151
|
Sun Y, Zhu C, Huang L, Luo C, Ju P, Chen J. Identification of key modules in metabolic syndrome induced by second-generation antipsychotics based on co-expression network analysis. Comput Struct Biotechnol J 2024; 23:723-731. [PMID: 38292473 PMCID: PMC10826125 DOI: 10.1016/j.csbj.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
Background Second-generation antipsychotics (SGAs) frequently cause metabolic syndrome (MetS), which raises the risk of heart disease, type 2 diabetes, morbid obesity, atherosclerosis, and hypertension. MetS also impairs cognitive function in patients with schizophrenia. However, the fundamental reasons of MetS caused by SGAs are not yet fully understood. Thus, we aimed to identify potential therapeutic targets for MetS induced by SGAs. Methods The serum biochemical parameters and the RNA-sequencing of peripheral blood mononuclear cells were measured in three groups (healthy controls and patients with schizophrenia with and without MetS taking SGAs). The study of the weighted gene co-expression network was utilized to pinpoint modules that were significantly connected to clinical markers. Results Statistical analysis showed significant differences in triglyceride and high-density lipoprotein among the three groups. The TNF signaling pathway, TGF-β signaling pathway, fatty acid metabolism, NF-kappa B signaling pathway, MAPK signaling pathway, and Toll-like receptor signaling pathway were the pathways that were primarily enriched in the two unique co-expression network modules that were found. Finally, five specific genes (TNF, CXCL8, IL1B, TIMP1, and ESR1) associated with metabolism and immunity pathways were identified. Conclusions This study indicated that SGAs differentially induced MetS of patients with schizophrenia through metabolic and inflammation-related pathways. Therefore, the potential side effects of drugs on inflammatory processes need to be considered when using SGAs for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Ying Sun
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Cuizhen Zhu
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, China
- Anhui Clinical Center for Mental and Psychological Diseases, Hefei Fourth People's Hospital, Hefei, Anhui, China
- Anhui Mental Health Center, Hefei, Anhui, China
| | - Lixuan Huang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Luo
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peijun Ju
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
- Shanghai Institute of Traditional Chinese Medicine for Mental Health, Shanghai, China
| | - Jianhua Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
- Shanghai Institute of Traditional Chinese Medicine for Mental Health, Shanghai, China
- Yueyang Hospital of Integrated Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
152
|
Chen B, de Launoit E, Meseguer D, Garcia Caceres C, Eichmann A, Renier N, Schneeberger M. The interactions between energy homeostasis and neurovascular plasticity. Nat Rev Endocrinol 2024; 20:749-759. [PMID: 39054359 DOI: 10.1038/s41574-024-01021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Food intake and energy expenditure are sensed and processed by multiple brain centres to uphold energy homeostasis. Evidence from the past decade points to the brain vasculature as a new critical player in regulating energy balance that functions in close association with the local neuronal networks. Nutritional imbalances alter many properties of the neurovascular system (such as neurovascular coupling and blood-brain barrier permeability), thus suggesting a bidirectional link between the nutritional milieu and neurovascular health. Increasing numbers of people are consuming a Western diet (comprising ultra-processed food with high-fat and high-sugar content) and have a sedentary lifestyle, with these factors contributing to the current obesity epidemic. Emerging pharmacological interventions (for example, glucagon-like peptide 1 receptor agonists) successfully trigger weight loss. However, whether these approaches can reverse the detrimental effects of long-term exposure to the Western diet (such as neurovascular uncoupling, neuroinflammation and blood-brain barrier disruption) and maintain stable body weight in the long-term needs to be clarified in addition to possible adverse effects. Lifestyle interventions revert the nutritional trigger for obesity and positively affect our overall health, including the cardiovascular system. This Perspective examines how lifestyle interventions affect the neurovascular system and neuronal networks.
Collapse
Affiliation(s)
- Bandy Chen
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Elisa de Launoit
- Sorbonne Université, Institut Du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS UMR 7225, Paris, France
| | - David Meseguer
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Cristina Garcia Caceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich & German Center for Diabetes Research (DZD), Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anne Eichmann
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, Paris, France
| | - Nicolas Renier
- Sorbonne Université, Institut Du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Marc Schneeberger
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
- Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
153
|
Zur Nedden S, Safari MS, Weber D, Kuenkel L, Garmsiri C, Lang L, Orset C, Freret T, Haelewyn B, Hotze M, Kwiatkowski M, Sarg B, Faserl K, Savic D, Skvortsova II, Krogsdam A, Carollo S, Trajanoski Z, Oberacher H, Zlotek D, Ostermaier F, Cameron A, Baier G, Baier-Bitterlich G. Protein kinase N1 deficiency results in upregulation of cerebral energy metabolism and is highly protective in in vivo and in vitro stroke models. Metabolism 2024; 161:156039. [PMID: 39332493 DOI: 10.1016/j.metabol.2024.156039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND AND AIM We recently identified protein kinase N1 (PKN1) as a master regulator of brain development. However, its function in the adult brain has not been clearly established. In this study, we assessed the cerebral energetic phenotype of wildtype (WT) and global Pkn1 knockout (Pkn1-/-) animals under physiological and pathophysiological conditions. METHODS Cerebral energy metabolism was analyzed by 13C6-glucose tracing in vivo and real time seahorse analysis of extracellular acidification rates as well as mitochondrial oxygen consumption rates (OCR) of brain slice punches in vitro. Isolated WT and Pkn1-/- brain mitochondria were tested for differences in OCR with different substrates. Metabolite levels were determined by mass spectrometric analysis in brain slices under control and energetic stress conditions, induced by oxygen-glucose deprivation and reperfusion, an in vitro model of ischemic stroke. Differences in enzyme activities were assessed by enzymatic assays, western blotting and bulk RNA sequencing. A middle cerebral artery occlusion stroke model was used to analyze lesion volumes and functional recovery in WT and Pkn1-/- mice. RESULTS Pkn1 deficiency resulted in a remarkable upregulation of cerebral energy metabolism, in vivo and in vitro. This was due to two separate mechanisms involving an enhanced glycolytic flux and higher pyruvate-induced mitochondrial OCR. Mechanistically we show that Pkn1-/- brain tissue exhibits an increased activity of the glycolysis rate-limiting enzyme phosphofructokinase. Additionally, glucose-1,6-bisphosphate levels, a metabolite that increases mitochondrial pyruvate uptake, were elevated upon Pkn1 deficiency. Consequently, Pkn1-/- brain slices had more ATP and a greater accumulation of ATP degradation metabolites during energetic stress. This translated into increased phosphorylation and activity of adenosine monophosphate (AMP)-activated protein kinase (AMPK) during in vitro stroke. Accordingly, Pkn1-/- brain slices showed a post-ischemic transcriptional upregulation of energy metabolism pathways and Pkn1 deficiency was strongly protective in in vitro and in vivo stroke models. While inhibition of mitochondrial pyruvate uptake only moderately affected the protective phenotype, inhibition of AMPK in Pkn1-/- slices increased post-ischemic cell death in vitro. CONCLUSION This is the first study to comprehensively demonstrate an essential and unique role of PKN1 in cerebral energy metabolism, regulating glycolysis and mitochondrial pyruvate-induced respiration. We further uncovered a highly protective phenotype of Pkn1 deficiency in both, in vitro and in vivo stroke models, validating inhibition of PKN1 as a promising new therapeutic target for the development of novel stroke therapies.
Collapse
Affiliation(s)
- Stephanie Zur Nedden
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria.
| | - Motahareh S Safari
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Dido Weber
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Louisa Kuenkel
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Carolin Garmsiri
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Luisa Lang
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Cyrille Orset
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute of Blood and Brain @ Caen-Normandie (BB@C), GIP Cyceron, Caen, France
| | - Tom Freret
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute of Blood and Brain @ Caen-Normandie (BB@C), GIP Cyceron, Caen, France
| | - Benoît Haelewyn
- Normandie University, UNICAEN, ESR3P, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute of Blood and Brain @ Caen-Normandie (BB@C), GIP Cyceron, Caen, France
| | - Madlen Hotze
- University of Innsbruck, Department of Biochemistry, Institute of Bioanalytic & Intermediary Metabolism, 6020 Innsbruck, Austria
| | - Marcel Kwiatkowski
- University of Innsbruck, Department of Biochemistry, Institute of Bioanalytic & Intermediary Metabolism, 6020 Innsbruck, Austria
| | - Bettina Sarg
- Medical University of Innsbruck, CCB-Biocenter, Institute of Medical Biochemistry, Protein Core Facility, 6020 Innsbruck, Austria
| | - Klaus Faserl
- Medical University of Innsbruck, CCB-Biocenter, Institute of Medical Biochemistry, Protein Core Facility, 6020 Innsbruck, Austria
| | - Dragana Savic
- Medical University of Innsbruck, Laboratory for Experimental and Translational Research on Radiation Oncology (EXTRO-Lab), Department of Therapeutic Radiology and Oncology, 6020 Innsbruck, Austria; Tyrolean Cancer Research Institute, Innsbruck A-6020, Austria
| | - Ira-Ida Skvortsova
- Medical University of Innsbruck, Laboratory for Experimental and Translational Research on Radiation Oncology (EXTRO-Lab), Department of Therapeutic Radiology and Oncology, 6020 Innsbruck, Austria; Tyrolean Cancer Research Institute, Innsbruck A-6020, Austria
| | - Anne Krogsdam
- Medical University of Innsbruck, CCB-Biocenter, Institute of Bioinformatics, 6020 Innsbruck, Austria
| | - Sandro Carollo
- Medical University of Innsbruck, CCB-Biocenter, Institute of Bioinformatics, 6020 Innsbruck, Austria
| | - Zlatko Trajanoski
- Medical University of Innsbruck, CCB-Biocenter, Institute of Bioinformatics, 6020 Innsbruck, Austria
| | - Herbert Oberacher
- Medical University of Innsbruck, Institute of Legal Medicine and Core Facility Metabolomics, 6020 Innsbruck, Austria
| | - Dominik Zlotek
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Florian Ostermaier
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Angus Cameron
- Kinase Biology Laboratory, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Gottfried Baier
- Medical University of Innsbruck, Institute for Cell Genetics, 6020 Innsbruck, Austria
| | - Gabriele Baier-Bitterlich
- Medical University of Innsbruck, CCB-Biocenter, Institute of Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| |
Collapse
|
154
|
Berriel Diaz M, Rohm M, Herzig S. Cancer cachexia: multilevel metabolic dysfunction. Nat Metab 2024; 6:2222-2245. [PMID: 39578650 DOI: 10.1038/s42255-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024]
Abstract
Cancer cachexia is a complex metabolic disorder marked by unintentional body weight loss or 'wasting' of body mass, driven by multiple aetiological factors operating at various levels. It is associated with many malignancies and significantly contributes to cancer-related morbidity and mortality. With emerging recognition of cancer as a systemic disease, there is increasing awareness that understanding and treatment of cancer cachexia may represent a crucial cornerstone for improved management of cancer. Here, we describe the metabolic changes contributing to body wasting in cachexia and explain how the entangled action of both tumour-derived and host-amplified processes induces these metabolic changes. We discuss energy homeostasis and possible ways that the presence of a tumour interferes with or hijacks physiological energy conservation pathways. In that context, we highlight the role played by metabolic cross-talk mechanisms in cachexia pathogenesis. Lastly, we elaborate on the challenges and opportunities in the treatment of this devastating paraneoplastic phenomenon that arise from the complex and multifaceted metabolic cross-talk mechanisms and provide a status on current and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany.
| |
Collapse
|
155
|
Roy SC, Sapkota S, Pasula MB, Briski KP. In Vivo Glucose Transporter-2 Regulation of Dorsomedial Versus Ventrolateral VMN Astrocyte Metabolic Sensor and Glycogen Metabolic Enzyme Gene Expression in Female Rat. Neurochem Res 2024; 49:3367-3382. [PMID: 39306597 DOI: 10.1007/s11064-024-04246-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024]
Abstract
Astrocyte glycogenolysis shapes ventromedial hypothalamic nucleus (VMN) regulation of glucostasis in vivo. Glucose transporter-2 (GLUT2), a plasma membrane glucose sensor, controls hypothalamic primary astrocyte culture glycogen metabolism in vitro. In vivo gene silencing tools and single-cell laser-catapult-microdissection/multiplex qPCR techniques were used here to examine whether GLUT2 governs dorsomedial (VMNdm) and/or ventrolateral (VMNvl) VMN astrocyte metabolic sensor and glycogen metabolic enzyme gene profiles. GLUT2 gene knockdown diminished astrocyte GLUT2 mRNA in both VMN divisions. Hypoglycemia caused GLUT2 siRNA-reversible up-regulation of this gene profile in the VMNdm, but down-regulated VMNvl astrocyte GLUT2 transcription. GLUT2 augmented baseline VMNdm and VMNvl astrocyte glucokinase (GCK) gene expression, but increased (VMNdm) or reduced (VMNvl) GCK transcription during hypoglycemia. GLUT2 imposed opposite control, namely stimulation versus inhibition of VMNdm or VMNvl astrocyte 5'-AMP-activated protein kinase-alpha 1 and -alpha 2 gene expression, respectively. GLUT2 stimulated astrocyte glycogen synthase (GS) gene expression in each VMN division. GLUT2 inhibited transcription of the AMP-sensitive glycogen phosphorylase (GP) isoform GP-brain type (GPbb) in each site, yet diminished (VMNdm) or augmented (VMNvl) astrocyte GP-muscle type (GPmm) mRNA. GLUT2 enhanced VMNdm and VMNvl glycogen accumulation during euglycemia, and curbed hypoglycemia-associated VMNdm glycogen depletion. Results show that VMN astrocytes exhibit opposite, division-specific GLUT2 transcriptional responsiveness to hypoglycemia. Data document divergent GLUT2 control of GCK, AMPK catalytic subunit, and GPmm gene profiles in VMNdm versus VMNvl astrocytes. Ongoing studies seek to determine how differential GLUT2 regulation of glucose and energy sensor function and glycogenolysis in each VMN location may affect local neuron responses to hypoglycemia.
Collapse
Affiliation(s)
- Sagor C Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA
| | - Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA
| | - Madhu Babu Pasula
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, 71201, USA.
- UL System Foundation and Willis-Knighton Health Systems Professorship in Toxicology, College of Pharmacy, University of Louisiana at Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA, 71201, USA.
| |
Collapse
|
156
|
Padilha DMH, Mendes MCS, Takahashi MES, Lascala F, Silveira MN, Pozzuto L, Carrilho LAO, Guerra LD, Moreira RCL, Branbilla SR, Ramos CD, Carvalheira JBC. Subcutaneous adipose tissue radiodensity: An emerging risk factor for severe COVID-19. Nutrition 2024; 128:112561. [PMID: 39277984 DOI: 10.1016/j.nut.2024.112561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/13/2024] [Accepted: 07/25/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND Adipose tissue radiodensity and metabolic activity may influence COVID-19 outcomes. This study evaluated the association between adipose tissue characteristics and clinical outcomes in COVID-19 patients. METHODS Two retrospective cohorts of hospitalized COVID-19 patients were analyzed. Subcutaneous adipose tissue radiodensity (SATR) and visceral adipose tissue radiodensity were assessed by computed tomography. Fluorine-18-labelled fluorodeoxyglucose PET/computed tomography measured adipose tissue metabolic activity. Associations with mortality, length of stay, ventilation requirement, and complications were examined using regression analyses. RESULTS High SATR was independently associated with increased mortality risk (OR: 2.70; P = 0.033), longer hospitalization (P < 0.001), higher rates of mechanical ventilation (P = 0.007), and complications: acute kidney injury (P = 0.001), secondary infection (P = 0.007), shock (P = 0.010), and pulmonary embolism (P = 0.011). SATR positively correlated with SAT glucose uptake (ρ = 0.52) and negatively with leptin levels (ρ = -0.48). CONCLUSIONS Elevated SATR at COVID-19 diagnosis predicts disease severity and worse outcomes. SATR is a potential prognostic biomarker for acute and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Daniela M H Padilha
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil; Nestlé Health Science, Lausanne, Switzerland
| | - Maria C S Mendes
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil; Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Maria E S Takahashi
- Institute of Physics "Gleb Wataghin", University of Campinas, Campinas SP, Brazil
| | - Fabiana Lascala
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Marina N Silveira
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Lara Pozzuto
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Larissa A O Carrilho
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Lívia D Guerra
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Rafaella C L Moreira
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Sandra R Branbilla
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - Celso Darío Ramos
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil
| | - José B C Carvalheira
- Department of Anesthesiology, Oncology and Radiology, School of Medical Sciences, University of Campinas, Campinas SP, Brazil.
| |
Collapse
|
157
|
Ajwani J, Hwang E, Portillo B, Lieu L, Wallace B, Kabahizi A, He Z, Dong Y, Grose K, Williams KW. Upregulation of Xbp1 in NPY/AgRP neurons reverses diet-induced obesity and ameliorates leptin and insulin resistance. Neuropeptides 2024; 108:102461. [PMID: 39180950 PMCID: PMC11568921 DOI: 10.1016/j.npep.2024.102461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/27/2024]
Abstract
The molecular mechanisms underlying neuronal leptin and insulin resistance in obesity and diabetes are not fully understood. In this study, we show that induction of the unfolded protein response transcription factor, spliced X-box binding protein 1 (Xbp1s), in Agouti-Related Peptide (AgRP) neurons alone, is sufficient to not only protect against but also significantly reverse diet-induced obesity (DIO) as well as improve leptin and insulin sensitivity, despite activation of endoplasmic reticulum stress. We also demonstrate that constitutive expression of Xbp1s in AgRP neurons contributes to improved insulin sensitivity and glucose tolerance. Together, our results identify critical molecular mechanisms linking ER stress in arcuate AgRP neurons to acute leptin and insulin resistance as well as liver glucose metabolism in DIO and diabetes.
Collapse
Affiliation(s)
- Jason Ajwani
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Eunsang Hwang
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Bryan Portillo
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Linh Lieu
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Briana Wallace
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Anita Kabahizi
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Zhenyan He
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Yanbin Dong
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kyle Grose
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
158
|
Mukherjee S, Kumar D, Guha D. Insights of probiotics as an alternative medicine for cancer therapy, mechanism, and applications. MEDICINE IN MICROECOLOGY 2024; 22:100111. [DOI: 10.1016/j.medmic.2024.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
159
|
Yamashima T. 4-Hydroxynonenal from Mitochondrial and Dietary Sources Causes Lysosomal Cell Death for Lifestyle-Related Diseases. Nutrients 2024; 16:4171. [PMID: 39683565 DOI: 10.3390/nu16234171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Excessive consumption of vegetable oils such as soybean and canolla oils containing ω-6 polyunsaturated fatty acids is considered one of the most important epidemiological factors leading to the progression of lifestyle-related diseases. However, the underlying mechanism of vegetable-oil-induced organ damage is incompletely elucidated. Since proopiomelanocortin (POMC) neurons in the hypothalamus are related to the control of appetite and energy expenditure, their cell degeneration/death is crucial for the occurrence of obesity. In patients with metabolic syndrome, saturated fatty acids, especially palmitate, are used as an energy source. Since abundant reactive oxygen species are produced during β-oxidation of the palmitate in mitochondria, an increased amount of 4-hydroxy-2-nonenal (4-HNE) is endogenously generated from linoleic acids constituting cardiolipin of the inner membranes. Further, due to the daily intake of deep-fried foods and/or high-fat diets cooked using vegetable oils, exogenous 4-HNE being generated via lipid peroxidation during heating is incorporated into the blood. By binding with atheromatous and/or senile plaques, 4-HNE inactivates proteins via forming hybrid covalent chemical addition compounds and causes cellular dysfunction and tissue damage by the specific oxidation carbonylation. 4-HNE overstimulates G-protein-coupled receptors to induce abnormal Ca2+ mobilization and µ-calpain activation. This endogenous and exogenous 4-HNE synergically causes POMC neuronal degeneration/death and obesity. Then, the resultant metabolic disorder facilitates degeneration/death of hippocampal neurons, pancreatic β-cells, and hepatocytes. Hsp70.1 is a molecular chaperone which is crucial for both protein quality control and the stabilization of lysosomal limiting membranes. Focusing on the monkey hippocampus after ischemia, previously we formulated the 'calpain-cathepsin hypothesis', i.e., that calpain-mediated cleavage of carbonylated Hsp70.1 is a trigger of programmed neuronal death. This review aims to report that in diverse organs, lysosomal cell degeneration/death occurs via the calpain-cathepsin cascade after the consecutive injections of synthetic 4-HNE in monkeys. Presumably, 4-HNE is a root substance of lysosomal cell death for lifestyle-related diseases.
Collapse
Affiliation(s)
- Tetsumori Yamashima
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Takara-machi 13-1, Kanazawa 920-8040, Japan
| |
Collapse
|
160
|
Jia Y, Wei Z, Feng J, Lei M, Yang Y, Liu J, Ma Y, Chen W, Huang G, Genin GM, Guo X, Li Y, Xu F. A Heart Rate Matched Patch for Mechano-Chemical Treatment of Myocardial Infarction: Optimal Design and Transspecies Application. RESEARCH (WASHINGTON, D.C.) 2024; 7:0517. [PMID: 39582687 PMCID: PMC11582187 DOI: 10.34133/research.0517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 11/26/2024]
Abstract
After myocardial infarction (MI), ventricular dilation and the microscopic passive stretching of the infarcted border zone is the meaning contributor to the continuous expansion of myocardial fibrosis. Epicardial hydrogel patches have been demonstrated to alleviate this sequela of MI in small-animal models. However, these have not been successfully translated to humans or even large animals, in part because of challenges in attaining both the greater stiffness and slower viscoelastic relaxation that mathematical models predict to be optimal for application to larger, slower-beating hearts. Here, using borate-based dynamic covalent chemistry, we develop an injectable "heart rate matched" viscoelastic gelatin (VGtn) hydrogel with a gel point tunable across the stiffnesses and frequencies that are predicted to transspecies and cross-scale cardiac repair after MI. Small-animal experiments demonstrated that, compared to heart rate mismatched patches, the heart rate matched VGtn patches inhibited ventricular bulging and attenuated stress concentrations in the myocardium after MI. In particular, the viscoelastic patch can coordinate the microscopic strain at the infarction boundary. VGtn loaded with anti-fibrotic agents further reduced myocardial damage and promoted angiogenesis in the myocardium. The tuned heart rate matched patches demonstrated similar benefits in a larger-scale and lower heart rate porcine MI model. Results suggest that heart rate matched VGtn patches may hold potential for clinical translation.
Collapse
Affiliation(s)
- Yuanbo Jia
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, P.R. China
- Key Laboratory of Surgical Critical Care and Life Support (Xi’an Jiaotong University), Ministry of Education, Xi’an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Zhao Wei
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Jinteng Feng
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Meng Lei
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Yanshen Yang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Jingyi Liu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Yufei Ma
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| | - Weiguo Chen
- Department of Cardiology, Tangdu Hospital,
the Air Force Military Medical University, Xi’an, Shaanxi 710038, P.R. China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering,
Wuhan University, Wuhan 430072, P.R. China
| | - Guy M. Genin
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
- Department of Mechanical Engineering & Materials Science,
Washington University in St. Louis, St. Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology,
Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Xiaogang Guo
- Department of Cardiology, the First Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou 310003, P.R. China
| | - Yan Li
- Department of Cardiology, Tangdu Hospital,
the Air Force Military Medical University, Xi’an, Shaanxi 710038, P.R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049, P.R. China
- MOE Key Laboratory of Biomedical Information Engineering,
School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, P.R. China
| |
Collapse
|
161
|
Smeda M, Maleki EH, Jasztal A. A possible role of plasmin-dependent activation of TGF-β in cancer-associated thrombosis: Implications for therapy. Cancer Metastasis Rev 2024; 44:2. [PMID: 39576347 PMCID: PMC11584501 DOI: 10.1007/s10555-024-10222-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024]
Abstract
While the prevalence of cancer-associated thrombosis (CAT) is high in cancer patients, its molecular mechanisms have not been fully elucidated. Moreover, the risks of recurrent CAT events and mortality remain high in cancer patients despite the introduction of anticoagulant/antiplatelet therapy. Here, we discuss the possibility that increased plasmin activity driven by anticoagulant/antiplatelet treatment might be the major mechanism responsible for the activation of an excess of cancer-derived transforming growth factor-beta (TGF-β) originating from cancer cells and the tumour microenvironment. Hence, high coagulation and fibrinolysis rates in cancer patients may be linked to high rates of TGF-β activation, especially the excess of TGF-β derived from cancer cells. In turn, high TGF-β activation could contribute directly to maintaining high thrombotic risk and CAT recurrence in cancer patients since TGF-β signalling increases gene expression and secretion of the fibrinolysis inhibitor plasminogen activator inhibitor 1 (PAI1). Thus, TGF-β could directly contribute to the high number of deaths among patients with cancer experiencing CAT, despite anticoagulant/antiplatelet treatment. In a longer-term perspective, increased TGF-β activation, by supporting a pro-coagulant cancer microenvironment, might also accelerate cancer progression. This review aims to discuss the published evidence that might support the scenario described above, and to put forward the hypothesis that cancer patients experiencing CAT events would largely benefit from anti-TGF-β therapy.
Collapse
Affiliation(s)
- Marta Smeda
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, 30-348, Poland.
| | - Ebrahim H Maleki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, 30-348, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, 30-348, Poland
| |
Collapse
|
162
|
Tamayo-Molina YS, Giraldo MA, Rodríguez BA, Machado-Rodríguez G. A biological rhythm in the hypothalamic system links sleep-wake cycles with feeding-fasting cycles. Sci Rep 2024; 14:28897. [PMID: 39572629 PMCID: PMC11582708 DOI: 10.1038/s41598-024-77915-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/28/2024] [Indexed: 11/24/2024] Open
Abstract
The hypothalamus senses the appetite-regulating hormones and also coordinates the metabolic function in alignment with the circadian rhythm. This alignment is essential to maintain the physiological conditions that prevent clinically important comorbidities, such as obesity or type-2 diabetes. However, a complete model of the hypothalamus that relates food intake with circadian rhythms and appetite hormones has not yet been developed. In this work, we present a computational model that accurately allows interpreting neural activity in terms of hormone regulation and sleep-wake cycles. We used a conductance-based model, which consists of a system of four differential equations that considers the ionotropic and metabotropic receptors, and the input currents from homeostatic hormones. We proposed a logistic function that fits available experimental data of insulin hormone concentration and added it into a short-term ghrelin model that served as an input to our dynamical system. Our results show a double oscillatory system, one synchronized by light-regulated sleep-wake cycles and the other by food-regulated feeding-fasting cycles. We have also found that meal timing frequency is highly relevant for the regulation of the hypothalamus neurons. We therefore present a mathematical model to explore the plausible link between the circadian rhythm and the endogenous food clock.
Collapse
Affiliation(s)
- Y S Tamayo-Molina
- Biophysics Group, Institute of Physics, University of Antioquia, Medellin, Colombia.
- Grupo de Fundamentos y Enseñanza de la Física y los Sistemas Dinámicos, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellin, Colombia.
| | - M A Giraldo
- Biophysics Group, Institute of Physics, University of Antioquia, Medellin, Colombia.
| | - B A Rodríguez
- Grupo de Fundamentos y Enseñanza de la Física y los Sistemas Dinámicos, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellin, Colombia
| | - G Machado-Rodríguez
- Biophysics Group, Institute of Physics, University of Antioquia, Medellin, Colombia
- Grupo de Fundamentos y Enseñanza de la Física y los Sistemas Dinámicos, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellin, Colombia
| |
Collapse
|
163
|
Liu S, Zhang X, Wang W, Li X, Sun X, Zhao Y, Wang Q, Li Y, Hu F, Ren H. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol Cancer 2024; 23:261. [PMID: 39574178 PMCID: PMC11580516 DOI: 10.1186/s12943-024-02165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024] Open
Abstract
Metabolic alterations, a hallmark of cancer, enable tumor cells to adapt to their environment by modulating glucose, lipid, and amino acid metabolism, which fuels rapid growth and contributes to treatment resistance. In primary breast cancer, metabolic shifts such as the Warburg effect and enhanced lipid synthesis are closely linked to chemotherapy failure. Similarly, metastatic lesions often display distinct metabolic profiles that not only sustain tumor growth but also confer resistance to targeted therapies and immunotherapies. The review emphasizes two major aspects: the mechanisms driving metabolic resistance in both primary and metastatic breast cancer, and how the unique metabolic environments in metastatic sites further complicate treatment. By targeting distinct metabolic vulnerabilities at both the primary and metastatic stages, new strategies could improve the efficacy of existing therapies and provide better outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Shan Liu
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xingda Zhang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenzheng Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Sun
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuqian Zhao
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qi Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingpu Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Fangjie Hu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - He Ren
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
164
|
Monti M, Limarzi F, Oboldi D, Sbrancia M, Pallotti MC, Miserocchi G, Ghini V, Zanuccoli S, Cagnazzo S, Frassineti GL. Squamous cell carcinoma of the stomach: focus on a heterogeneous disease at diagnosis. Case report and literature review. Front Oncol 2024; 14:1419923. [PMID: 39634259 PMCID: PMC11614721 DOI: 10.3389/fonc.2024.1419923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Primary squamous cell carcinoma (SCC) can originate in different parts of the body, including the head, neck, lung, bronchus, cervix uteri, esophagus, and cardia, and subsequently metastasize to the stomach. Primary gastric squamous cell carcinoma (GSCC) is a rare disease. To better understand GSCC, we present the case of a 72-year-old woman with a primary GSCC. A chest and abdominal CT scan highlighted a 36×26 mm mass with a 41 mm longitudinal diameter, which included the origin of the celiac tripod. The disease appeared to originate exophytically from the gastric wall. An ultrasound-endoscopy showed a hypoechoic formation with not well-defined margins measuring 40×30 mm involving the origin of the celiac tripod, about 10 mm from the gastric wall. An endoscopic fine-needle aspiration showed a poorly differentiated carcinoma. A PET/CT scan showed a hyperaccumulation of the known expansive formation at the celiac tripod (SUV 11.9) without specific cleavage planes from the stomach. A gastroscopy showed a regular esophagus and an absence of gastric protruding lesions. In the subcardial area, on the posterior wall, there was a slightly raised sub-centimetric area covered by bleeding mucosa where the biopsy had been performed. The pathological report showed chronic gastritis. An eco-endoscopy confirmed a hypoechoic neoformation measuring 30×40 mm that appeared to originate from the muscular layer of the gastric wall. The biopsy report was positive for broad-spectrum cytokeratins (AE1/AE3), CK5/6/7, p40, p63 and negative for CK20, PAS, TTF-1, anti-smooth muscle actin, CD45 (LCA), ERG, and S100. The clinical picture suggested poorly differentiated carcinoma with squamous differentiation. We analyzed the main classifications of GSCC cases and compared their characteristics. It is clear that to have an appropriate definition of GSCC, well-defined diagnostic criteria are needed. Currently, there is no consensus. For practical purposes, it would be better to include a panel of CK and p40 to distinguish GSCC from adenocarcinoma. A GSCC outside the mucosa is not rare and could be a true entity.
Collapse
Affiliation(s)
- Manlio Monti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | | | - Devil Oboldi
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Monica Sbrancia
- Gastroenterology and Digestive Endoscopy Unit, “Morgagni-Pierantoni” Hospital, Forlì, Italy
- Gastroenterology and Digestive Endoscopy Unit, “Maurizio Bufalini” Hospital, Cesena, Italy
| | - Maria Caterina Pallotti
- Palliative Care Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giulia Miserocchi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Virginia Ghini
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sofia Zanuccoli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Cagnazzo
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
165
|
Lu Y, Lu C, Luo Z, Chen P, Xiong H, Li W. Association between the systemic inflammatory response index and mortality in patients with sarcopenia. PLoS One 2024; 19:e0312383. [PMID: 39556533 PMCID: PMC11573146 DOI: 10.1371/journal.pone.0312383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/06/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Sarcopenia is closely linked to inflammation; however, the association between the systemic inflammatory response index (SIRI) and mortality in patients with sarcopenia remains unclear. This study aims to explore the relationship between SIRI and mortality in sarcopenia patients. METHODS We analyzed data from ten cycles of the National Health and Nutrition Examination Survey (NHANES) spanning 1999 to 2018, selecting 3,141 sarcopenia patients. Mortality data were obtained from the National Death Index up to December 31, 2019. Participants were divided into three groups based on the ranking of their SIRI values. The association between SIRI and mortality was assessed using Cox proportional hazards models, with smooth curve fitting employed to test the correlation. Sensitivity analyses, subgroup analyses, and interaction tests were conducted to validate the stability of the findings. RESULTS A total of 101,316 individuals were included in this study. During a median follow-up of 10.4 years (minimum follow-up time of approximately 0.08 years, maximum follow-up time of 20.75 years), 667 participants died. Kaplan-Meier (KM) analysis indicated a higher risk of mortality in the SIRI Q3 group. Cox regression analysis showed a significant association between the SIRI Q3 group and all-cause mortality [HR 1.24 (95% CI: 1.05, 1.47)] and cardiovascular disease mortality [HR 1.46 (95% CI: 1.04, 2.04)]. Subgroup analysis revealed that SIRI was significantly associated with all-cause mortality across various demographic characteristics (e.g., gender, diabetes, hypertension, cardiovascular disease). Sensitivity analysis, excluding participants with cardiovascular disease, those who died within two years of follow-up, and those under 50 years old, indicated higher hazard ratios (HRs) for all-cause and cardiovascular mortality in the SIRI Q3 group. CONCLUSION This study demonstrates a significant association between SIRI and an increased risk of mortality in sarcopenia patients aged 20 years and older.
Collapse
Affiliation(s)
- Yifan Lu
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chengyin Lu
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhiqiang Luo
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Pei Chen
- The First Hospital of Hunan University Chinese Medicine, Changsha, Hunan, China
| | - Hui Xiong
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- The First Hospital of Hunan University Chinese Medicine, Changsha, Hunan, China
| | - Wangyang Li
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
166
|
Huang L, Liu P, Du Y, Bazan JF, Pan D, Chen Q, Lee A, Kola VSR, Wolfe SA, Wang YX. A brown fat-enriched adipokine, ASRA, is a leptin receptor antagonist that stimulates appetite. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.12.557454. [PMID: 37745491 PMCID: PMC10515849 DOI: 10.1101/2023.09.12.557454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The endocrine control of food intake remains incompletely understood, and whether the leptin receptor (LepR)-mediated anorexigenic pathway in the hypothalamus is negatively regulated by a humoral factor is unknown. Here, we identify an appetite-stimulating factor - ASRA - that represents a peripheral signal of energy deficit and orthosterically antagonizes LepR signaling. Asra encodes an 8 kD protein that is abundantly and selectively expressed in adipose tissue and to a lesser extent, in liver. ASRA associates with autophagy vesicles and its secretion is enhanced by energy deficiency. In vivo, fasting and cold stimulate Asra expression and increase its protein concentration in cerebrospinal fluid. Asra overexpression attenuates LepR signaling, leading to elevated blood glucose and development of severe hyperphagic obesity. Conversely, either adipose- or liver-specific Asra knockout mice display increased leptin sensitivity, improved glucose homeostasis, reduced food intake, resistance to high-fat diet-induced obesity, and blunted cold-evoked feeding response. Mechanistically, ASRA acts as a high affinity antagonist of LepR. AlphaFold2-multimer prediction and mutational studies suggest that a core segment of ASRA binds to the immunoglobin-like domain of LepR, similar to the 'site 3' recognition of the A-B loop of leptin. While administration of recombinant wild-type ASRA protein promotes food intake and increases blood glucose in a LepR signaling-dependent manner, point mutation within ASRA that disrupts LepR-binding results in a loss of these effects. Our studies reveal a previously unknown endocrine mechanism in appetite regulation and have important implications for our understanding of leptin resistance.
Collapse
Affiliation(s)
- Lei Huang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - Pengpeng Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - Yong Du
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - J Fernando Bazan
- Bioconsulting llc, Stillwater, MN, USA
- Unit of Structural Biology, VIB-UGent Center for Inflammation Research, Gent, Belgium
| | - Dongning Pan
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Present address: Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Qingbo Chen
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alexandra Lee
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vijaya Sudhakara Rao Kola
- Department of Medicine and Division of Hematology/Oncology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yong-Xu Wang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
167
|
Surov A, Pönisch W, Borggrefe J, Meyer HJ. CT-defined muscle density as a prognostic factor in multiple myeloma undergoing autologous stem cell therapy: a retrospective single center study. J Cancer Res Clin Oncol 2024; 150:499. [PMID: 39546043 PMCID: PMC11567988 DOI: 10.1007/s00432-024-06009-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE Skeletal muscle quality assessment can be performed by cross-sectional imaging. Skeletal muscle density (SMD) identified to be of prognostic relevance of several clinically outcomes in patients with hematological diseases. The purpose of the present study was to establish the effect of SMD on overall survival (OS) and progression-free survival (PFS) in patients with multiple myeloma (MM). METHODS All patients with MM were retrospectively analyzed between 2009 and 2019. 127 patients were included into the analysis. Whole-body computed tomography (CT) was used to calculate skeletal muscle index (SMI), SMD, albumin-gauge score and intramuscular adipose tissue content (IMAC). RESULTS Overall, 28 patients (22.0%) of the patient sample died. In the discrimination analysis muscle density was higher in non-survivors compared to survivors (mean 30.8 ± 12.5 versus 24.1 ± 15.8, p = 0.03) and IMAC was lower in non-survivors (- 0.66 ± 1.8 versus - 0.25 ± 0.21, p = 0.01). These differences, however, were not demonstrated in the logistic regression analysis, which could not show prognostic relevance for the investigated muscle density parameters on PFS or OS. CONCLUSION CT-defined muscle density parameters have no prognostic relevance on survival in patients with MM undergoing autologous stem cell therapy, which was demonstrated in a comprehensive analysis. These results corroborate previous smaller studies that body composition might have a limited role in this tumor entity.
Collapse
Affiliation(s)
- Alexey Surov
- Department of Radiology and Nuclear Medicine, Ruhr-University of Bochum, Muehlenkreiskliniken Minden, Bochum, Germany.
| | - Wolfram Pönisch
- Department of Hematology and Cell Therapy, University of Leipzig, Leipzig, Germany
| | - Jan Borggrefe
- Department of Radiology and Nuclear Medicine, Ruhr-University of Bochum, Muehlenkreiskliniken Minden, Bochum, Germany
| | - Hans-Jonas Meyer
- Department of Diagnostic and Interventional Radiology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
168
|
Mendes N, Zanesco A, Aguiar C, Rodrigues-Luiz GF, Silva D, Campos J, Camara NOS, Moraes-Vieira P, Araujo E, Velloso LA. CXCR3-expressing myeloid cells recruited to the hypothalamus protect against diet-induced body mass gain and metabolic dysfunction. eLife 2024; 13:RP95044. [PMID: 39535032 PMCID: PMC11560133 DOI: 10.7554/elife.95044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Microgliosis plays a critical role in diet-induced hypothalamic inflammation. A few hours after a high-fat diet (HFD), hypothalamic microglia shift to an inflammatory phenotype, and prolonged fat consumption leads to the recruitment of bone marrow-derived cells to the hypothalamus. However, the transcriptional signatures and functions of these cells remain unclear. Using dual-reporter mice, this study reveals that CX3CR1-positive microglia exhibit minimal changes in response to a HFD, while significant transcriptional differences emerge between microglia and CCR2-positive recruited myeloid cells, particularly affecting chemotaxis. These recruited cells also show sex-specific transcriptional differences impacting neurodegeneration and thermogenesis. The chemokine receptor CXCR3 is emphasized for its role in chemotaxis, displaying notable differences between recruited cells and resident microglia, requiring further investigation. Central immunoneutralization of CXCL10, a ligand for CXCR3, resulted in increased body mass and decreased energy expenditure, especially in females. Systemic chemical inhibition of CXCR3 led to significant metabolic changes, including increased body mass, reduced energy expenditure, elevated blood leptin, glucose intolerance, and decreased insulin levels. This study elucidates the transcriptional differences between hypothalamic microglia and CCR2-positive recruited myeloid cells in diet-induced inflammation and identifies CXCR3-expressing recruited immune cells as protective in metabolic outcomes linked to HFD consumption, establishing a new concept in obesity-related hypothalamic inflammation.
Collapse
Affiliation(s)
- Natalia Mendes
- School of Medical Sciences, Department of Translational Medicine (Section of Pharmacology), University of CampinasCampinasBrazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Ariane Zanesco
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Cristhiane Aguiar
- Laboratory of Immunometabolism, Institute of Biology - University of Campinas, BrazilCampinasBrazil
| | - Gabriela F Rodrigues-Luiz
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa CatarinaFlorianópolisBrazil
| | - Dayana Silva
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Jonathan Campos
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Niels Olsen Saraiva Camara
- Laboratory for Transplantation Immunobiology, Institute of Biomedical Sciences, University of Sao PauloSao PauloBrazil
| | - Pedro Moraes-Vieira
- Laboratory of Immunometabolism, Institute of Biology - University of Campinas, BrazilCampinasBrazil
| | - Eliana Araujo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
- Faculty of Nursing, University of CampinasCampinasBrazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
- National Institute of Science and Technology on NeuroimmunomodulationRio de JaneiroBrazil
| |
Collapse
|
169
|
Azimzadeh M, Cheah PS, Ling KH. Brain insulin resistance in Down syndrome: Involvement of PI3K-Akt/mTOR axis in early-onset of Alzheimer's disease and its potential as a therapeutic target. Biochem Biophys Res Commun 2024; 733:150713. [PMID: 39307112 DOI: 10.1016/j.bbrc.2024.150713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/27/2024] [Accepted: 09/16/2024] [Indexed: 10/06/2024]
Abstract
Down syndrome (DS) is the most common genetic cause of intellectual impairment, characterised by an extra copy of chromosome 21. After the age of 40, DS individuals are highly susceptible to accelerated ageing and the development of early-onset Alzheimer-like neuropathology. In the context of DS, the brain presents a spectrum of neuropathological mechanisms and metabolic anomalies. These include heightened desensitisation of brain insulin and insulin-like growth factor-1 (IGF-1) reactions, compromised mitochondrial functionality, escalated oxidative stress, reduced autophagy, and the accumulation of amyloid beta and tau phosphorylation. These multifaceted factors intertwine to shape the intricate landscape of DS-related brain pathology. Altered brain insulin signalling is linked to Alzheimer's disease (AD). This disruption may stem from anomalies in the extracellular aspect (insulin receptor) or the intracellular facet, involving the inhibition of insulin receptor substrate 1 (IRS1). Both domains contribute to the intricate mechanism underlying this dysregulation. The PI3K-Akt/mammalian target of the rapamycin (mTOR) axis is a crucial intracellular element of the insulin signalling pathway that connects numerous physiological processes in the cell cycle. In age-related neurodegenerative disorders like AD, aberrant modulation of the PI3K-Akt signalling cascade is a key factor contributing to their onset. Aberrant and sustained hyperactivation of the PI3K/Akt-mTOR axis in the DS brain is implicated in early symptoms of AD development. Targeting the PI3K-Akt/mTOR pathway may help delay the onset of early-onset AD in individuals with DS, offering a potential way to slow disease progression and enhance their quality of life.
Collapse
Affiliation(s)
- Mansour Azimzadeh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Malaysian Research Institute on Ageing (MyAgeing®), Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Malaysian Research Institute on Ageing (MyAgeing®), Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
170
|
Bombassaro B, Araujo EP, Velloso LA. The hypothalamus as the central regulator of energy balance and its impact on current and future obesity treatments. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240082. [PMID: 39876968 PMCID: PMC11771753 DOI: 10.20945/2359-4292-2024-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/10/2024] [Indexed: 01/31/2025]
Abstract
The hypothalamus is a master regulator of energy balance in the body. First-order hypothalamic neurons localized in the arcuate nucleus sense systemic signals that indicate the energy stores in the body. Through distinct projections, arcuate nucleus neurons communicate with second-order neurons, which are mostly localized in the paraventricular nucleus and in the lateral hypothalamus. The signals then proceed to third- and fourth-order neurons that activate complex responses aimed at maintaining whole-body energy homeostasis. During the last 30 years, since the identification of leptin in 1994, there has been a great advance in the unveiling of the hypothalamic and extra-hypothalamic neuronal networks that control energy balance. This has contributed to the characterization of the mechanisms by which glucagon-like peptide-1 receptor agonists promote body mass reduction and has opened new windows of opportunity for the development of drugs to treat obesity. This review presents an overview of the mechanisms involved in the hypothalamic regulation of energy balance and discusses how advancements in this field are contributing to the development of new pharmacological strategies to treat obesity.
Collapse
Affiliation(s)
- Bruna Bombassaro
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Eliana P Araujo
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Licio A Velloso
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| |
Collapse
|
171
|
Lima H, Etchebehere M, Bogoni M, Torricelli C, Nogueira-Lima E, Deflon VM, Lima M, Etchebehere E. Theranostics Nuclear Medicine in Prostate Cancer. Pharmaceuticals (Basel) 2024; 17:1483. [PMID: 39598394 PMCID: PMC11597825 DOI: 10.3390/ph17111483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024] Open
Abstract
Theranostic Nuclear Medicine is based on the idea of combining the same molecule (or drug) with different radioisotopes for both diagnosis and treatment, a concept that emerged in the early 1940s with the use of radioactive iodine for thyroid diseases. Theranostic Nuclear Medicine has since expanded to diseases of higher incidence, such as prostate cancer, with several imaging methods used to assess the extent of the disease and the corresponding radiopharmaceuticals used for treatment. For example, by detecting osteoblastic metastases by bone scintigraphy, corresponding radiopharmaceuticals with therapeutic properties can be administered to eliminate or reduce pain associated with metastases and/or determine overall survival gain. The purpose of this review is to discuss the role of Theranostic Nuclear Medicine in prostate cancer, addressing the main diagnostic imaging studies with their corresponding treatments in the Theranostic model.
Collapse
Affiliation(s)
- Helena Lima
- Faculdade de Medicina, Pontifícia Universidade Católica de Campinas (PUCC), Campinas 13087-571, Brazil;
| | - Marina Etchebehere
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo 05652-000, Brazil;
| | - Mateos Bogoni
- Hospital Erasto Gaertner, Curitiba 81520-060, Brazil;
- Diagnóstico Avançado por Imagem (DAPI), Curitiba 80430-210, Brazil
| | - Caroline Torricelli
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas 13083-888, Brazil; (C.T.); (E.N.-L.); (M.L.)
| | - Ellen Nogueira-Lima
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas 13083-888, Brazil; (C.T.); (E.N.-L.); (M.L.)
| | - Victor M. Deflon
- São Carlos Institute of Chemistry, University of São Paulo (USP), São Carlos 13566-590, Brazil;
| | - Mariana Lima
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas 13083-888, Brazil; (C.T.); (E.N.-L.); (M.L.)
- Medicina Nuclear de Campinas (Grupo MND), Campinas 13020-432, Brazil
| | - Elba Etchebehere
- Division of Nuclear Medicine, University of Campinas (UNICAMP), Campinas 13083-888, Brazil; (C.T.); (E.N.-L.); (M.L.)
- Medicina Nuclear de Campinas (Grupo MND), Campinas 13020-432, Brazil
| |
Collapse
|
172
|
Kim DH, Lee MJ, Kang D, Lee JY, Park S, Khang AR, Bae JH, Kim JY, Kim SH, Kang YH, Yi D. Empagliflozin Attenuates High-Glucose-Induced Astrocyte Activation and Inflammation via NF-κB Pathway. Curr Issues Mol Biol 2024; 46:12417-12427. [PMID: 39590331 PMCID: PMC11592667 DOI: 10.3390/cimb46110737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibitors regulate blood glucose levels in patients with type 2 diabetes mellitus and may also exert anti-inflammatory and anti-atherosclerotic effects by promoting M2 macrophage polarization. Although SGLT2 is expressed in brain regions that influence glucose balance and cognitive function, its roles in the central nervous system are unclear. This study investigated the effects of empagliflozin (EMPA), an SGLT2 inhibitor, on hypothalamic inflammation associated with metabolic diseases. Mice were subjected to a high-fat diet (HFD) for varying durations (3 d, 3 weeks, and 16 weeks) and treated with EMPA for 3 weeks (NFD, NFD + EMPA, HFD, HFD + EMPA; n = 5/group). EMPA regulated the expression of astrocyte markers and pro-inflammatory cytokine mRNA in the hypothalamus of HFD-induced mice, which was linked to regulation of the NF-κB pathway. Under hyperglycemic conditions, EMPA may mitigate hypothalamic inflammation by modulating astrocyte activation via the NF-κB pathway. Our findings demonstrated that EMPA possesses therapeutic potential beyond merely lowering blood glucose levels, opening new avenues for addressing inflammation and providing neuroprotection in metabolic disease management.
Collapse
Affiliation(s)
- Dong Hee Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
| | - Min Jin Lee
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Dasol Kang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
| | - Ji Young Lee
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
| | - Sujin Park
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
| | - Ah Reum Khang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Ji Hyun Bae
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Joo Yeon Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Su Hyun Kim
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Yang Ho Kang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| | - Dongwon Yi
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50621, Republic of Korea; (D.H.K.); (M.J.L.); (D.K.); (J.Y.L.); (S.P.); (A.R.K.); (J.H.B.); (J.Y.K.); (S.H.K.); (Y.H.K.)
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50621, Republic of Korea
| |
Collapse
|
173
|
Xie R, Luo Y, Bao B, Wu X, Guo J, Wang J, Qu X, Che X, Zheng C. The Role of Fatty Acid Metabolism, the Related Potential Biomarkers, and Targeted Therapeutic Strategies in Gastrointestinal Cancers. Drug Dev Res 2024; 85:e70014. [PMID: 39527665 DOI: 10.1002/ddr.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/12/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Gastrointestinal cancer has emerged as a significant global health concern due to its high incidence and mortality, limited effectiveness of early detection, suboptimal treatment outcomes, and poor prognosis. Metabolic reprogramming is a prominent feature of cancer, and fatty acid metabolism assumes a pivotal role in bridging glucose metabolism and lipid metabolism. Fatty acids play important roles in cellular structural composition, energy supply, signal transduction, and other lipid-related processes. Changes in the levels of fatty acid metabolite may indicate the malignant transformation of gastrointestinal cells, which have an impact on the prognosis of patients and can be used as a marker to monitor the efficacy of anticancer therapy. Therefore, targeting key enzymes involved in fatty acid metabolism, either as monotherapy or in combination with other agents, is a promising strategy for anticancer treatment. This article reviews the potential mechanisms of fatty acid metabolism disorders in the occurrence and development of gastrointestinal tumors, and summarizes the related potential biomarkers and anticancer strategies.
Collapse
Affiliation(s)
- Ruixi Xie
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Luo
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bowen Bao
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinshu Wu
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jia Guo
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jin Wang
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaofang Che
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chunlei Zheng
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Oncology, Shanghai Electric Power Hospital, Shanghai, China
| |
Collapse
|
174
|
Yang N, Zhou P, Lyu J, Ren J, Nie X, Zhao S, Ye Y. Prognostic value of sarcopenia and myosteatosis alterations on survival outcomes for esophageal squamous cell carcinoma before and after radiotherapy. Nutrition 2024; 127:112536. [PMID: 39182329 DOI: 10.1016/j.nut.2024.112536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVE We assessed the impact and prognostic significance of alterations in muscle quality and quantity (myosteatosis and sarcopenia, respectively) in patients with esophageal cancer treated with radiotherapy (RT). METHODS We retrospectively pooled 258 patients with esophageal squamous cell cancer who underwent RT. Myosteatosis and sarcopenia were determined based on the skeletal muscle index derived from the muscle area and attenuation at the L3 level from computed tomography images. Subgroups were formed as 2 subgroups of non-sarcopenia/myosteatosis and sarcopenia/myosteatosis (with or without other muscle status) at either timepoint of RT, 3 subgroups of only-sarcopenia, only myosteatosis (without other muscle status), and the co-presence of sarcopenia and myosteatosis at either timepoint of RT, as well as 4 subgroups of continuous sarcopenia/myosteatosis, developed sarcopenia/myosteatosis, reduced sarcopenia/myosteatosis and non-sarcopenia/myosteatosis according to alterations of muscle status at both timepoints of RT. Overall survival (OS) was compared. Univariate and multivariate analyses based on Cox regression identified independent risk factors for prognosis. RESULTS Either pre- or post-RT, patients with sarcopenia and myosteatosis (with or without other muscle status) had poor OS. Patients with only myosteatosis (without other muscle status) showed the best OS (1352 days pre-RT vs. 1648 days post-RT), while patients with concurrent myosteatosis and sarcopenia had the worst OS (907 days pre-RT vs. 706 days post-RT). The ascending order of OS for sarcopenia alterations was as follows: continuous sarcopenia (1093 days), non-sarcopenia (1740 days), developed sarcopenia (2187 days), and reduced sarcopenia (2208 days) (P = 0.002). The ascending order of OS for myosteatosis alterations was ranked as follows: continuous myosteatosis (1165 days), reduced myosteatosis (1275 days), developed myosteatosis (1783 days), and non-myosteatosis (1942 days) (P = 0.061). Univariate and multivariate Cox regression analyses revealed that increased age, longer tumor length, developed myosteatosis, and continuous myosteatosis were independent prognostic factors for OS. CONCLUSIONS Muscle mass status at presentation and alterations in patients with esophageal cancer before and after RT should be considered prognostic indicators.
Collapse
Affiliation(s)
- Ningjing Yang
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Zhou
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Jiahua Lyu
- Department of Radiotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Ren
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinyu Nie
- Department of Radiotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Sijia Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yihong Ye
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
175
|
Connolly BJ, Saxton SN. Recent updates on the influence of iron and magnesium on vascular, renal, and adipose inflammation and possible consequences for hypertension. J Hypertens 2024; 42:1848-1861. [PMID: 39258532 PMCID: PMC11451934 DOI: 10.1097/hjh.0000000000003829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
The inflammatory status of the kidneys, vasculature, and perivascular adipose tissue (PVAT) has a significant influence on blood pressure and hypertension. Numerous micronutrients play an influential role in hypertension-driving inflammatory processes, and recent reports have provided bases for potential targeted modulation of these micronutrients to reduce hypertension. Iron overload in adipose tissue macrophages and adipocytes engenders an inflammatory environment and may contribute to impaired anticontractile signalling, and thus a treatment such as chelation therapy may hold a key to reducing blood pressure. Similarly, magnesium intake has proven to greatly influence inflammatory signalling and concurrent hypertension in both healthy animals and in a model for chronic kidney disease, demonstrating its potential clinical utility. These findings highlight the importance of further research to determine the efficacy of micronutrient-targeted treatments for the amelioration of hypertension and their potential translation into clinical application.
Collapse
Affiliation(s)
- Benjamin J Connolly
- Divison of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | | |
Collapse
|
176
|
Malani K, Pradhan S, Madani MM, Roberts MB, Shadyab A, Allison M, Brasky TM, Schnatz PF, Snetselaar L, Eaton CB. Association of Diet Quality With Risk of Incident Rheumatoid Arthritis in the Women's Health Initiative. J Acad Nutr Diet 2024; 124:1451-1473. [PMID: 39025233 DOI: 10.1016/j.jand.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/03/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Various foods and nutrients are linked with higher or lower risk of rheumatoid arthritis (RA), yet these associations are inconsistent across studies. Limited research has been done evaluating the association between diet quality and RA in a larger-scale prospective study on postmenopausal women. OBJECTIVE The objective of this study was to evaluate the association between dietary quality and risk of incident RA in postmenopausal women. DESIGN This was a prospective cohort study as part of the Women's Health Initiative (WHI), with an average follow-up time of 8.1 years. Baseline diet was measured using a food frequency questionnaire (FFQ). Diet quality was evaluated by the Healthy Eating Index (HEI)-2015 total score. In addition, intake of food groups and nutrients that align with HEI-2015 components was assessed. PARTICIPANTS/SETTING Postmenopausal women (N = 109 591) were included in this study, which was conducted at various clinical centers across the United States with recruitment from 1993 to 1998. Women's Health Initiative participants who were missing outcome data, had unreliable/missing FFQ data, or had RA at baseline were excluded. MAIN OUTCOME MEASURES The primary outcome measure was incident RA. Statistical analyses performed Multivariable Cox proportional regression analysis was performed evaluating the association of diet quality with self-reported physician-diagnosed RA after adjusting for age, race, ethnicity, education status, income, and body mass index (BMI). RESULTS During 857 517 person-years of follow-up, 5823 incident RA cases were identified. After adjustment for multiple comparisons, compared with quartile 1, quartiles 2, 3, and 4 of the HEI-2015 total scores were associated with lower RA risks of 1%, 10%, and 19%, respectively (P-trend < .001). Greater consumption of total fruits (P-trend = .014), whole fruits (P-trend < .0002), total vegetables (P-trend = .008), greens and beans (P-trend < .0002), whole grains (P-trend = .008), and dairy (P-trend = .018) were significantly associated with lower rates of incident RA. Conversely, higher consumption of saturated fat (P-trend = .002) was significantly associated with higher rates of incident RA. CONCLUSION A higher-quality diet reflected by higher HEI-2015 total scores was inversely associated with incident RA in postmenopausal women.
Collapse
Affiliation(s)
- Kanika Malani
- Warren Alpert School of Medicine at Brown University, Providence, RI.
| | - Sushaili Pradhan
- Brown University Center for Primary Care and Prevention, Pawtucket, RI
| | | | - Mary B Roberts
- Brown University Center for Primary Care and Prevention, Pawtucket, RI
| | - Aladdin Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA
| | - Matthew Allison
- University of California San Diego School of Medicine, San Diego, CA
| | | | - Peter F Schnatz
- Sidney Kimmel Medical College at Thomas Jefferson University, West Reading, PA
| | | | - Charles B Eaton
- Brown University Center for Primary Care and Prevention, Pawtucket, RI; Department of Epidemiology, Brown University School of Public Health, Pawtucket, RI; Department of Family Medicine, Warren Alpert Medical School of Brown University, Pawtucket, RI
| |
Collapse
|
177
|
Chen X, Song Y, Hong Y, Zhang X, Li Q, Zhou H. "NO" controversy?: A controversial role in insulin signaling of diabetic encephalopathy. Mol Cell Endocrinol 2024; 593:112346. [PMID: 39151653 DOI: 10.1016/j.mce.2024.112346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/14/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Insulin, a critical hormone in the human body, exerts its effects by binding to insulin receptors and regulating various cellular processes. While nitric oxide (NO) plays an important role in insulin secretion and acts as a mediator in the signal transduction pathway between upstream molecules and downstream effectors, holds a significant position in the downstream signal network of insulin. Researches have shown that the insulin-NO system exhibits a dual regulatory effect within the central nervous system, which is crucial in the regulation of diabetic encephalopathy (DE). Understanding this system holds immense practical importance in comprehending the targets of existing drugs and the development of potential therapeutic interventions. This review extensively examines the characterization of insulin, NO, Nitric oxide synthase (NOS), specific NO pathway, their interconnections, and the mechanisms underlying their regulatory effects in DE, providing a reference for new therapeutic targets of DE.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ying Song
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China; Hangzhou King's Bio-pharmaceutical Technology Co., Ltd, Hangzhou, Zhejiang, 310007, China.
| | - Ye Hong
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xiaomin Zhang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qisong Li
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Hongling Zhou
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
178
|
Grandhi N, Liu L, Wang M, Thomas T, Schoen M, Sanfilippo K, Gao F, Colditz GA, Carson KR, Janakiram M, Chang SH. Association between glucagon-like peptide-1 receptor agonist use and progression of monoclonal gammopathy of uncertain significance to multiple myeloma among patients with diabetes. JNCI Cancer Spectr 2024; 8:pkae095. [PMID: 39514091 PMCID: PMC11643351 DOI: 10.1093/jncics/pkae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/26/2024] [Accepted: 09/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND In patients with diabetes and monoclonal gammopathy of uncertain significance (MGUS), the impact of glucagon-like peptide-1 (GLP-1) receptor agonists on the natural history of MGUS is unknown. We aimed to assess the association of GLP-1 receptor agonist use in the progression of MGUS to multiple myeloma in patients with diabetes. METHODS This is a population-based cohort study of veterans diagnosed with MGUS from 2006 to 2021 with a prior diagnosis of diabetes. A validated natural language processing algorithm was used to confirm MGUS and progression to multiple myeloma. We performed 1:2 matching for individuals with and without GLP-1 receptor agonist exposure. The Gray test was performed to detect the difference in cumulative incidence functions for progression by GLP-1 receptor agonist use status. The association between time-varying GLP-1 receptor agonist use and progression was estimated through multivariable-adjusted hazard ratio using a stratified Fine-Gray distribution hazard model, with death as a competing event and stratum for the matched patient triad. RESULTS Our matched cohort included 1097 individuals with MGUS who had ever used GLP-1 receptor agonists and the matched 2194 patients who had never used GLP-1 receptor agonists. Overall, 2.6% of individuals progressed in the GLP-1 receptor agonist ever use group compared with 5.0% in the GLP-1 receptor agonist never use group. Cumulative incidence functions were statistically significantly different between the exposed and unexposed groups (P = .02). GLP-1 receptor agonist use vs no use was associated with decreased progression to multiple myeloma (hazard ratio = 0.45, 95% confidence interval = 0.22 to 0.93, P = .03). CONCLUSIONS For patients with diabetes and MGUS, GLP-1 receptor agonist use is associated with a 55% reduction in risk of progression from MGUS to multiple myeloma compared with no use.
Collapse
Affiliation(s)
- Nikhil Grandhi
- Research Service, St Louis VA Medical Center, St Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Lawrence Liu
- Research Service, St Louis VA Medical Center, St Louis, MO, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Mei Wang
- Research Service, St Louis VA Medical Center, St Louis, MO, USA
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Theodore Thomas
- Research Service, St Louis VA Medical Center, St Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Martin Schoen
- Research Service, St Louis VA Medical Center, St Louis, MO, USA
- Department of Medicine, Saint Louis University School of Medicine, St Louis, MO, USA
| | - Kristen Sanfilippo
- Research Service, St Louis VA Medical Center, St Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Graham A Colditz
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Kenneth R Carson
- Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Su-Hsin Chang
- Research Service, St Louis VA Medical Center, St Louis, MO, USA
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
179
|
Chiu PE, Fu Z, Tsai YC, Tsai CY, Hsu WJ, Chou LW, Lai DW. Fu's subcutaneous needling promotes axonal regeneration and remyelination by inhibiting inflammation and endoplasmic reticulum stress. Transl Res 2024; 273:46-57. [PMID: 38950695 DOI: 10.1016/j.trsl.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 05/03/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Fu's subcutaneous needling (FSN) is a traditional Chinese acupuncture procedure used to treat pain-related neurological disorders. Moreover, the regulation of inflammatory cytokines may provide a favorable environment for peripheral nerve regeneration. In light of this, FSN may be an important novel therapeutic strategy to alleviate pain associated with peripheral neuropathy; however, the underlying molecular mechanisms remain unclear. This study revealed that patients who had osteoarthritis with peripheral neuropathic pain significantly recovered after 1 to 2 weeks of FSN treatment according to the visual analog scale, Western Ontario and McMaster Universities Osteoarthritis Index, Lequesne index, walking speed, and passive range of motion. Similarly, we demonstrated that FSN treatment in an animal model of chronic constriction injury (CCI) significantly improved sciatic nerve pain using paw withdrawal thresholds, sciatic functional index scores, and compound muscle action potential amplitude tests. In addition, transmission electron microscopy images of sciatic nerve tissue showed that FSN effectively reduced axonal swelling, abnormal myelin sheaths, and the number of organelle vacuoles in CCI-induced animals. Mechanistically, RNA sequencing and gene set enrichment analysis revealed significantly reduced inflammatory pathways, neurotransmitters, and endoplasmic reticulum stress pathways and increased nerve regeneration factors in the FSN+CCI group, compared with that in the CCI group. Finally, immunohistochemistry, immunoblotting and enzyme-linked immunosorbent assay showed similar results in the dorsal root ganglia and sciatic nerve. Our findings suggest that FSN can effectively ameliorate peripheral neuropathic pain by regulate inflammation and endoplasmic reticulum stress, thereby determine its beneficial application in patients with peripheral nerve injuries.
Collapse
Affiliation(s)
- Po-En Chiu
- Department of Chinese Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Zhonghua Fu
- Institute of Fu's Subcutaneous Needling, Beijing University of Chinese Medicine, Beijing, China; Clinical Medical College of Acupuncture & Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi-Ching Tsai
- Department of Immune Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Chia-Yun Tsai
- Experimental Animal Center, Department of Molecular Biology and Cell Research, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Wei-Jen Hsu
- Experimental Animal Center, Department of Molecular Biology and Cell Research, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Li-Wei Chou
- Department of Physical Medicine and Rehabilitation, China Medical University Hospital, Taichung, Taiwan; Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan; Department of Physical Medicine and Rehabilitation, Asia University Hospital, Asia University, Taichung, Taiwan.
| | - De-Wei Lai
- Experimental Animal Center, Department of Molecular Biology and Cell Research, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan; Department of Pharmacy and Master Program, Tajen University, Pingtung, Taiwan; Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.
| |
Collapse
|
180
|
Fragkiadaki V, Panagiotidis E, Vlontzou E, Kalathas T, Paschali A, Kypraios C, Chatzipavlidou V, Datseris I. Correlation of PSA blood levels with standard uptake value maximum (SUV max ) and total metabolic tumor volume (TMTV) in 18F-PSMA-1007 and 18F-choline PET/CT in patients with biochemically recurrent prostate cancer. Nucl Med Commun 2024; 45:924-930. [PMID: 39082074 DOI: 10.1097/mnm.0000000000001881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
OBJECTIVES In this prospective study, we investigated the correlation between prostate-specific antigen (PSA) levels in the blood of patients with prostate cancer in biochemical recurrence after radical treatment with the semiquantitative parameters standard uptake value maximum (SUV max ) and the total metabolic tumor volume (TMTV) in the metastatic foci depicted in 18F-prostate-specific membrane antigen (PSMA)-1007 and 18F-choline PET/computed tomography (CT) imaging. METHODS We prospectively examined 104 patients with biochemical relapse of prostate cancer after primary definitive treatment. All patients underwent one 18F-PSMA-1007 and one 18F-choline PET/CT examination in randomized order within a time frame of 10 days and were followed for at least 6 months (182 ± 10 days). The semiquantitative parameters of SUV max and metabolic tumor volume (MTV) of each neoplastic lesion in PET/CT imaging were calculated, and further summation of each MTV value was done to calculate the TMTV. RESULTS According to the Spearman correlation analysis, a positive correlation was found between PSA levels and SUV max and TMTV scores in the metastatic foci of 18F-PSMA-1007 PET/CT ( r = 0.24 and 0.35, respectively; P < 0.05) and SUV max in the lesions of 18F-choline PET/CT ( r = 0.28; P < 0.0239). However, a positive but NS correlation was demonstrated between values of PSA and TMTV for each lesion in the 18F-choline PET/CT study ( r = 0.22; P = 0.0795). The detection rate of the different PSA levels with a cutoff of 1 ng/ml was higher for 18F-PSMA-1007 than 18F-choline. CONCLUSION In biochemical relapse patients there is a positive correlation between PSA levels in the blood and the semiquantitative parameters SUV max and TMTV of the metastatic foci in the 18F-PSMA-1007 and 18F-Choline PET/CT imaging.
Collapse
Affiliation(s)
| | | | - Evaggelia Vlontzou
- Department of Nuclear Medicine, Evaggelismos General Hospital, Athens and
| | | | - Anna Paschali
- Department of Nuclear Medicine, Theageneio Cancer Center, Thessaloniki,
| | | | | | - Ioannis Datseris
- Department of Nuclear Medicine, Evaggelismos General Hospital, Athens and
| |
Collapse
|
181
|
Xiang Y, Shen L, Xue Y, Wang Z, Zhou R, Cao Y, Zhu Z, Xu P, Yu X, Fang P, Shang W. Efficacy and safety of diacerein monotherapy in adults with obesity: A randomized, double-blind, placebo-controlled trial. Diabetes Obes Metab 2024; 26:5293-5303. [PMID: 39192530 DOI: 10.1111/dom.15881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024]
Abstract
AIM To assess the efficacy and safety of diacerein monotherapy in adults with obesity. METHODS Forty-two adults with obesity participated in the study and were randomly assigned to receive diacerein or placebo in addition to lifestyle modification for 14 weeks, in a double-blinded fashion. Differences in changes in body weight, body composition, metabolic variables, fatty liver-related indicators, cardiovascular system variables, lifestyle score and metabolic factors were compared. RESULTS Post-treatment weight loss percentage from baseline was -6.56% (-8.71%, -4.41%) in the diacerein group and -0.59% (-2.74%, 1.56%) in the placebo group. Compared with the placebo group, the diacerein group showed significant improvements in body composition, metabolic variables and indicators related to fatty liver. In addition, after 14 weeks of treatment, diacerein led to a significant reduction in serum visfatin concentration versus the placebo group. The reductions in total body fat mass and visceral fat area mediated the weight loss induced by diacerein. No significant differences were found between the groups in the number of adverse events and safety variables. CONCLUSIONS For adults with obesity, diacerein led to a clinically meaningful weight loss and provided multiple metabolic benefits with acceptable safety. These results support that diacerein is a promising candidate medicine to be developed for obesity management.
Collapse
Affiliation(s)
- Yingying Xiang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lixuan Shen
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yingying Xue
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziwei Wang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruonan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Cao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziwei Zhu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pingyuan Xu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xizhong Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenbin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
182
|
Khan H, Naseem T, Kaushik P, Narang J, Khan R, Panwar S, Parvez S. Decoding paradoxical links of cytokine markers in cognition: Cross talk between physiology, inflammaging, and Alzheimer's disease- related cognitive decline. Ageing Res Rev 2024; 101:102535. [PMID: 39374831 DOI: 10.1016/j.arr.2024.102535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Recent research has revolutionized our understanding of memory consolidation by emphasizing the critical role of astrocytes, microglia, and immune cells in through cytokine signaling. Cytokines, compact proteins, play pivotal roles in neuronal development, synaptic transmission, and normal aging. This review explores the cellular mechanisms contributing to cognitive decline in inflammaging and Alzheimer's disease, highlighting the paradoxical effects of most studied cytokines (IL-1, IL-6, TNF-α) in brain function, which act as a double-edged sword in brain physiology, acting both as facilitators of healthy cognitive function and as a potential contributor to cognitive decline.
Collapse
Affiliation(s)
- Hiba Khan
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Talib Naseem
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Jagriti Narang
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Siddharth Panwar
- School of Computing and Electrical Engineering, Indian Institute of Technology, Mandi, Himachal Pradesh 175075, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
183
|
Lin WL, Nguyen THY, Huang WT, Guo HR, Wu LM. Sarcopenia and survival in colorectal cancer without distant metastasis: a systematic review and meta-analysis. J Gastroenterol Hepatol 2024; 39:2250-2259. [PMID: 38986533 DOI: 10.1111/jgh.16681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND AND AIM Despite prior attempts to evaluate the effects of sarcopenia on survival among patients with colorectal cancer (CRC), the results of these studies have not been consistent. The present study aimed to evaluate the association between sarcopenia and survival among patients having CRC without distant metastasis by aggregating multiple studies. METHODS We performed a literature search using computerized databases and identified additional studies from among the bibliographies of retrieved articles. The quality of each study was evaluated using the Newcastle-Ottawa Scale, and meta-analyses were performed to evaluate overall survival (OS) and disease-free survival (DFS). RESULTS Thirteen studies with up to 6600 participants were included in the meta-analyses, with a mean age of 63.6 years (range: 18-93 years). We found that preoperative sarcopenia was associated with worse OS (hazard ratio [HR]: 1.61; 95% confidence interval [CI]: 1.38-1.88) and worse DFS (HR: 1.57; 95% CI: 1.10-2.24). Compared with patients without sarcopenia after tumor resection, those with postoperative sarcopenia had worse OS (HR: 1.76; 95% CI: 1.47-2.10) and DFS (HR: 1.79; 95% CI: 1.46-2.20). CONCLUSION These meta-analyses suggest that sarcopenia, no matter observed before or after tumor resection, is associated with worse OS and DFS in patients with CRC who have no distant metastasis.
Collapse
Affiliation(s)
- Wen-Li Lin
- Center for Quality Management, Chi Mei Medical Center, Tainan City, Taiwan
- School of Nursing, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Thi-Hoang-Yen Nguyen
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan City, Taiwan
| | - Wen-Tsung Huang
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan City, Taiwan
| | - How-Ran Guo
- Department of Environmental and Occupational Health, National Cheng Kung University, Tainan City, Taiwan
- Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Li-Min Wu
- School of Nursing, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung City, Taiwan
| |
Collapse
|
184
|
Li Q, Tong Y, Chen J, Xie T. Targeting programmed cell death via active ingredients from natural plants: a promising approach to cancer therapy. Front Pharmacol 2024; 15:1491802. [PMID: 39584140 PMCID: PMC11582395 DOI: 10.3389/fphar.2024.1491802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/09/2024] [Indexed: 11/26/2024] Open
Abstract
Cancer is a serious public health problem in humans, and prevention and control strategies are still necessary. Therefore, the development of new therapeutic drugs is urgently needed. Targeting programmed cell death, particularly via the induction of cancer cell apoptosis, is one of the cancer treatment approaches employed. Recently, an increasing number of studies have shown that compounds from natural plants can target programmed cell death and kill cancer cells, laying the groundwork for use in future anticancer treatments. In this review, we focus on the latest research progress on the role and mechanism of natural plant active ingredients in different forms of programmed cell death, such as apoptosis, autophagy, necroptosis, ferroptosis, and pyroptosis, to provide a strong theoretical basis for the clinical development of antitumor drugs.
Collapse
Affiliation(s)
- Qian Li
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yan Tong
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jianxiang Chen
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Tian Xie
- School of Pharmacy and Department of Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
185
|
Spinedi E, Docena GH. Physiopathological Roles of White Adiposity and Gut Functions in Neuroinflammation. Int J Mol Sci 2024; 25:11741. [PMID: 39519291 PMCID: PMC11546880 DOI: 10.3390/ijms252111741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
White adipose tissue (WAT) and the gut are involved in the development of neuroinflammation when an organism detects any kind of injury, thereby triggering metainflammation. In fact, the autonomous nervous system innervates both tissues, although the complex role played by the integrated sympathetic, parasympathetic, and enteric nervous system functions have not been fully elucidated. Our aims were to investigate the participation of inflamed WAT and the gut in neuroinflammation. Firstly, we conducted an analysis into how inflamed peripheral WAT plays a key role in the triggering of metainflammation. Indeed, this included the impact of the development of local insulin resistance and its metabolic consequences, a serious hypothalamic dysfunction that promotes neurodegeneration. Then, we analyzed the gut-brain axis dysfunction involved in neuroinflammation by examining cell interactions, soluble factors, the sensing of microbes, and the role of dysbiosis-related mechanisms (intestinal microbiota and mucosal barriers) affecting brain functions. Finally, we targeted the physiological crosstalk between cells of the brain-WAT-gut axis that restores normal tissue homeostasis after injury. We concluded the following: because any injury can result not only in overall insulin resistance and dysbiosis, which in turn can impact upon the brain, but that a high-risk of the development of neuroinflammation-induced neurodegenerative disorder can also be triggered. Thus, it is imperative to avoid early metainflammation by applying appropriate preventive (e.g., lifestyle and diet) or pharmacological treatments to cope with allostasis and thus promote health homeostasis.
Collapse
Affiliation(s)
- Eduardo Spinedi
- Centro de Endocrinología Experimental y Aplicada (CENEXA-UNLP-CONICET-CICPBA), University of La Plata Medical School, La Plata 1900, Argentina
| | - Guillermo Horacio Docena
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP-UNLP-CONICET-CICPBA), School of Sciences, University of La Plata, La Plata 1900, Argentina
| |
Collapse
|
186
|
Yang Y, Liang W, Fan K, Yang T, Cheng J. Clinical features of pituitary carcinoma: analysis based on a case report and literature review. Front Endocrinol (Lausanne) 2024; 15:1440247. [PMID: 39544231 PMCID: PMC11560426 DOI: 10.3389/fendo.2024.1440247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Introduction Pituitary carcinoma (PC) is an extremely rare tumor of the adenohypophysis, which manifests as craniospinal dissemination and/or systemic metastasis. The diagnosis of PC is particularly difficult, as the clinical diagnosis only can be made after the metastasis is found. Owing to the complex diagnostic process and less effective treatments, the clinical prognosis of PC is usually very poor. Hence, it is of great significance to illustrate the diagnosis and treatment course of PC. Methods In this case report, we described a 48-year-old male patient who was diagnosed with pituitary adenoma (PA) initially and then was diagnosed with PC eventually after spinal cord metastasis was found, and we illustrated the treatment course as well. Furthermore, we summarized all the published case reports until now and provided a comprehensive review of the diagnosis, treatment, prediction, and clinical outcome of PC. Results and Conclusions We found that most PC patients had adrenocorticotropic hormone/prolactin (ACTH/PRL)-secreting tumors, Ki-67 ≥ 10%, and P53 positivity, which may have the potential to predict the transformation from PA to PC; surgery excision combined with temozolomide (TMZ) and radiotherapy is helpful to prolong the survival of PC patients.
Collapse
Affiliation(s)
- Yongxiang Yang
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Wanlin Liang
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
- School of Clinical Medicine &The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Kexia Fan
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Tao Yang
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Jingmin Cheng
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
187
|
Zhang Y, Wang R, Liu T, Wang R. Exercise as a Therapeutic Strategy for Obesity: Central and Peripheral Mechanisms. Metabolites 2024; 14:589. [PMID: 39590824 PMCID: PMC11596326 DOI: 10.3390/metabo14110589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a complex, multifactorial condition involving excessive fat accumulation due to an imbalance between energy intake and expenditure, with its global prevalence steadily rising. This condition significantly increases the risk of chronic diseases, including sarcopenia, type 2 diabetes, and cardiovascular diseases, highlighting the need for effective interventions. Exercise has emerged as a potent non-pharmacological approach to combat obesity, targeting both central and peripheral mechanisms that regulate metabolism, energy expenditure, and neurological functions. In the central nervous system, exercise influences appetite, mood, and cognitive functions by modulating the reward system and regulating appetite-controlling hormones to manage energy intake. Concurrently, exercise promotes thermogenesis in adipose tissue and regulates endocrine path-ways and key metabolic organs, such as skeletal muscle and the liver, to enhance fat oxidation and support energy balance. Despite advances in understanding exercise's role in obesity, the precise interaction between the neurobiological and peripheral metabolic pathways remains underexplored, particularly in public health strategies. A better understanding of these interactions could inform more comprehensive obesity management approaches by addressing both central nervous system influences on behavior and peripheral metabolic regulation. This review synthesizes recent insights into these roles, highlighting potential therapeutic strategies targeting both systems for more effective obesity interventions.
Collapse
Affiliation(s)
- Yiyin Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Ruwen Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| |
Collapse
|
188
|
Jafari E, Dadgar H, Zarei A, Samimi R, Manafi-Farid R, Divband G, Nikkholgh B, Fallahi B, Amini H, Ahmadzadehfar H, Keshavarz A, Assadi M. The role of [68Ga]Ga-PSMA PET/CT in primary staging of newly diagnosed prostate cancer: predictive value of PET-derived parameters for risk stratification through machine learning. Clin Transl Imaging 2024; 12:669-682. [DOI: 10.1007/s40336-024-00666-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/21/2024] [Indexed: 01/03/2025]
|
189
|
Zhang Z, Wang L, Li X, Miao Y, Li D. Integrating Network Pharmacology, Molecular Docking and Experimental Validation to Explore the Pharmacological Mechanisms of Quercetin Against Diabetic Wound. Int J Med Sci 2024; 21:2837-2850. [PMID: 39512686 PMCID: PMC11539386 DOI: 10.7150/ijms.100468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024] Open
Abstract
The chronic non-healing diabetic wound (DW) has remained a challenge to both the society and individuals. Previous studies suggested dietary moderate consumption of quercetin (QCT) are beneficial in preventing diabetic complications, including non-healing DW. However, there were few studies that have investigated QCT-related underlying molecular mechanisms against DW. In the present study, we for the first-time combined network pharmacology with molecular docking and experimental validation to investigate QCT-related therapeutic targets and mechanisms for treating DW. Finally, 191 QCT-related targets and 1750 DW-related pathogenetic targets were obtained from online databases. After removing duplicates, a total of 90 potential therapeutic targets of quercetin for treating DW were ultimately identified. Furthermore, 7 targets with higher degree including IL-6, EGFR, SRC, TNF, AKT1, JUN and MMP9 were predicted as central therapeutic targets of QCT for treating DW. Functional enrichment analysis demonstrated that QCT exerted strong levels of multitargeting regulatory activity. In addition, the KEGG enrichment analysis indicated that several signaling pathways including AGE-RAGE signaling pathway in diabetic complications, IL-17, PI3k-AKT, TNF, HIF-1, VEGF were predicted as key regulators of QCT for treating DW. Molecular docking results suggested that QCT had strong binding activity with the predicted targets. In addition, verification experiments suggested that QCT could significantly attenuated the expression of inflammatory cytokines and the regulation of PI3K-AKT signaling pathway was probably a vital mechanism involved in the pharmacological mechanism of QCT for treating DW. Taken together, combined network pharmacological with experimental validation, we for the first time systematically investigated associated-therapeutic targets and potential pathways of QCT for DW treatment. Our study might provide theoretical basis for DW treatment.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of General Surgery & VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Lei Wang
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Xuan Li
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning Province, 110122, China
| | - Dongyu Li
- Department of General Surgery & VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| |
Collapse
|
190
|
Stathori G, Hatziagapiou K, Mastorakos G, Vlahos NF, Charmandari E, Valsamakis G. Endocrine-Disrupting Chemicals, Hypothalamic Inflammation and Reproductive Outcomes: A Review of the Literature. Int J Mol Sci 2024; 25:11344. [PMID: 39518897 PMCID: PMC11545284 DOI: 10.3390/ijms252111344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Endocrine-disrupting chemicals (EDCs) are environmental and industrial agents that interfere with hormonal functions. EDC exposure is linked to various endocrine diseases, especially in reproduction, although the mechanisms remain unclear and effects vary among individuals. Neuroinflammation, particularly hypothalamic inflammation, is an emerging research area with implications for endocrine-related diseases like obesity. The hypothalamus plays a crucial role in regulating reproduction, and its inflammation can adversely affect reproductive health. EDCs can cross the blood-brain barrier, potentially causing hypothalamic inflammation and disrupting the reproductive axis. This review examines the existing literature on EDC-mediated hypothalamic inflammation. Our findings suggest that exposure to 2,3,7,8-tetrachloro-dibenzo-p-dioxin (TCDD), polychlorinated biphenyl (PCB), tributyltin (TBT), phthalates, bisphenol A (BPA), and chlorpyrifos (CPF) in animals is linked to hypothalamic inflammation, specifically affecting the hypothalamic centers of the gonadotropic axis. To our knowledge, this is the first comprehensive review on this topic, indicating hypothalamic inflammation as a possible mediator between EDC exposure and reproductive dysfunction. Further human studies are needed to develop effective prevention and treatment strategies against EDC exposure.
Collapse
Affiliation(s)
- Galateia Stathori
- Center for Prevention and Management of Overweight and Obesity, Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (G.S.); (E.C.)
| | - Kyriaki Hatziagapiou
- Division of Endocrinology, Metabolism and Diabetes, ENDO-ERN Center for Rare Pediatric Endocrine Disorders, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece;
- Department of Physiotherapy, School of Health and Care Sciences, University of West Attica, 12243 Egaleo, Greece
| | - George Mastorakos
- Second Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, ‘Aretaieion’ University Hospital, 11528 Athens, Greece; (G.M.); (N.F.V.)
| | - Nikolaos F. Vlahos
- Second Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, ‘Aretaieion’ University Hospital, 11528 Athens, Greece; (G.M.); (N.F.V.)
| | - Evangelia Charmandari
- Center for Prevention and Management of Overweight and Obesity, Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (G.S.); (E.C.)
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Georgios Valsamakis
- Second Department of Obstetrics and Gynecology, Medical School, National and Kapodistrian University of Athens, ‘Aretaieion’ University Hospital, 11528 Athens, Greece; (G.M.); (N.F.V.)
| |
Collapse
|
191
|
Lan T, Lu Y, He J, Zhan C, Wang X, Shao X, Hu Z. Global, reginal, national burden and risk factors in female breast cancer from 1990 to 2021. iScience 2024; 27:111045. [PMID: 39435147 PMCID: PMC11491723 DOI: 10.1016/j.isci.2024.111045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/31/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
This study was to assess the burden, trends, and risk factors associated with female breast cancer from 1990 to 2021 based on the Global Burden of Disease (GBD) 2021 study. In 2021, there were 20.32 million prevalent cases, 2.08 million incident cases, 0.66 million death cases, and 20.26 million disability-adjusted life years (DALYs). It presented an ascending trend in the age-standardized rates of prevalence and incidence over the past 32 years. The age-standardized DALYs rate (ASDR) increased slightly during 2012-2021. The DALYs increase was primarily driven by population aging and growth. High red meat intake accounted for the highest proportion of ASDR. Breast cancer burden attributed to metabolic risks increased, especially in the regions with low social-development index (SDI) and limited health systems. Dietary, behavior, and metabolic risk factors should be controlled to diminish breast cancer burden, especially in countries with lower SDI.
Collapse
Affiliation(s)
- Tian Lan
- Department of Breast Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Yunyan Lu
- Department of Cardiology, The First People’s Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, Zhejiang, China
| | - Jiawei He
- Department of Breast Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Chenni Zhan
- Department of Breast Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Xiaojia Wang
- Department of Medical Oncology (Breast Cancer), Cancer Hospital of the University of Chinese Academy of Sciences/Zhejiang Cancer Hospital, Hangzhou, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiying Shao
- Department of Medical Oncology (Breast Cancer), Cancer Hospital of the University of Chinese Academy of Sciences/Zhejiang Cancer Hospital, Hangzhou, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Zujian Hu
- Department of Breast Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
192
|
Schrank Y, Fontes R, Perozo AFDF, Araújo PB, Pinheiro MFMC, Gomes DMV, Vieira LMF. Proposal for fasting insulin and HOMA-IR reference intervals based on an extensive Brazilian laboratory database. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230483. [PMID: 39529982 PMCID: PMC11554367 DOI: 10.20945/2359-4292-2023-0483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 05/28/2024] [Indexed: 11/16/2024]
Abstract
Objective Fasting insulin and the homeostasis model assessment of insulin resistance (HOMA-IR) index are relatively simple and reliable noninvasive markers of insulin resistance (IR). Given the relevance of correctly diagnosing IR, we emphasize the importance of establishing reliable reference intervals (RIs) for these markers. This study aimed to determine the RIs of fasting insulin and HOMA-IR index in adults living in Rio de Janeiro and, secondarily, to verify potential RI differences between sexes. Subjects and methods Serum insulin levels of 146,497 individuals (ages 20-60 years) who underwent blood sampling in the state of Rio de Janeiro were obtained retrospectively through access to an extensive laboratory database. Insulin was measured using the electrochemiluminescence immunoassay method. After applying exclusion criteria, 21,684 individuals (18,576 [86%] women) were included. The RIs were estimated using a computational mining approach that integrates a selection of R packages. Results The 95% RIs in women and men and in the overall population were, respectively, 2.54-13.30 μU/mL (15.3-80.12 pmol/L), 2.43-11.89 μU/mL (14.6-71.7 pmol/L), and 2.52-13.14 μU/mL (15.2-79.2 pmol/L) for fasting insulin levels and 0.39-2.86, 0.38-2.81, and 0.39-2.86 for HOMA-IR values. Despite significant differences in insulin levels and HOMA-IR index between men and women, the use of sex-specific RIs was not justified. Conclusion The RIs of fasting insulin levels and HOMA-IR values found in the overall population can be applied to both sexes. Thus, for our population, we suggest the RIs of 2.52-13.14 μU/mL (15.1-78.8 pmol/L) for fasting insulin and 0.39-2.86 for the HOMA-IR index.
Collapse
Affiliation(s)
- Yolanda Schrank
- Diagnósticos da América S.A.São PauloSPBrasilDiagnósticos da América S.A. (DASA), São Paulo, SP, Brasil
- Hospital Federal de BonsucessoRio de JaneiroRJBrasilHospital Federal de Bonsucesso, Rio de Janeiro RJ, Brasil
| | - Rosita Fontes
- Diagnósticos da América S.A.São PauloSPBrasilDiagnósticos da América S.A. (DASA), São Paulo, SP, Brasil
- Instituto Estadual de Diabetes e Endocrinologia Luiz CapriglioneRio de JaneiroRJBrasilInstituto Estadual de Diabetes e Endocrinologia Luiz Capriglione (IEDE), Rio de Janeiro, RJ, Brasil
| | | | - Paula Bruna Araújo
- Rio de JaneiroRJBrasilIndependent Researcher, Rio de Janeiro, RJ, Brasil
| | - Maria Fernanda Miguens Castelar Pinheiro
- Diagnósticos da América S.A.São PauloSPBrasilDiagnósticos da América S.A. (DASA), São Paulo, SP, Brasil
- Instituto Estadual de Diabetes e Endocrinologia Luiz CapriglioneRio de JaneiroRJBrasilInstituto Estadual de Diabetes e Endocrinologia Luiz Capriglione (IEDE), Rio de Janeiro, RJ, Brasil
| | | | - Luisane Maria Falci Vieira
- Diagnósticos da América S.A.São PauloSPBrasilDiagnósticos da América S.A. (DASA), São Paulo, SP, Brasil
- International Federation of Clinical Chemistry and Laboratory Medicine Task Force Global Reference Intervals DatabaseMilanoItalyInternational Federation of Clinical Chemistry and Laboratory Medicine Task Force Global Reference Intervals Database, Milano, Italy
| |
Collapse
|
193
|
Arora M, Singh AK, Kumar A, Singh H, Pathak P, Grishina M, Yadav JP, Verma A, Kumar P. Semisynthetic phytochemicals in cancer treatment: a medicinal chemistry perspective. RSC Med Chem 2024; 15:3345-3370. [PMID: 39430100 PMCID: PMC11484407 DOI: 10.1039/d4md00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer is the uncontrolled proliferation of abnormal cells that invade other areas, spread to other organs, and cause metastases, which is the most common cause of death. A review of all FDA-approved new molecular entities (NMEs) shows that natural products and derivatives account for over one-third of all NMEs. Before 1940, unmodified products and derivatives accounted for 43% and 14% of NME registrations, respectively. Since then, the share of unmodified products has decreased to 9.5% of all approved NMEs, while the share of derivatives has increased to 28%. Since the 1940s, semi-synthetic and synthetic derivatives of natural substances have gained importance, and this trend continues to date. In this study, we have discussed in detail isolated phytoconstituents with chemical modifications that are either FDA-approved or under clinical trials, such as podophyllotoxin, Taxol (paclitaxel, docetaxel), vinca alkaloids (vincristine, vinblastine), camptothecin, genistein, cephalotaxine, rohitukine, and many more, which may act as essential leads to the development of novel anticancer agents. Furthermore, we have also discussed recent developments in the most potent semisynthetic phytoconstituents, their unique properties, and their importance in cancer treatment.
Collapse
Affiliation(s)
- Meghna Arora
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Ankit Kumar Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Adarsh Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Harshwardhan Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| | - Prateek Pathak
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, School of Pharmacy, GITAM (Deemed to be University) Hyderabad Campus India
| | - Maria Grishina
- Laboratory of Computational Modeling of Drugs, Higher Medical and Biological School, South Ural State University Chelyabinsk 454008 Russia
| | - Jagat Pal Yadav
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
- Pharmacology Research Laboratory, Faculty of Pharmaceutical Sciences, Rama University Kanpur 209217 India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences Prayagraj 211007 India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Ghudda Bathinda 151401 India
| |
Collapse
|
194
|
Wohlfert AJ, Phares J, Granholm AC. The mTOR Pathway: A Common Link Between Alzheimer's Disease and Down Syndrome. J Clin Med 2024; 13:6183. [PMID: 39458132 PMCID: PMC11508835 DOI: 10.3390/jcm13206183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Down syndrome (DS) is a chromosomal condition that causes many systemic dysregulations, leading to several possible age-related diseases including Alzheimer's disease (AD). This may be due to the triplication of the Amyloid precursor protein (APP) gene or other alterations in mechanistic pathways, such as the mTOR pathway. Impairments to upstream regulators of mTOR, such as insulin, PI3K/AKT, AMPK, and amino acid signaling, have been linked to amyloid beta plaques (Aβ) and neurofibrillary tangles (NFT), the most common AD pathologies. However, the mechanisms involved in the progression of pathology in human DS-related AD (DS-AD) are not fully investigated to date. Recent advancements in omics platforms are uncovering new insights into neurodegeneration. Genomics, spatial transcriptomics, proteomics, and metabolomics are novel methodologies that provide more data in greater detail than ever before; however, these methods have not been used to analyze the mTOR pathways in connection to DS-AD. Using these new techniques can unveil unexpected insights into pathological cellular mechanisms through an unbiased approach.
Collapse
Affiliation(s)
- Abigail J. Wohlfert
- Department of Modern Human Anatomy and Cell & Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Jeremiah Phares
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| | - Ann-Charlotte Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA;
| |
Collapse
|
195
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
196
|
Hassanzadeh K, Liu J, Maddila S, Mouradian MM. Posttranslational Modifications of α-Synuclein, Their Therapeutic Potential, and Crosstalk in Health and Neurodegenerative Diseases. Pharmacol Rev 2024; 76:1254-1290. [PMID: 39164116 PMCID: PMC11549938 DOI: 10.1124/pharmrev.123.001111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024] Open
Abstract
α-Synuclein (α-Syn) aggregation in Lewy bodies and Lewy neurites has emerged as a key pathogenetic feature in Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Various factors, including posttranslational modifications (PTMs), can influence the propensity of α-Syn to misfold and aggregate. PTMs are biochemical modifications of a protein that occur during or after translation and are typically mediated by enzymes. PTMs modulate several characteristics of proteins including their structure, activity, localization, and stability. α-Syn undergoes various posttranslational modifications, including phosphorylation, ubiquitination, SUMOylation, acetylation, glycation, O-GlcNAcylation, nitration, oxidation, polyamination, arginylation, and truncation. Different PTMs of a protein can physically interact with one another or work together to influence a particular physiological or pathological feature in a process known as PTMs crosstalk. The development of detection techniques for the cooccurrence of PTMs in recent years has uncovered previously unappreciated mechanisms of their crosstalk. This has led to the emergence of evidence supporting an association between α-Syn PTMs crosstalk and synucleinopathies. In this review, we provide a comprehensive evaluation of α-Syn PTMs, their impact on misfolding and pathogenicity, the pharmacological means of targeting them, and their potential as biomarkers of disease. We also highlight the importance of the crosstalk between these PTMs in α-Syn function and aggregation. Insight into these PTMS and the complexities of their crosstalk can improve our understanding of the pathogenesis of synucleinopathies and identify novel targets of therapeutic potential. SIGNIFICANCE STATEMENT: α-Synuclein is a key pathogenic protein in Parkinson's disease and other synucleinopathies, making it a leading therapeutic target for disease modification. Multiple posttranslational modifications occur at various sites in α-Synuclein and alter its biophysical and pathological properties, some interacting with one another to add to the complexity of the pathogenicity of this protein. This review details these modifications, their implications in disease, and potential therapeutic opportunities.
Collapse
Affiliation(s)
- Kambiz Hassanzadeh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Jun Liu
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Santhosh Maddila
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| |
Collapse
|
197
|
Sapkota S, Roy SC, Briski KP. Dorsomedial Ventromedial Hypothalamic Nucleus Growth Hormone-Releasing Hormone Neuron Steroidogenic Factor-1 Gene Targets in Female Rat. ASN Neuro 2024; 16:2403345. [PMID: 39401164 PMCID: PMC11792125 DOI: 10.1080/17590914.2024.2403345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/07/2024] [Indexed: 10/18/2024] Open
Abstract
The prospect that the ventromedial hypothalamic nucleus (VMN) transcription factor steroidogenic factor-1/NR5A1 (SF-1) may exert sex-dimorphic control of glucose counterregulation is unresolved. Recent studies in male rats show that SF-1 regulates transcription of co-expressed hypoglycemia-sensitive neurochemicals in dorsomedial VMN growth hormone-releasing hormone (Ghrh) neurons. Gene knockdown and laser-catapult-microdissection/single-cell multiplex qPCR techniques were used here in a female rat model to determine if SF-1 control of Ghrh neuron transmitter marker, energy sensor, and estrogen receptor (ER) variant mRNAs varies according to sex. Data show that in females, hypoglycemia elicits a gain of SF-1 inhibitory control of VMNdm Ghrh neuron Ghrh and Ghrh-receptor gene profiles and loss of augmentation of glutaminase transcription; SF-1 gene silencing diminished eu- and hypoglycemic patterns of neuronal nitric oxide gene transcription. SF-1 imposes divergent control of baseline and hypoglycemic glutamate decarboxylase65 (GAD)-1 (stimulatory) versus GAD2 (inhibitory) mRNAs in that sex. SF-1 stimulates baseline VMNdm Ghrh neuron PRKAA1/AMPKα1 and PRKAA2/AMPKα2 gene expression, yet causes opposite changes in these gene profiles during hypoglycemia. SF-1 exerts glucose-dependent control of ER-alpha and G-protein-coupled ER-1 transcription, but blunts ER-beta gene profiles during eu- and hypoglycemia. In females, SF-1 knockdown did not affect hypercorticosteronemia or hyperglucagonemia, but blunted hypoglycemic suppression of growth hormone secretion. Results show that SF-1 expression is critical for female rat VMNdm Ghrh neuron counterregulatory neurochemical, AMPK catalytic subunit, and ER gene transcription responses to hypoglycemia. Sex differences in direction of SF-1 control of distinctive gene profiles may result in observed disparities in SF-1 regulation of counterregulatory hormone secretion between sexes.
Collapse
Affiliation(s)
- Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Sagor C. Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Karen P. Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| |
Collapse
|
198
|
Grudet F, Martinot E, Godin P, Bérubé M, Chédotal A, Boerboom D. Slit1 inhibits ovarian follicle development and female fertility in mice†. Biol Reprod 2024; 111:834-844. [PMID: 38943353 PMCID: PMC11473917 DOI: 10.1093/biolre/ioae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024] Open
Abstract
Previous in vitro studies have suggested that SLIT ligands could play roles in regulating ovarian granulosa cell proliferation and gene expression, as well as luteolysis. However, no in vivo study of Slit gene function has been conducted to date. Here, we investigated the potential role of Slit1 in ovarian biology using a Slit1-null mouse model. Female Slit1-null mice were found to produce larger litters than their wild-type counterparts due to increased ovulation rates. Increased ovarian weights in Slit1-null animals were found to be due to the presence of greater numbers of healthy antral follicles with similar numbers of atretic ones, suggesting both an increased rate of follicle recruitment and a decreased rate of atresia. Consistent with this, treatment of cultured granulosa cells with exogenous SLIT1 induced apoptosis in presence or absence of follicle-stimulating hormone, but had no effect on cell proliferation. Although few alterations in the messenger RNA levels of follicle-stimulating hormone-responsive genes were noted in granulosa cells of Slit1-null mice, luteinizing hormone target gene mRNA levels were greatly increased. Finally, increased phospho-AKT levels were found in granulosa cells isolated from Slit1-null mice, and SLIT1 pretreatment of cultured granulosa cells inhibited the ability of both follicle-stimulating hormone and luteinizing hormone to increase AKT phosphorylation, suggesting a mechanism whereby SLIT1 could antagonize gonadotropin signaling. These findings therefore represent the first evidence for a physiological role of a SLIT ligand in the ovary, and define Slit1 as a novel autocrine/paracrine regulator of follicle development.
Collapse
Affiliation(s)
- Florine Grudet
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Emmanuelle Martinot
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Philippe Godin
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Michael Bérubé
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| |
Collapse
|
199
|
Sadia H, Sumalatha G. Evaluation of <i>Saussurea lappa</i> on Oxidative Stress and Cognition in Aluminium-induced Alzheimer’s Disease Rats. JOURNAL OF NATURAL REMEDIES 2024:2015-2025. [DOI: 10.18311/jnr/2024/43482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/01/2024] [Indexed: 01/05/2025]
Abstract
Background: Oxidative stress and neurodegenerative illnesses, such as Alzheimer's disease (AD), are closely associated. There has been a lot of thought put into finding medicinal plants with nootropic properties to slow the onset and course of AD. Objective: The study aimed to evaluate the methanolic extract of Saussurea lappa clarke (MESC) on oxidative stress and cognitive ability induced by aluminium exposure. Methods: Wistar albino rats were chosen for the study. About 30 animals were selected and grouped into 5 with 6 animals in each group. Group I served as control, group II served as disease induced (Aluminium-induced), group III, IV and V were administered with standard drug – Donepezil Hcl, and MESC at two doses – 200 and 400 mg/kg. The behavioural studies were examined by using certain apparatus like Passive Avoidance (PA) test, Elevated Plus Maze, Y- Maze and Actophotometer. Determination of anti-oxidant enzymes – Catalase (CAT) and thiobarbituric acid reactive substances (TBARS) along with acetylcholinesterase (AChE) levels which was done in rat’s brain homogenate. Results: In the PA test, administration of MESC at doses of 200 and 400 mg/kg significantly (**p< 0.01) lengthened step-through latency (STL) in rats on day 30 compared to the positive control group. Animals at MESC (200 & 400 mg/kg) showed noticeably higher memory retention (MR) rates as compared to the disease-control group. Additionally, administration of MESC (200 and 400 mg/kg) significantly (**p< 0.01) raised CAT and declined the concentration of TBARS. AChE concentration was significantly (**p< 0.01) reduced at the dose of MESC at 200 and 400 mg/kg as compared to the positive control group. Conclusion: The present study showed that MESC had a strong nootropic effect on brain antioxidant indicators and cognitive function in rats exposed to aluminium-induced oxidative stress and cognitive impairment. These findings may be investigated in the treatment of neurodegenerative diseases, including AD.
Collapse
|
200
|
Wang XJ. Pituitary metastasis from lung adenocarcinoma. World J Clin Cases 2024; 12:6155-6158. [PMID: 39371557 PMCID: PMC11362882 DOI: 10.12998/wjcc.v12.i28.6155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 08/13/2024] Open
Abstract
Pituitary tumor is a common neuroendocrine tumor, but there are also rare clinical metastases at this site, which are generally transferred from extrabellar tumors. Although the clinical incidence is low, the prognosis is poor. The purpose of this editorial is to discuss further the relevant knowledge of pituitary metastases and remind clinicians to prevent missed diagnosis and improve the prognosis of these patients.
Collapse
Affiliation(s)
- Xue-Jian Wang
- Department of Neurosurgery, Affiliated Hospital 2 of Nantong University, Nantong 226000, Jiangsu Province, China
| |
Collapse
|