Pub Date : 2025-04-09DOI: 10.1007/s43440-025-00721-w
Song-Hee Lee, Haeri Lee, Yong-Jin Kwon, Seul-Ki Kim, Eun-Bi Seo, Jie Ohn Sohn, Byung-Hak Kim, Jung-Youl Park, Sang-Kyu Ye
Background: Breast cancer remains the leading cause of cancer incidence and mortality among women worldwide, with triple-negative breast cancer (TNBC) posing significant treatment challenges. The dysregulation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway contributes to tumor progression, making it a potential therapeutic target. Chalcones, known for their diverse biological activities, including anti-cancer effects, hold promise for drug development. This study explores the anti-cancer activity of (E)-4-(3-(2-(benzyloxy)-6-hydroxyphenyl)-3-oxoprop-1-en-1-yl)benzoic acid (chalcone-9), a novel chalcone derivative.
Methods: The cytotoxic effects of chalcone-9 were evaluated in breast cancer cell lines, including TNBC lines MDA-MB-231 and MDA-MB-468. Western blotting and qRT-PCR were used to analyze the impact on JAK1, JAK2, STAT1, and STAT3 activation and their downstream gene expression. In silico molecular docking analysis was conducted to determine whether chalcone-9 can interact with JAK1 and JAK2. A wound healing assay was used to observe the effect of chalcone-9 on tumor cell migration, and flow cytometry was employed to analyze whether chalcone-9 inhibits tumor cell cycle progression and induces apoptosis. The expression of apoptosis markers was also assessed.
Results: Chalcone-9 exhibited dose-dependent cytotoxicity in breast cancer cell lines, with TNBC cells showing higher sensitivity. Chalcone-9 effectively inhibited the activation of JAK1, JAK2, STAT1, and STAT3, outperforming conventional JAK/STAT inhibitors. The structure of chalcone-9 was confirmed to stably interact with JAK1 and JAK2 proteins. It also suppressed STAT1 and STAT3 target gene expression, reduced tumor cell migration, and induced apoptosis, as evidenced by PARP and caspase cleavage and decreased survivin levels.
Conclusions: Chalcone-9 demonstrates significant anti-cancer activity, particularly against TNBC. By targeting the JAK/STAT pathway and promoting apoptosis, chalcone-9 emerges as a promising therapeutic candidate for aggressive breast cancers.
{"title":"Chalcone-9: a novel inhibitor of the JAK-STAT pathway with potent anti-cancer effects in triple-negative breast cancer cells.","authors":"Song-Hee Lee, Haeri Lee, Yong-Jin Kwon, Seul-Ki Kim, Eun-Bi Seo, Jie Ohn Sohn, Byung-Hak Kim, Jung-Youl Park, Sang-Kyu Ye","doi":"10.1007/s43440-025-00721-w","DOIUrl":"https://doi.org/10.1007/s43440-025-00721-w","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer remains the leading cause of cancer incidence and mortality among women worldwide, with triple-negative breast cancer (TNBC) posing significant treatment challenges. The dysregulation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway contributes to tumor progression, making it a potential therapeutic target. Chalcones, known for their diverse biological activities, including anti-cancer effects, hold promise for drug development. This study explores the anti-cancer activity of (E)-4-(3-(2-(benzyloxy)-6-hydroxyphenyl)-3-oxoprop-1-en-1-yl)benzoic acid (chalcone-9), a novel chalcone derivative.</p><p><strong>Methods: </strong>The cytotoxic effects of chalcone-9 were evaluated in breast cancer cell lines, including TNBC lines MDA-MB-231 and MDA-MB-468. Western blotting and qRT-PCR were used to analyze the impact on JAK1, JAK2, STAT1, and STAT3 activation and their downstream gene expression. In silico molecular docking analysis was conducted to determine whether chalcone-9 can interact with JAK1 and JAK2. A wound healing assay was used to observe the effect of chalcone-9 on tumor cell migration, and flow cytometry was employed to analyze whether chalcone-9 inhibits tumor cell cycle progression and induces apoptosis. The expression of apoptosis markers was also assessed.</p><p><strong>Results: </strong>Chalcone-9 exhibited dose-dependent cytotoxicity in breast cancer cell lines, with TNBC cells showing higher sensitivity. Chalcone-9 effectively inhibited the activation of JAK1, JAK2, STAT1, and STAT3, outperforming conventional JAK/STAT inhibitors. The structure of chalcone-9 was confirmed to stably interact with JAK1 and JAK2 proteins. It also suppressed STAT1 and STAT3 target gene expression, reduced tumor cell migration, and induced apoptosis, as evidenced by PARP and caspase cleavage and decreased survivin levels.</p><p><strong>Conclusions: </strong>Chalcone-9 demonstrates significant anti-cancer activity, particularly against TNBC. By targeting the JAK/STAT pathway and promoting apoptosis, chalcone-9 emerges as a promising therapeutic candidate for aggressive breast cancers.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143812120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The treatment landscape for Multiple Sclerosis (MS) has increased significantly over the past few decades, thanks to the introduction of disease-modifying therapies (DMTs). Fingolimod, siponimod, ozanimod, and ponesimod belong to the newer generation of oral DMTs categorized as sphingosine 1-phosphate receptor modulators (S1PRMs). Because of their mechanism of action, they may increase the risk of lymphopenia, which could influence the therapeutic management of people with MS. The aim of this study was to describe and compare the reporting frequency of lymphopenia related to four S1PRMs.
Methods: Individual case safety reports (ICSRs) were retrieved from the European spontaneous reporting system database (EudraVigilance) from January 1st, 2022, to December 31st, 2023. The reporting odds ratios (RORs) were computed to compare the reporting probability of lymphopenia between a S1PRM versus each other.
Results: We retrieved 4017 ICSRs, of which 521 (13%) reported lymphopenia associated with fingolimod (53.3%), siponimod (38.4%), ozanimod (5.4%), and ponesimod (2.1%). The most common reporting source was the healthcare professional (94.2%), and more than half of the ICSRs (62.6%) reported serious lymphopenia. Fingolimod was associated with a lower reporting frequency of lymphopenia compared to siponimod. Both siponimod and fingolimod were associated with a higher reporting frequency of lymphopenia compared to ozanimod; siponimod also had a higher reporting probability in comparison with ponesimod.
Conclusions: The most relevant clinical implication of the disproportionality analysis is to increase the awareness of the risk of lymphopenia related to these drugs, thus supporting proactive monitoring and optimizing treatment strategies for people with MS.
Clinical trial number: Not applicable.
{"title":"Lymphopenia associated with sphingosine 1-phosphate receptor modulators (S1PRMs) in multiple sclerosis: analysis of European pharmacovigilance data.","authors":"Nunzia Balzano, Raffaella Di Napoli, Federica Fraenza, Daniele Di Giulio Cesare, Ornella Moreggia, Mirko Cardillo, Cristina Scavone, Giorgia Teresa Maniscalco, Annalisa Capuano, Liberata Sportiello","doi":"10.1007/s43440-025-00725-6","DOIUrl":"https://doi.org/10.1007/s43440-025-00725-6","url":null,"abstract":"<p><strong>Background: </strong>The treatment landscape for Multiple Sclerosis (MS) has increased significantly over the past few decades, thanks to the introduction of disease-modifying therapies (DMTs). Fingolimod, siponimod, ozanimod, and ponesimod belong to the newer generation of oral DMTs categorized as sphingosine 1-phosphate receptor modulators (S1PRMs). Because of their mechanism of action, they may increase the risk of lymphopenia, which could influence the therapeutic management of people with MS. The aim of this study was to describe and compare the reporting frequency of lymphopenia related to four S1PRMs.</p><p><strong>Methods: </strong>Individual case safety reports (ICSRs) were retrieved from the European spontaneous reporting system database (EudraVigilance) from January 1st, 2022, to December 31st, 2023. The reporting odds ratios (RORs) were computed to compare the reporting probability of lymphopenia between a S1PRM versus each other.</p><p><strong>Results: </strong>We retrieved 4017 ICSRs, of which 521 (13%) reported lymphopenia associated with fingolimod (53.3%), siponimod (38.4%), ozanimod (5.4%), and ponesimod (2.1%). The most common reporting source was the healthcare professional (94.2%), and more than half of the ICSRs (62.6%) reported serious lymphopenia. Fingolimod was associated with a lower reporting frequency of lymphopenia compared to siponimod. Both siponimod and fingolimod were associated with a higher reporting frequency of lymphopenia compared to ozanimod; siponimod also had a higher reporting probability in comparison with ponesimod.</p><p><strong>Conclusions: </strong>The most relevant clinical implication of the disproportionality analysis is to increase the awareness of the risk of lymphopenia related to these drugs, thus supporting proactive monitoring and optimizing treatment strategies for people with MS.</p><p><strong>Clinical trial number: </strong>Not applicable.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143812121","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-08DOI: 10.1007/s43440-025-00722-9
Józef Muszyński, Agnieszka Bienert, Rasha Wafaie Elsorady, Filip Rybakowski
Schizophrenia is a primary health concern, imposing a significant burden on both patients and healthcare systems globally. It is a disease with a complex etiology in which both genetic and environmental factors are involved. Despite numerous studies, the mechanism of its origin is still not fully understood. The hypotheses are synaptic, serotonergic, muscarinic, dopaminergic, microRNA-related, and neurodegenerative theories. Treatment to date is mainly based on antipsychotic drugs that act on the dopaminergic system. Although they are effective in reducing positive symptoms, their effect on negative and cognitive symptoms is limited, and their use is often associated with numerous side effects. A breakthrough in the treatment of schizophrenia came with the approval of the first drug with a non-dopaminergic mechanism of action, which opens up new therapeutic possibilities. As a result, there is intensive research into innovative substances that could increase the effectiveness of treatment and improve the quality of life of patients. In this review, we present the current state of knowledge about schizophrenia, its prevalence, risk factors, and its impact on patients' functioning. We pay special attention to new therapeutic directions, including drugs that affect systems other than the dopaminergic one, which could open up new prospects for treating the condition.
{"title":"New pharmacological approaches in the treatment of schizophrenia.","authors":"Józef Muszyński, Agnieszka Bienert, Rasha Wafaie Elsorady, Filip Rybakowski","doi":"10.1007/s43440-025-00722-9","DOIUrl":"https://doi.org/10.1007/s43440-025-00722-9","url":null,"abstract":"<p><p>Schizophrenia is a primary health concern, imposing a significant burden on both patients and healthcare systems globally. It is a disease with a complex etiology in which both genetic and environmental factors are involved. Despite numerous studies, the mechanism of its origin is still not fully understood. The hypotheses are synaptic, serotonergic, muscarinic, dopaminergic, microRNA-related, and neurodegenerative theories. Treatment to date is mainly based on antipsychotic drugs that act on the dopaminergic system. Although they are effective in reducing positive symptoms, their effect on negative and cognitive symptoms is limited, and their use is often associated with numerous side effects. A breakthrough in the treatment of schizophrenia came with the approval of the first drug with a non-dopaminergic mechanism of action, which opens up new therapeutic possibilities. As a result, there is intensive research into innovative substances that could increase the effectiveness of treatment and improve the quality of life of patients. In this review, we present the current state of knowledge about schizophrenia, its prevalence, risk factors, and its impact on patients' functioning. We pay special attention to new therapeutic directions, including drugs that affect systems other than the dopaminergic one, which could open up new prospects for treating the condition.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143811812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-01Epub Date: 2025-01-27DOI: 10.1007/s43440-025-00698-6
Ali Lesani, Fatemeh Mashaknejadian Behbahani, Mohammad Amin Manavi, Razieh Mohammad Jafari, Hamed Shafaroodi, Saman Khosravi, Ahmad Reza Dehpour
Background: Epilepsy, a neurological disorder characterized by recurrent seizures, presents considerable difficulties in treatment, particularly when dealing with drug-resistant cases. Dapsone, recognized for its anti-inflammatory properties, holds promise as a potential therapeutic option. However, its effectiveness in epilepsy requires further investigation. The aim of this study is to explore the effects of dapsone on seizure activity and neuroinflammation, particularly through the nuclear factor erythroid-2-related factor (Nrf2)/ Heme Oxygenase 1 (HO-1) and NOD-like receptor family pyrin domain-containing 3 (NLRP3) pathways, to better understand its therapeutic potential.
Methods: To evaluate the effects of dapsone, two seizure models were utilized in mice: pentylenetetrazole (PTZ)-induced clonic seizures and maximal electroshock (MES)-induced generalized tonic-clonic seizures (GTCS) in mice. The impact of dapsone on neuroinflammatory markers and oxidative stress pathways, specifically Nrf2/HO-1 and NLRP3, as well as interleukin-1β (IL-1β), IL-8, and IL-18, was assessed using Western blotting and ELISA techniques.
Results: In this study, dapsone (2, 5, 10, and 20 mg/kg, ip) showcased a significant increase in clonic seizure threshold following intravenous infusion of PTZ. Notably, doses of 5, 10, and 20 mg/kg exhibited increased latency and decreased the number of seizures. Additionally, dapsone at 10 and 20 mg/kg prevented the incidence of GTCS and subsequent mortality in the MES model. Furthermore, Dapsone demonstrated modulation of Nrf2/ HO-1 and NLRP3 IL-1 β/IL-18 pathways.
Conclusion: This study highlights the therapeutic potential of dapsone in epilepsy, emphasizing the involvement of Nrf2/HO-1 and NLRP3 pathways. These findings provide a foundation for future clinical research aimed at developing dapsone-based therapies for drug-resistant epilepsy.
{"title":"Acute anticonvulsant effects of dapsone on PTZ- and MES-induced seizures in mice: NLRP3 inflammasome inhibition and Nrf2/HO-1 pathway preservation.","authors":"Ali Lesani, Fatemeh Mashaknejadian Behbahani, Mohammad Amin Manavi, Razieh Mohammad Jafari, Hamed Shafaroodi, Saman Khosravi, Ahmad Reza Dehpour","doi":"10.1007/s43440-025-00698-6","DOIUrl":"10.1007/s43440-025-00698-6","url":null,"abstract":"<p><strong>Background: </strong>Epilepsy, a neurological disorder characterized by recurrent seizures, presents considerable difficulties in treatment, particularly when dealing with drug-resistant cases. Dapsone, recognized for its anti-inflammatory properties, holds promise as a potential therapeutic option. However, its effectiveness in epilepsy requires further investigation. The aim of this study is to explore the effects of dapsone on seizure activity and neuroinflammation, particularly through the nuclear factor erythroid-2-related factor (Nrf2)/ Heme Oxygenase 1 (HO-1) and NOD-like receptor family pyrin domain-containing 3 (NLRP3) pathways, to better understand its therapeutic potential.</p><p><strong>Methods: </strong>To evaluate the effects of dapsone, two seizure models were utilized in mice: pentylenetetrazole (PTZ)-induced clonic seizures and maximal electroshock (MES)-induced generalized tonic-clonic seizures (GTCS) in mice. The impact of dapsone on neuroinflammatory markers and oxidative stress pathways, specifically Nrf2/HO-1 and NLRP3, as well as interleukin-1β (IL-1β), IL-8, and IL-18, was assessed using Western blotting and ELISA techniques.</p><p><strong>Results: </strong>In this study, dapsone (2, 5, 10, and 20 mg/kg, ip) showcased a significant increase in clonic seizure threshold following intravenous infusion of PTZ. Notably, doses of 5, 10, and 20 mg/kg exhibited increased latency and decreased the number of seizures. Additionally, dapsone at 10 and 20 mg/kg prevented the incidence of GTCS and subsequent mortality in the MES model. Furthermore, Dapsone demonstrated modulation of Nrf2/ HO-1 and NLRP3 IL-1 β/IL-18 pathways.</p><p><strong>Conclusion: </strong>This study highlights the therapeutic potential of dapsone in epilepsy, emphasizing the involvement of Nrf2/HO-1 and NLRP3 pathways. These findings provide a foundation for future clinical research aimed at developing dapsone-based therapies for drug-resistant epilepsy.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"450-462"},"PeriodicalIF":3.6,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047438","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-01Epub Date: 2025-01-08DOI: 10.1007/s43440-024-00692-4
Rakesh Kumar Singh
Amyloid beta 1-42 (Aβ1-42) peptide is one of the most studied disease-related amyloidogenic peptides implicated in the pathophysiology of Alzheimer's disease (AD). Despite significant scientific breakthroughs in the recent past, the existing non-transgenic animal models do not demonstrate accurate pathology of AD progression. This review has presented a concise mechanistic understanding of the intranasal amyloid-based animal model of AD, along with its advantages, challenges, and major limitations. Furthermore, discussions on how to combat these challenges to pave the road toward developing novel therapeutics for AD, have also been included. Preclinical exploration of repeated intranasal amyloid-beta exposure would certainly aid the translational development of a robust animal model of AD. This will also provide a better understanding of disease progression and pathology in the intranasal animal model.
{"title":"Intranasal amyloid model of Alzheimer's disease - potential opportunities and challenges.","authors":"Rakesh Kumar Singh","doi":"10.1007/s43440-024-00692-4","DOIUrl":"10.1007/s43440-024-00692-4","url":null,"abstract":"<p><p>Amyloid beta 1-42 (Aβ<sub>1-42</sub>) peptide is one of the most studied disease-related amyloidogenic peptides implicated in the pathophysiology of Alzheimer's disease (AD). Despite significant scientific breakthroughs in the recent past, the existing non-transgenic animal models do not demonstrate accurate pathology of AD progression. This review has presented a concise mechanistic understanding of the intranasal amyloid-based animal model of AD, along with its advantages, challenges, and major limitations. Furthermore, discussions on how to combat these challenges to pave the road toward developing novel therapeutics for AD, have also been included. Preclinical exploration of repeated intranasal amyloid-beta exposure would certainly aid the translational development of a robust animal model of AD. This will also provide a better understanding of disease progression and pathology in the intranasal animal model.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"425-433"},"PeriodicalIF":3.6,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142953119","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-01Epub Date: 2024-11-28DOI: 10.1007/s43440-024-00683-5
Mohammad Sheibani, Azam Hosseinzadeh, Iman Fatemi, Ali Jamshidi Naeini, Saeed Mehrzadi
Melatonin, a hormone primarily produced by the pineal gland, exhibits a range of physiological functions that extend beyond its well-known role in regulating circadian rhythms. This hormone influences energy metabolism, modulates insulin sensitivity, and plays a significant role in controlling sleep patterns and food intake. Notably, melatonin is also synthesized in various peripheral organs, including the gastrointestinal system and pancreas, suggesting its function as a local hormone. The presence of melatonin receptors in the pancreas underscores its relevance in pancreatic physiology. Pancreatic disorders, such as diabetes mellitus (DM), pancreatitis, and pancreatic cancer, often stem from inflammatory processes. The majority of these conditions are characterized by dysregulated immune responses and oxidative stress. Melatonin's anti-inflammatory properties are mediated through the inhibition of pro-inflammatory cytokines and the activation of antioxidant enzymes, which help to mitigate cellular damage. Furthermore, melatonin has demonstrated pro-apoptotic effects on cancer cells, promoting cell death in malignant tissues while preserving healthy cells. Thus, melatonin emerges as a multifaceted agent with significant therapeutic potential for pancreatic disorders. Its ability to reduce inflammation and oxidative stress positions it as a promising adjunct therapy for conditions such as diabetes mellitus, pancreatitis, and pancreatic cancer. By modulating immune responses and enhancing cellular resilience through antioxidant mechanisms, melatonin not only addresses the symptoms but also targets the underlying pathophysiological processes associated with these disorders. This review aims to categorize and summarize the impacts of melatonin on pancreatic functions and disorders, emphasizing its potential as a therapeutic agent for managing pancreatic dysfunctions. Future research should focus on elucidating the precise mechanisms by which melatonin exerts its protective effects on pancreatic tissues and exploring optimal dosing strategies for clinical applications. The integration of melatonin into treatment regimens may enhance existing therapies and offer new hope for individuals suffering from pancreatic dysfunctions.
{"title":"Practical application of melatonin for pancreas disorders: protective roles against inflammation, malignancy, and dysfunctions.","authors":"Mohammad Sheibani, Azam Hosseinzadeh, Iman Fatemi, Ali Jamshidi Naeini, Saeed Mehrzadi","doi":"10.1007/s43440-024-00683-5","DOIUrl":"10.1007/s43440-024-00683-5","url":null,"abstract":"<p><p>Melatonin, a hormone primarily produced by the pineal gland, exhibits a range of physiological functions that extend beyond its well-known role in regulating circadian rhythms. This hormone influences energy metabolism, modulates insulin sensitivity, and plays a significant role in controlling sleep patterns and food intake. Notably, melatonin is also synthesized in various peripheral organs, including the gastrointestinal system and pancreas, suggesting its function as a local hormone. The presence of melatonin receptors in the pancreas underscores its relevance in pancreatic physiology. Pancreatic disorders, such as diabetes mellitus (DM), pancreatitis, and pancreatic cancer, often stem from inflammatory processes. The majority of these conditions are characterized by dysregulated immune responses and oxidative stress. Melatonin's anti-inflammatory properties are mediated through the inhibition of pro-inflammatory cytokines and the activation of antioxidant enzymes, which help to mitigate cellular damage. Furthermore, melatonin has demonstrated pro-apoptotic effects on cancer cells, promoting cell death in malignant tissues while preserving healthy cells. Thus, melatonin emerges as a multifaceted agent with significant therapeutic potential for pancreatic disorders. Its ability to reduce inflammation and oxidative stress positions it as a promising adjunct therapy for conditions such as diabetes mellitus, pancreatitis, and pancreatic cancer. By modulating immune responses and enhancing cellular resilience through antioxidant mechanisms, melatonin not only addresses the symptoms but also targets the underlying pathophysiological processes associated with these disorders. This review aims to categorize and summarize the impacts of melatonin on pancreatic functions and disorders, emphasizing its potential as a therapeutic agent for managing pancreatic dysfunctions. Future research should focus on elucidating the precise mechanisms by which melatonin exerts its protective effects on pancreatic tissues and exploring optimal dosing strategies for clinical applications. The integration of melatonin into treatment regimens may enhance existing therapies and offer new hope for individuals suffering from pancreatic dysfunctions.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"315-332"},"PeriodicalIF":3.6,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142740123","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-01DOI: 10.1007/s43440-025-00716-7
Randa Mohammed Zaki, Mohamed A M Ali, Mayada Said, Anis Ahmad Chaudhary, Fehmi Boufahja, Obaid Afzal, Abdelghafar M Abu-Elsaoud, Alyaa S Abdel Halim
Statins, recognized for their lipid-lowering capabilities, have demonstrated osteoanabolic and anti-resorptive effects on bone metabolism. The effects encompass the overexpression of bone morphogenetic proteins, heightened osteoblast activity, and the control of inflammation. Nevertheless, conventional systemic administration of statins has difficulties, including restricted bone bioavailability and possible adverse effects. Recent improvements in targeted and localized drug delivery are revolutionizing the therapeutic landscape for statins in bone applications. This review consolidates existing knowledge regarding the molecular processes by which statins influence bone metabolism and describes novel drug delivery methods such as nano-carriers, biomaterial scaffolds, and controlled-release systems. It seeks to address current knowledge deficiencies and offer insights into how enhanced bioavailability and specificity can optimize the efficiency of statins in bone regeneration. The review integrates molecular insights with novel pharmacological strategies to inform future research and clinical applications, pinpointing critical areas for exploration, such as optimal dose, delivery safety, and clinical efficacy.
{"title":"Molecular mechanisms underlying the effects of statins on bone metabolism: an evolving paradigm of statins delivery modalities for bone regeneration.","authors":"Randa Mohammed Zaki, Mohamed A M Ali, Mayada Said, Anis Ahmad Chaudhary, Fehmi Boufahja, Obaid Afzal, Abdelghafar M Abu-Elsaoud, Alyaa S Abdel Halim","doi":"10.1007/s43440-025-00716-7","DOIUrl":"https://doi.org/10.1007/s43440-025-00716-7","url":null,"abstract":"<p><p>Statins, recognized for their lipid-lowering capabilities, have demonstrated osteoanabolic and anti-resorptive effects on bone metabolism. The effects encompass the overexpression of bone morphogenetic proteins, heightened osteoblast activity, and the control of inflammation. Nevertheless, conventional systemic administration of statins has difficulties, including restricted bone bioavailability and possible adverse effects. Recent improvements in targeted and localized drug delivery are revolutionizing the therapeutic landscape for statins in bone applications. This review consolidates existing knowledge regarding the molecular processes by which statins influence bone metabolism and describes novel drug delivery methods such as nano-carriers, biomaterial scaffolds, and controlled-release systems. It seeks to address current knowledge deficiencies and offer insights into how enhanced bioavailability and specificity can optimize the efficiency of statins in bone regeneration. The review integrates molecular insights with novel pharmacological strategies to inform future research and clinical applications, pinpointing critical areas for exploration, such as optimal dose, delivery safety, and clinical efficacy.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754111","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-01DOI: 10.1007/s43440-025-00719-4
Arkadiusz Adamiszak, Julia Drobińska, Izabela Niewiadomska-Wojnałowicz, Katarzyna Derwich, Edmund Grześkowiak, Agnieszka Bienert
Background: The lack of information on drug-drug interactions in the pediatric population significantly complicates making effective therapeutic decisions. Our study aimed to analyze the rate and risk factors as well as present potential drug-drug interactions (pDDIs) specifically for pediatric patients from the pediatric hemato-oncologic unit, including acute lymphoblastic leukemia (ALL) patients.
Methods: We conducted a six-month prospective study in which clinical pharmacists examined medical records once a week to look for pDDIs using the Lexicomp® Drug Interactions Checker. Spearman's rank coefficient, logistic regression, and the U-Mann-Whitney test were used to identify correlations, analyze risk factors for pDDIs, and compare ALL patients with non-ALL patients, respectively. Recommendations were provided for the D and X pDDIs categories.
Results: We identified 507 pDDIs in 119 screened patients, 388 of which were clinically relevant. Nearly 68% of the patients were exposed to at least one significant interaction. The number of pDDIs was positively correlated with the number of medications (rs=0.75, p < 0.001), off-label used drugs (rs=0.42, p < 0.001), comorbidities (rs=0.21, p = 0.019), and hospitalization length (rs=0.48, p < 0.001). The multivariate analysis revealed that at least 7 administered medications (OR = 8.63; 95% CI = 2.92-25.47) and 13 days in the hospital (OR = 3.47; 95% CI = 1.31-9.19) were risk factors for pDDIs. Furthermore, patients treated for ALL represent an at-risk group with a statistically higher number of drugs taken and pDDIs identified.
Conclusions: Limited data on drug-drug interactions in the pediatric population emphasizes the need for close collaboration between clinical pharmacists and clinicians to improve the safety and effectiveness of pharmacotherapy.
{"title":"Potential drug-drug interactions analysis in Polish pediatric hemato-oncologic unit, including acute lymphoblastic leukemia patients.","authors":"Arkadiusz Adamiszak, Julia Drobińska, Izabela Niewiadomska-Wojnałowicz, Katarzyna Derwich, Edmund Grześkowiak, Agnieszka Bienert","doi":"10.1007/s43440-025-00719-4","DOIUrl":"https://doi.org/10.1007/s43440-025-00719-4","url":null,"abstract":"<p><strong>Background: </strong>The lack of information on drug-drug interactions in the pediatric population significantly complicates making effective therapeutic decisions. Our study aimed to analyze the rate and risk factors as well as present potential drug-drug interactions (pDDIs) specifically for pediatric patients from the pediatric hemato-oncologic unit, including acute lymphoblastic leukemia (ALL) patients.</p><p><strong>Methods: </strong>We conducted a six-month prospective study in which clinical pharmacists examined medical records once a week to look for pDDIs using the Lexicomp<sup>®</sup> Drug Interactions Checker. Spearman's rank coefficient, logistic regression, and the U-Mann-Whitney test were used to identify correlations, analyze risk factors for pDDIs, and compare ALL patients with non-ALL patients, respectively. Recommendations were provided for the D and X pDDIs categories.</p><p><strong>Results: </strong>We identified 507 pDDIs in 119 screened patients, 388 of which were clinically relevant. Nearly 68% of the patients were exposed to at least one significant interaction. The number of pDDIs was positively correlated with the number of medications (r<sub>s</sub>=0.75, p < 0.001), off-label used drugs (r<sub>s</sub>=0.42, p < 0.001), comorbidities (r<sub>s</sub>=0.21, p = 0.019), and hospitalization length (r<sub>s</sub>=0.48, p < 0.001). The multivariate analysis revealed that at least 7 administered medications (OR = 8.63; 95% CI = 2.92-25.47) and 13 days in the hospital (OR = 3.47; 95% CI = 1.31-9.19) were risk factors for pDDIs. Furthermore, patients treated for ALL represent an at-risk group with a statistically higher number of drugs taken and pDDIs identified.</p><p><strong>Conclusions: </strong>Limited data on drug-drug interactions in the pediatric population emphasizes the need for close collaboration between clinical pharmacists and clinicians to improve the safety and effectiveness of pharmacotherapy.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754114","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-01Epub Date: 2025-01-21DOI: 10.1007/s43440-025-00697-7
Dariusz Łaszczych, Aleksandra Czernicka, Katarzyna Łaszczych
Type 1 diabetes (T1D) is an autoimmune disease that leads to the progressive destruction of insulin-producing β cells, resulting in lifelong insulin dependence and a range of severe complications. Beyond conventional glycemic control, innovative therapeutic strategies are needed to address the underlying disease mechanisms. Recent research has highlighted gamma-aminobutyric acid (GABA) as a promising therapeutic target for T1D due to its dual role in modulating both β cell survival and immune response within pancreatic islets. GABA signaling supports β cell regeneration, inhibits α cell hyperactivity, and promotes α-to-β cell transdifferentiation, contributing to improved islet function. Moreover, GABA's influence extends to mitigating T1D complications, including nephropathy, neuropathy, and retinopathy, as well as regulating central nervous system pathways involved in glucose metabolism. This review consolidates the latest advances in GABA-related T1D therapies, covering animal preclinical and human clinical studies and examining the therapeutic potential of GABA receptor modulation, combination therapies, and dietary interventions. Emphasis is placed on the translational potential of GABA-based approaches to enhance β cell viability and counteract autoimmune processes in T1D. Our findings underscore the therapeutic promise of GABA signaling modulation as a novel approach for T1D treatment and encourage further investigation into this pathway's role in comprehensive diabetes management.
{"title":"Targeting GABA signaling in type 1 diabetes and its complications- an update on the state of the art.","authors":"Dariusz Łaszczych, Aleksandra Czernicka, Katarzyna Łaszczych","doi":"10.1007/s43440-025-00697-7","DOIUrl":"10.1007/s43440-025-00697-7","url":null,"abstract":"<p><p>Type 1 diabetes (T1D) is an autoimmune disease that leads to the progressive destruction of insulin-producing β cells, resulting in lifelong insulin dependence and a range of severe complications. Beyond conventional glycemic control, innovative therapeutic strategies are needed to address the underlying disease mechanisms. Recent research has highlighted gamma-aminobutyric acid (GABA) as a promising therapeutic target for T1D due to its dual role in modulating both β cell survival and immune response within pancreatic islets. GABA signaling supports β cell regeneration, inhibits α cell hyperactivity, and promotes α-to-β cell transdifferentiation, contributing to improved islet function. Moreover, GABA's influence extends to mitigating T1D complications, including nephropathy, neuropathy, and retinopathy, as well as regulating central nervous system pathways involved in glucose metabolism. This review consolidates the latest advances in GABA-related T1D therapies, covering animal preclinical and human clinical studies and examining the therapeutic potential of GABA receptor modulation, combination therapies, and dietary interventions. Emphasis is placed on the translational potential of GABA-based approaches to enhance β cell viability and counteract autoimmune processes in T1D. Our findings underscore the therapeutic promise of GABA signaling modulation as a novel approach for T1D treatment and encourage further investigation into this pathway's role in comprehensive diabetes management.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"409-424"},"PeriodicalIF":3.6,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143009873","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-04-01Epub Date: 2025-01-08DOI: 10.1007/s43440-024-00688-0
Snigdha Sama, Sunjoo Cho, Ashish K Rehni, Wenche Jy, Kunjan R Dave
Background: Current therapies to treat excessive bleeding are associated with significant complications, which may outweigh their benefits. Red blood cell-derived microparticles (RMPs) are a promising hemostatic agent. Previous studies demonstrated that they reduce bleeding in animal models, correct coagulation defects in patient blood, and have an excellent safety profile. However, their exact mechanism of action is not known. We investigated the potential role of RMPs on primary and secondary hemostasis.
Methods: To evaluate the effects of RMPs, prepared using high-pressure extrusion, on primary hemostasis, we employed platelet aggregometry with platelet inhibitors, eptifibatide, and ticagrelor, with and without RMPs. To evaluate their effects on secondary hemostasis, we employed thromboelastography with plasma deficient in factors VII, VIII, IX, XI, and XII with and without RMPs.
Results: We found that RMPs significantly increased collagen-induced platelet aggregation. However, there were no significant differences with and without RMP in the presence of the platelet inhibitors, indicating that RMPs may work through these receptors, either directly or indirectly. For secondary hemostasis, RMPs significantly decreased clotting times for plasma deficient in factors VII, VIII, IX, and XI but not in XII.
Conclusions: Our results indicate that RMPs enhance primary hemostasis and both pathways of secondary hemostasis.
{"title":"Red cell microparticles produced using high-pressure extrusion enhance both primary and secondary hemostasis.","authors":"Snigdha Sama, Sunjoo Cho, Ashish K Rehni, Wenche Jy, Kunjan R Dave","doi":"10.1007/s43440-024-00688-0","DOIUrl":"10.1007/s43440-024-00688-0","url":null,"abstract":"<p><strong>Background: </strong>Current therapies to treat excessive bleeding are associated with significant complications, which may outweigh their benefits. Red blood cell-derived microparticles (RMPs) are a promising hemostatic agent. Previous studies demonstrated that they reduce bleeding in animal models, correct coagulation defects in patient blood, and have an excellent safety profile. However, their exact mechanism of action is not known. We investigated the potential role of RMPs on primary and secondary hemostasis.</p><p><strong>Methods: </strong>To evaluate the effects of RMPs, prepared using high-pressure extrusion, on primary hemostasis, we employed platelet aggregometry with platelet inhibitors, eptifibatide, and ticagrelor, with and without RMPs. To evaluate their effects on secondary hemostasis, we employed thromboelastography with plasma deficient in factors VII, VIII, IX, XI, and XII with and without RMPs.</p><p><strong>Results: </strong>We found that RMPs significantly increased collagen-induced platelet aggregation. However, there were no significant differences with and without RMP in the presence of the platelet inhibitors, indicating that RMPs may work through these receptors, either directly or indirectly. For secondary hemostasis, RMPs significantly decreased clotting times for plasma deficient in factors VII, VIII, IX, and XI but not in XII.</p><p><strong>Conclusions: </strong>Our results indicate that RMPs enhance primary hemostasis and both pathways of secondary hemostasis.</p>","PeriodicalId":19947,"journal":{"name":"Pharmacological Reports","volume":" ","pages":"508-516"},"PeriodicalIF":3.6,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11911262/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142953134","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}