase and RA pathways were identified as potential inhibitors.
Currently, no effective therapeutic agents for NBS4 exist. Immunotherapy which has proven effective in treating various cancers, is currently used to treat NBS4 and has a 40% to 50% survival rate. This paper investigates two possible treatment targets for NBS4, Csk and RA signalling pathways as possible candidates for a multi-target drug. Four potential inhibitors have been identified.
The PI3K (phosphoinositide 3-kinase)-alpha isoform is found upregulated in 30–40% of breast cancer. Currently, there are limited selective and specific drugs that target PI3K-alpha, and no natural therapeutic option is available. This study aims to develop natural hybrid antagonists of PI3K-alpha for breast cancer therapeutics.
25 pan-PI3K and PI3K-alpha targeting drugs were obtained from various sources, including the COCONUT (Collection of Open Natural Products) database. On the parent dataset, high throughput virtual screening (HTVS), standard precision (SP) docking, and extra precision (XP) docking were performed to produce Murcko scaffolds and heterogenous fragments. Murcko scaffolds are hybridized with fragments of natural compounds (Category 1) and drugs (Category 2), respectively. Hybrids are docked with HTVS, SP, and XP, followed by induced fit docking and ADME (absorption, distribution, metabolism, and excretion) prediction. MM/GBSA (molecular mechanics/generalized Born and surface area) was performed on the docked poses.
Highest docking scores of –13.354 kcal/mol and –12.670 kcal/mol were achieved by hybrids in Category 1 and Category 2, respectively. MM/GBSA free energy ranged from –51.14 kcal/mol to –72.66 kcal/mol. In terms of binding docking, pharmacological properties, and Lipinski’s rule of five, the natural hybrids outperformed the parent drugs.
PI3K-alpha kinase proteins can be targeted with natural-drug hybrid antagonists for breast cancer treatment. Hybrid molecules, such as NH-01 and NH-06, show better binding with promising ADME properties. Thus, in vivo and in vitro testing is necessary to prove the value of such hybrids.
The PI3K (phosphoinositide 3-kinase)-alpha isoform is found upregulated in 30–40% of breast cancer. Currently, there are limited selective and specific drugs that target PI3K-alpha, and no natural therapeutic option is available. This study aims to develop natural hybrid antagonists of PI3K-alpha for breast cancer therapeutics.
25 pan-PI3K and PI3K-alpha targeting drugs were obtained from various sources, including the COCONUT (Collection of Open Natural Products) database. On the parent dataset, high throughput virtual screening (HTVS), standard precision (SP) docking, and extra precision (XP) docking were performed to produce Murcko scaffolds and heterogenous fragments. Murcko scaffolds are hybridized with fragments of natural compounds (Category 1) and drugs (Category 2), respectively. Hybrids are docked with HTVS, SP, and XP, followed by induced fit docking and ADME (absorption, distribution, metabolism, and excretion) prediction. MM/GBSA (molecular mechanics/generalized Born and surface area) was performed on the docked poses.
Highest docking scores of –13.354 kcal/mol and –12.670 kcal/mol were achieved by hybrids in Category 1 and Category 2, respectively. MM/GBSA free energy ranged from –51.14 kcal/mol to –72.66 kcal/mol. In terms of binding docking, pharmacological properties, and Lipinski’s rule of five, the natural hybrids outperformed the parent drugs.
PI3K-alpha kinase proteins can be targeted with natural-drug hybrid antagonists for breast cancer treatment. Hybrid molecules, such as NH-01 and NH-06, show better binding with promising ADME properties. Thus, in vivo and in vitro testing is necessary to prove the value of such hybrids.
The development of selective and potent antitumor agents remains a significant challenge. This study aimed to synthesize and evaluate biaryl hydroxy-1,2,3-triazoles and 9H-fluorene-1,2,3-triazole hybrids, inspired by previously identified bioactive 1,2,3-triazoles, for their cytotoxic potential against human cancer cell lines.
A library of 13 biaryl hydroxy-1,2,3-triazoles and 11 fluorene-1,2,3-triazoles was synthesized using optimized Suzuki and telescopic one-pot reactions, with yields ranging from 16% to 97%. The cytotoxicity of these compounds was tested against HCT-116 (colorectal cancer), SNB-19 (astrocytoma), MDA-MB-231 (triple-negative breast cancer), and MOLM-13 (acute myeloid leukemia, FLT3-ITD mutant) cell lines.
Two fluorene-triazoles, 1-(2-bromophenyl)-4-(9H-fluoren-9-yl)-1H-1,2,3-triazole (
The results highlight the critical role of bromine substitution on the aryl azide-derived ring in modulating cytotoxic activity. The study reinforces the potential of rigid fluorene-based scaffolds as promising leads for the development of targeted therapies against FLT3-mutant leukemia, aligning with previous reports on 1,2,3-triazole hybrids antiproliferative activity in leukemia models.
The development of selective and potent antitumor agents remains a significant challenge. This study aimed to synthesize and evaluate biaryl hydroxy-1,2,3-triazoles and 9H-fluorene-1,2,3-triazole hybrids, inspired by previously identified bioactive 1,2,3-triazoles, for their cytotoxic potential against human cancer cell lines.
A library of 13 biaryl hydroxy-1,2,3-triazoles and 11 fluorene-1,2,3-triazoles was synthesized using optimized Suzuki and telescopic one-pot reactions, with yields ranging from 16% to 97%. The cytotoxicity of these compounds was tested against HCT-116 (colorectal cancer), SNB-19 (astrocytoma), MDA-MB-231 (triple-negative breast cancer), and MOLM-13 (acute myeloid leukemia, FLT3-ITD mutant) cell lines.
Two fluorene-triazoles, 1-(2-bromophenyl)-4-(9H-fluoren-9-yl)-1H-1,2,3-triazole (
The results highlight the critical role of bromine substitution on the aryl azide-derived ring in modulating cytotoxic activity. The study reinforces the potential of rigid fluorene-based scaffolds as promising leads for the development of targeted therapies against FLT3-mutant leukemia, aligning with previous reports on 1,2,3-triazole hybrids antiproliferative activity in leukemia models.
Cancer remains the second leading cause of death globally, posing an ongoing threat to public health. A hallmark of cancer cells is their capacity to invade adjacent tissues and evolve into malignant forms, often resulting in aggressive tumors resistant to conventional treatments. At the heart of this therapeutic challenge are cancer stem cells (CSCs), which possess distinctive capabilities for self-renewal, differentiation, and generation of diverse tumor cell populations. These CSCs have been identified across multiple tissue types, including lung, colon, breast, pancreas, and ovary. Research has demonstrated that CSC subpopulations contribute significantly to therapeutic resistance, tumor recurrence, and metastasis by regulating multiple signaling pathways, making them compelling targets for cancer therapy. Notably, emerging evidence suggests that natural products may offer protective benefits against cancer development while potentially targeting CSCs. This review synthesizes current knowledge of CSCs, examining their identifying markers, isolation techniques, study methods, and associated signaling pathways. Additionally, we explore various natural products that specifically target CSCs across different cancer types, presenting potential strategies to address the persistent challenges of drug resistance and cancer relapse.
Cancer remains the second leading cause of death globally, posing an ongoing threat to public health. A hallmark of cancer cells is their capacity to invade adjacent tissues and evolve into malignant forms, often resulting in aggressive tumors resistant to conventional treatments. At the heart of this therapeutic challenge are cancer stem cells (CSCs), which possess distinctive capabilities for self-renewal, differentiation, and generation of diverse tumor cell populations. These CSCs have been identified across multiple tissue types, including lung, colon, breast, pancreas, and ovary. Research has demonstrated that CSC subpopulations contribute significantly to therapeutic resistance, tumor recurrence, and metastasis by regulating multiple signaling pathways, making them compelling targets for cancer therapy. Notably, emerging evidence suggests that natural products may offer protective benefits against cancer development while potentially targeting CSCs. This review synthesizes current knowledge of CSCs, examining their identifying markers, isolation techniques, study methods, and associated signaling pathways. Additionally, we explore various natural products that specifically target CSCs across different cancer types, presenting potential strategies to address the persistent challenges of drug resistance and cancer relapse.
Metabolic syndrome is a complex, multifactorial disorder, with emerging research emphasizing the significant role of gut health in its prevention and management. Recent studies suggest that dietary strategies promoting a healthy gut microbiome, including the incorporation of fiber, fermented foods, and healthy fats, are crucial for regulating metabolism. Additionally, the use of postbiotics and supplements, such as probiotics, omega-3 fatty acids, and polyphenols, provides promising avenues for enhancing metabolic health. This holistic approach to managing metabolic syndrome not only supports gut health but also offers the potential for improving long-term health outcomes. This review examines the influence of the gut microbiome on metabolism, highlighting the increasing significance of dietary strategies and supplements in managing metabolic syndrome.
Metabolic syndrome is a complex, multifactorial disorder, with emerging research emphasizing the significant role of gut health in its prevention and management. Recent studies suggest that dietary strategies promoting a healthy gut microbiome, including the incorporation of fiber, fermented foods, and healthy fats, are crucial for regulating metabolism. Additionally, the use of postbiotics and supplements, such as probiotics, omega-3 fatty acids, and polyphenols, provides promising avenues for enhancing metabolic health. This holistic approach to managing metabolic syndrome not only supports gut health but also offers the potential for improving long-term health outcomes. This review examines the influence of the gut microbiome on metabolism, highlighting the increasing significance of dietary strategies and supplements in managing metabolic syndrome.
This study aimed to computationally identify and optimize 4-hydroxy isoleucine (4HILe) derivatives from fenugreek as multitarget antidiabetic agents against α-glucosidase, α-amylase, and aldose reductase [PDB (protein data bank) IDs: 5NN8, 4GQR, 4QX4].
A multi-step computational workflow was employed to identify and optimize 4HILe derivatives as antidiabetic agents. Molecular docking using the Schrödinger Suite screened 23 ligands against three enzyme targets to evaluate binding affinities and interactions. Molecular dynamic (MD) simulations conducted with GROMACS (Groningen machine chemical simulations) over 100 ns assessed conformational stability through RMSD (root mean square deviation) and RMSF (root mean square fluctuation) analysis. Binding free energy calculations [MM-GBSA (molecular mechanics-generalized Born surface area)] and free energy landscape (FEL) studies are performed to validate the thermodynamics of protein-ligand interactions. Additionally, generative AI modeling using LigDream generated 100 novel compounds derived from 4HILe, subsequently validated through docking studies to identify promising inhibitors.
The study identified 4HILe-4, 2R-3S-4R-4HILe, and 4HILe-Amide-2 as potent derivatives with superior binding affinities [ΔG (Gibbs free energy): −49.3 to −42.3 kcal/mol] compared to co-crystal ligands (−45.3 kcal/mol), as determined by docking and MM-GBSA calculations. MD revealed stable protein-ligand complexes, evidenced by low RMSD values (0.2–0.4 nm) and minimal residue fluctuations (RMSF), confirming their structural integrity. The generative AI approach using LigDream also generated 100 novel 4HILe derivatives, with top candidates exhibiting strong docking scores and key molecular interactions against α-glucosidase, α-amylase, and aldose reductase. Notably, compound 10 (−9.424 kcal/mol), compound 4 (−8.167 kcal/mol), and compound 28 (−13.760 kcal/mol) emerged as promising inhibitors for further investigation.
The study highlights 4HILe derivatives as promising inhibitors for diabetes-associated enzymes, demonstrating robust binding and dynamic stability. Integrating molecular dynamics, free energy calculations, and AI-driven generative modeling provides a strong framework for accelerating antidiabetic drug discovery. These findings pave the way for experimental validation and the development of next-generation therapeutics targeting insulin resistance and hyperglycemia.
This study aimed to computationally identify and optimize 4-hydroxy isoleucine (4HILe) derivatives from fenugreek as multitarget antidiabetic agents against α-glucosidase, α-amylase, and aldose reductase [PDB (protein data bank) IDs: 5NN8, 4GQR, 4QX4].
A multi-step computational workflow was employed to identify and optimize 4HILe derivatives as antidiabetic agents. Molecular docking using the Schrödinger Suite screened 23 ligands against three enzyme targets to evaluate binding affinities and interactions. Molecular dynamic (MD) simulations conducted with GROMACS (Groningen machine chemical simulations) over 100 ns assessed conformational stability through RMSD (root mean square deviation) and RMSF (root mean square fluctuation) analysis. Binding free energy calculations [MM-GBSA (molecular mechanics-generalized Born surface area)] and free energy landscape (FEL) studies are performed to validate the thermodynamics of protein-ligand interactions. Additionally, generative AI modeling using LigDream generated 100 novel compounds derived from 4HILe, subsequently validated through docking studies to identify promising inhibitors.
The study identified 4HILe-4, 2R-3S-4R-4HILe, and 4HILe-Amide-2 as potent derivatives with superior binding affinities [ΔG (Gibbs free energy): −49.3 to −42.3 kcal/mol] compared to co-crystal ligands (−45.3 kcal/mol), as determined by docking and MM-GBSA calculations. MD revealed stable protein-ligand complexes, evidenced by low RMSD values (0.2–0.4 nm) and minimal residue fluctuations (RMSF), confirming their structural integrity. The generative AI approach using LigDream also generated 100 novel 4HILe derivatives, with top candidates exhibiting strong docking scores and key molecular interactions against α-glucosidase, α-amylase, and aldose reductase. Notably, compound 10 (−9.424 kcal/mol), compound 4 (−8.167 kcal/mol), and compound 28 (−13.760 kcal/mol) emerged as promising inhibitors for further investigation.
The study highlights 4HILe derivatives as promising inhibitors for diabetes-associated enzymes, demonstrating robust binding and dynamic stability. Integrating molecular dynamics, free energy calculations, and AI-driven generative modeling provides a strong framework for accelerating antidiabetic drug discovery. These findings pave the way for experimental validation and the development of next-generation therapeutics targeting insulin resistance and hyperglycemia.