Avsnitt

  • BUFFALO, NY- April 24, 2024 – A new #researchpaper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 7, entitled, “Using genetics and proteomics data to identify proteins causally related to COVID-19, healthspan and lifespan: a Mendelian randomization study.”

    The COVID-19 pandemic poses a heavy burden on public health and accounts for substantial mortality and morbidity. Proteins are building blocks of life, but specific proteins causally related to COVID-19, healthspan and lifespan, have not been systematically examined. In this new study, researchers Jie V. Zhao, Minhao Yao, and Zhonghua Liu from The University of Hong Kong and Columbia University conducted a Mendelian randomization study to assess the effects of 1,361 plasma proteins on COVID-19, healthspan and lifespan, using large GWAS of severe COVID-19 (up to 13,769 cases and 1,072,442 controls), COVID-19 hospitalization (32,519 cases and 2,062,805 controls) and SARS-COV2 infection (122,616 cases and 2,475,240 controls), healthspan (n = 300,477) and parental lifespan (~0.8 million of European ancestry).

    “We included both COVID-19 and healthspan and lifespan in the outcome, because COVID-19 which occurred in recent years reflects a new threat to longevity, whilst healthspan and lifespan reflect overall morbidity and mortality.”

    The researchers identified 35, 43, and 63 proteins for severe COVID, COVID-19 hospitalization, and SARS-COV2 infection, and 4, 32, and 19 proteins for healthspan, father’s attained age, and mother’s attained age. In addition to some proteins reported previously, such as SFTPD related to severe COVID-19, the team identified novel proteins involved in inflammation and immunity (such as ICAM-2 and ICAM-5 which affect COVID-19 risk, CXCL9, HLA-DRA and LILRB4 for healthspan and lifespan), apoptosis (such as FGFR2 and ERBB4 which affect COVID-19 risk and FOXO3 which affect lifespan) and metabolism (such as PCSK9 which lowers lifespan). They found 2, 2, and 3 proteins shared between COVID-19 and healthspan/lifespan, such as CXADR and LEFTY2, shared between severe COVID-19 and healthspan/lifespan. Three proteins affecting COVID-19 and seven proteins affecting healthspan/lifespan are targeted by existing drugs.

    “Our study provided novel insights into protein targets affecting COVID-19, healthspan and lifespan, with implications for developing new treatment and drug repurposing.”

    DOI - https://doi.org/10.18632/aging.205711

    Corresponding authors - Jie V. Zhao - [email protected], and Zhonghua Liu - [email protected]

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    [email protected]

  • BUFFALO, NY- April 23, 2024 – A new #researchpaper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 7, entitled, “The coupling between healthspan and lifespan in Caenorhabditis depends on complex interactions between compound intervention and genetic background.”

    Aging is characterized by declining health that results in decreased cellular resilience and neuromuscular function. The relationship between lifespan and health, and the influence of genetic background on that relationship, has important implications in the development of pharmacological anti-aging interventions.

    In this new study, researchers Stephen A. Banse, E. Grace Jackson, Christine A. Sedore, Brian Onken, David Hall, Anna Coleman-Hulbert, Phu Huynh, Theo Garrett, Erik Johnson, Girish Harinath, Delaney Inman, Suzhen Guo, Mackenzie Morshead, Jian Xue, Ron Falkowski, Esteban Chen, Christopher Herrera, Allie J. Kirsch, Viviana I. Perez, Max Guo, Gordon J. Lithgow, Monica Driscoll, and Patrick C. Phillips from the University of Oregon, The State University of New Jersey (Rutgers), The Buck Institute for Research on Aging, and National Institute on Aging assessed swimming performance as well as survival under thermal and oxidative stress across a nematode genetic diversity test panel to evaluate health effects for three compounds previously studied in the Caenorhabditis Intervention Testing Program and thought to promote longevity in different ways – NP1 (nitrophenyl piperazine-containing compound 1), propyl gallate, and resveratrol.

    “Overall, we find the relationships among median lifespan, oxidative stress resistance, thermotolerance, and mobility vigor to be complex.”

    The researchers showed that oxidative stress resistance and thermotolerance vary with compound intervention, genetic background, and age. The effects of tested compounds on swimming locomotion, in contrast, are largely species-specific. In this study, thermotolerance, but not oxidative stress or swimming ability, correlates with lifespan. Notably, some compounds exert strong impact on some health measures without an equally strong impact on lifespan.

    “Our results demonstrate the importance of assessing health and lifespan across genetic backgrounds in the effort to identify reproducible anti-aging interventions, with data underscoring how personalized treatments might be required to optimize health benefits.”

    DOI - https://doi.org/10.18632/aging.205743

    Corresponding authors - Gordon J. Lithgow - [email protected], Monica Driscoll - [email protected], and Patrick C. Phillips - [email protected]

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    [email protected]

  • Saknas det avsnitt?

    Klicka här för att uppdatera flödet manuellt.

  • BUFFALO, NY - April 22, 2024 – The Longevity & Aging Series is an enlightening and progressive video interview series presented by Aging, featuring esteemed researchers discussing the latest in aging research, with a focus on their studies published by Aging.

    In the premiere episode of the second season of the Longevity & Aging Series, Dr. Irina Conboy and Xiaoyue (Serafina) Mei from the Department of Bioengineering and QB3 at the University of California, Berkeley, discuss a priority research paper they co-authored and published in Aging’s Volume 15, Issue 17, entitled, “Fail-tests of DNA methylation clocks, and development of a noise barometer for measuring epigenetic pressure of aging and disease.”

    DOI - https://doi.org/10.18632/aging.205046

    Corresponding Author - Irina M. Conboy - [email protected]

    Video - https://www.youtube.com/watch?v=aYhUPpEPVv4

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205046

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, DNA methylation, epigenetics, clocks’ fail-test, biological noise

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Aging is indexed by PubMed/Medline (abbreviated as “Aging (Albany NY)”), PubMed Central, Web of Science: Science Citation Index Expanded (abbreviated as “Aging‐US” and listed in the Cell Biology and Geriatrics & Gerontology categories), Scopus (abbreviated as “Aging” and listed in the Cell Biology and Aging categories), Biological Abstracts, BIOSIS Previews, EMBASE, META (Chan Zuckerberg Initiative) (2018-2022), and Dimensions (Digital Science).

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    Media Contact
    18009220957
    [email protected]

  • In the season 2 premiere episode of the Longevity & Aging Series, Dr. Irina Conboy and Xiaoyue (Serafina) Mei from the Department of Bioengineering and QB3 at the University of California, discuss a research paper they co-authored that was published in Volume 15, Issue 17, of Aging (Aging-US), entitled, “Fail-tests of DNA methylation clocks, and development of a noise barometer for measuring epigenetic pressure of aging and disease.”

    DOI - https://doi.org/10.18632/aging.205046

    Corresponding Author - Irina M. Conboy - [email protected]

    Video interview - https://www.youtube.com/watch?v=aYhUPpEPVv4

    Transcript - https://aging-us.net/2024/04/22/longevity-aging-series-s2-e1-dr-irina-conboy-and-xiaoyue-serafina-mei/

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205046

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, DNA methylation, epigenetics, clocks’ fail-test, biological noise

    About Longevity & Aging Series

    In its second season, the Longevity & Aging Series is a monthly video series that features esteemed researchers discussing the latest in aging research with a focus on their studies published by Aging (Aging-US).

    Learn more - https://www.aging-us.com/longevity

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Aging is indexed by PubMed/Medline (abbreviated as “Aging (Albany NY)”), PubMed Central, Web of Science: Science Citation Index Expanded (abbreviated as “Aging‐US” and listed in the Cell Biology and Geriatrics & Gerontology categories), Scopus (abbreviated as “Aging” and listed in the Cell Biology and Aging categories), Biological Abstracts, BIOSIS Previews, EMBASE, META (Chan Zuckerberg Initiative) (2018-2022), and Dimensions (Digital Science).

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    Media Contact
    18009220957
    [email protected]

  • Aging is still shrouded in proverbial darkness. But, some researchers hypothesize that aging may be linked to stem cell exhaustion. Stemness, the ability of a cell to differentiate into various cell types, is an essential characteristic defining the functionality of stem cells. It has been observed that stem cells seem to diminish with age, although the precise role of stem cells in human aging remains to be elucidated.

    “Among the biological pathways associated with aging, we can highlight stem cell exhaustion, which argues that during normal aging, the decrease in the number or activity of these cells contributes to physiological dysfunction in aged tissues [4].”

    In a new study, researchers Gabriel Arantes dos Santos, Gustavo Daniel Vega Magdaleno and João Pedro de Magalhães from the Universidade de Sao Paulo, University of Birmingham and the University of Liverpool applied a machine learning method to detect stemness signatures from transcriptome data of healthy human tissues. Their research paper was published on April 4, 2024, and chosen as the cover of Aging’s Volume 16, Issue 7, entitled, “Evidence of a pan-tissue decline in stemness during human aging.”

    Full blog - https://aging-us.org/2024/04/first-evidence-of-a-pan-tissue-decline-in-stemness-during-human-aging/

    Paper DOI - https://doi.org/10.18632/aging.205717

    Corresponding author - João Pedro de Magalhães - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205717

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, longevity, stem cells, transcriptomics, senescence

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Aging is indexed by PubMed/Medline (abbreviated as “Aging (Albany NY)”), PubMed Central, Web of Science: Science Citation Index Expanded (abbreviated as “Aging‐US” and listed in the Cell Biology and Geriatrics & Gerontology categories), Scopus (abbreviated as “Aging” and listed in the Cell Biology and Aging categories), Biological Abstracts, BIOSIS Previews, EMBASE, META (Chan Zuckerberg Initiative) (2018-2022), and Dimensions (Digital Science).

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    Media Contact
    18009220957
    [email protected]

  • BUFFALO, NY- April 17, 2024 – A new #researchpaper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 7, entitled, “The impact of continuous and intermittent ketogenic diets on cognitive behavior, motor function, and blood lipids in TgF344-AD rats.”

    Studies suggest that ketogenic diets (KD) may improve memory in mouse models of aging and Alzheimer’s disease (AD). In this new study, researchers Jennifer M. Rutkowsky, Zabrisky Roland, Anthony Valenzuela, An B. Nguyen, Heui Hye Park, Natalie Six, Ilknur Dursun, Kyoungmi Kim, Pamela J. Lein, and Jon J. Ramsey from the University of California Davis and Istinye University determined whether a continuous or intermittent KD (IKD) enhanced cognitive behavior in the TgF344-AD rat model of AD.

    “[...] it remains to be determined whether long-term consumption of a ketogenic diet can mitigate declines in cognitive or motor behavior in a rat model of AD. Therefore, the current study aimed to determine whether a KD improves cognitive or motor behavior in the TgF344-AD rat.”

    At 6 months-old, TgF344-AD and wild-type (WT) littermates were placed on a control (CD), KD, or IKD (morning CD and afternoon KD) provided as two meals per day for 2 or 6 months. Cognitive and motor behavior and circulating β-hydroxybutyrate (BHB), AD biomarkers and blood lipids were assessed. Animals on a KD diet had elevated circulating BHB, with IKD levels intermediate to CD and KD.

    TgF344-AD rats displayed impaired spatial learning memory in the Barnes maze at 8 and 12 months of age and impaired motor coordination at 12 months of age. Neither KD nor IKD improved performance compared to CD. At 12 months of age, TgF344-AD animals had elevated blood lipids. IKD reduced lipids to WT levels with KD further reducing cholesterol below WT levels.

    “[...] the IKD or KD did not improve motor coordination or spatial learning memory compared to the control diet. However, KD, and to a lesser extent IKD, mitigated elevations in plasma lipids in the TgF344-AD rats. Furthermore, the KD diet decreased plasma levels of total Tau in females.”

    DOI - https://doi.org/10.18632/aging.205741

    Corresponding author - Jennifer M. Rutkowsky - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205741

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, ketogenic diet, Alzheimer’s disease, cognitive behavior, motor function, lipids

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    [email protected]

  • BUFFALO, NY- April 15, 2024 – A new #researchpaper was #published on the #cover of Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 7, entitled, “Evidence of a pan-tissue decline in stemness during human aging.”

    Despite their biological importance, the role of stem cells in human aging remains to be elucidated. In a new study, researchers Gabriel Arantes dos Santos, Gustavo Daniel Vega Magdaleno and João Pedro de Magalhães from the Universidade de Sao Paulo, University of Birmingham and the University of Liverpool applied a machine learning method to detect stemness signatures from transcriptome data of healthy human tissues.

    “In this work, we applied a machine learning methodology to GTEx transcriptome data and assigned stemness scores to 17,382 healthy samples from 30 human tissues aged between 20 and 79 years.”

    The team found that ~60% of the studied tissues exhibit a significant negative correlation between the subject's age and stemness score. The only significant exception was the uterus, where they observed an increased stemness with age. Moreover, the researchers observed that stemness is positively correlated with cell proliferation and negatively correlated with cellular senescence. Finally, they also observed a trend that hematopoietic stem cells derived from older individuals might have higher stemness scores.

    “In conclusion, we assigned stemness scores to human samples and show evidence of a pan-tissue loss of stemness during human aging, which adds weight to the idea that stem cell deterioration may contribute to human aging.”

    DOI - https://doi.org/10.18632/aging.205717

    Corresponding author - João Pedro de Magalhães - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205717

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, longevity, stem cells, transcriptomics, senescence

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Aging is indexed by PubMed/Medline (abbreviated as “Aging (Albany NY)”), PubMed Central, Web of Science: Science Citation Index Expanded (abbreviated as “Aging‐US” and listed in the Cell Biology and Geriatrics & Gerontology categories), Scopus (abbreviated as “Aging” and listed in the Cell Biology and Aging categories), Biological Abstracts, BIOSIS Previews, EMBASE, META (Chan Zuckerberg Initiative) (2018-2022), and Dimensions (Digital Science).

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    Media Contact
    18009220957
    [email protected]

  • BUFFALO, NY- April 10, 2024 – A new #researchpaper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 6, entitled, “Geraniol attenuates oxidative stress and neuroinflammation-mediated cognitive impairment in D galactose-induced mouse aging model.”

    D-galactose (D-gal) administration was proven to induce cognitive impairment and aging in rodents’ models. Geraniol (GNL) belongs to the acyclic isoprenoid monoterpenes. GNL reduces inflammation by changing important signaling pathways and cytokines, and thus it is plausible to be used as a medicine for treating disorders linked to inflammation. In this new study, researchers Peramaiyan Rajendran, Fatma J. Al-Saeedi, Rebai Ben Ammar, Basem M. Abdallah, Enas M. Ali, Najla Khaled Al Abdulsalam, Sujatha Tejavat, Duaa Althumairy, Vishnu Priya Veeraraghavan, Sarah Abdulaziz Alamer, Gamal M. Bekhet, and Emad A. Ahmed from King Faisal University, Kuwait University, Center of Biotechnology of Borj-Cedria, Saveetha University, Alexandria University, and Assiut University examined the therapeutic effects of GNL on D-gal-induced oxidative stress and neuroinflammation-mediated memory loss in mice.

    “Life expectancy in the 21st century is rising, resulting in more age-related illnesses, such as memory impairment and Alzheimer’s disease. In this study, GNL was studied for its protective effect on D-gal-induced aging in mice.”

    The study was conducted using six groups of mice (6 mice per group). The first group received normal saline, then D-gal (150 mg/wt) dissolved in normal saline solution (0.9%, w/v) was given orally for 9 weeks to the second group. In the III group, from the second week until the 10th week, mice were treated orally (without anesthesia) with D-gal (150 mg/kg body wt) and GNL weekly twice (40 mg/kg body wt) four hours later. Mice in Group IV were treated with GNL from the second week up until the end of the experiment. For comparison of young versus elderly mice, 4 month old (Group V) and 16-month-old (Group VI) control mice were used.

    “We evaluated the changes in antioxidant levels, PI3K/Akt levels, and Nrf2 levels. We also examined how D-gal and GNL treated pathological aging changes.”

    Administration of GNL induced a significant increase in spatial learning and memory with spontaneously altered behavior. Enhancing anti-oxidant and anti-inflammatory effects and activating PI3K/Akt were the mechanisms that mediated this effect. Further, GNL treatment upregulated Nrf2 and HO-1 to reduce oxidative stress and apoptosis. This was confirmed using 99mTc-HMPAO brain flow gamma bioassays.

    “Thus, our data suggested GNL as a promising agent for treating neuroinflammation-induced cognitive impairment.”

    DOI - https://doi.org/10.18632/aging.205677

    Corresponding authors - Peramaiyan Rajendran - [email protected], and Fatma J. Al-Saeedi - [email protected]

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​ and connect with us on social media.

    [email protected]

  • BUFFALO, NY- April 9, 2024 – A new #researchpaper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 6, entitled, “CMS121: a novel approach to mitigate aging-related obesity and metabolic dysfunction.”

    Modulated by differences in genetic and environmental factors, laboratory mice often show progressive weight gain, eventually leading to obesity and metabolic dyshomeostasis. The geroneuroprotector CMS121 has a positive effect on energy metabolism in a mouse model of type 2 diabetes. In this new study, researchers Alcir L. Dafre, Saadia Zahid, Jessica Jorge Probst, Antonio Currais, Jingting Yu, David Schubert, and Pamela Maher from Salk Institute for Biological Studies, National University of Sciences and Technology (NUST) and Federal University of Santa Catarina investigated the potential of CMS121 to counteract the metabolic changes observed during the ageing process of wild type mice.

    “This comprehensive analysis aimed to further understand how CMS121 influences the metabolic landscape, paving the way for potential therapeutic applications beyond its established geroneuroprotective benefits.”

    Control or CMS121-containing diets were supplied ad libitum for 6 months, and mice were sacrificed at the age of 7 months. Blood, adipose tissue, and liver were analyzed for glucose, lipids, and protein markers of energy metabolism. The CMS121 diet induced a 40% decrease in body weight gain and improved both glucose and lipid indexes. Lower levels of hepatic caspase 1, caspase 3, and NOX4 were observed with CMS121 indicating a lower liver inflammatory status. Adipose tissue from CMS121-treated mice showed increased levels of the transcription factors Nrf1 and TFAM, as well as markers of mitochondrial electron transport complexes, levels of GLUT4 and a higher resting metabolic rate. Metabolomic analysis revealed elevated plasma concentrations of short chain acylcarnitines and butyrate metabolites in mice treated with CMS121.

    “The diminished de novo lipogenesis, which is associated with increased acetyl-CoA, acylcarnitine, and butyrate metabolite levels, could contribute to safeguarding not only the peripheral system but also the aging brain. By mimicking the effects of ketogenic diets, CMS121 holds promise for metabolic diseases such as obesity and diabetes, since these diets are hard to follow over the long term.”

    DOI - https://doi.org/10.18632/aging.205673

    Corresponding authors - Pamela Maher - [email protected] and Alcir L. Dafre - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205673

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    [email protected]

  • Menopause marks the beginning of the next biological chapter in a woman’s life. Characterized by the natural ebb of reproductive hormones (particularly estrogen), menopause ushers in a new season of aging. This hormonal shift not only signifies a transition in fertility but also influences systemic health. The menopause-associated decline in estrogen has been associated with various health issues, including alterations in brain structure and function. However, the mechanics of this phenomenon are still poorly understood. A greater understanding of how menopause alters the brain could aid in the early detection, and possible prevention, of neurodegenerative disease.

    In a new study, researchers Gwang-Won Kim, Kwangsung Park, Yun-Hyeon Kim, and Gwang-Woo Jeong from Chonnam National University used neuroimaging to shed light on how menopause alters brain morphology and functional connectivity in postmenopausal women. On March 23, 2024, their research paper was published as the cover of Aging’s Volume 16, Issue 6, entitled, "Altered brain morphology and functional connectivity in postmenopausal women: automatic segmentation of whole-brain and thalamic subnuclei and resting-state fMRI."

    Full blog - https://aging-us.org/2024/04/how-menopause-changes-brain-structure-and-connectivity/

    Paper DOI - https://doi.org/10.18632/aging.205662

    Corresponding author - Gwang-Woo Jeong - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205662

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, brain morphology, functional connectivity, sex hormones, thalamic subnuclei

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Aging is indexed by PubMed/Medline (abbreviated as “Aging (Albany NY)”), PubMed Central, Web of Science: Science Citation Index Expanded (abbreviated as “Aging‐US” and listed in the Cell Biology and Geriatrics & Gerontology categories), Scopus (abbreviated as “Aging” and listed in the Cell Biology and Aging categories), Biological Abstracts, BIOSIS Previews, EMBASE, META (Chan Zuckerberg Initiative) (2018-2022), and Dimensions (Digital Science).

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    Media Contact
    18009220957
    [email protected]

  • BUFFALO, NY- April 3, 2024 – A new #research paper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 6, entitled, “Targeting mitochondrial dysfunction using methylene blue or mitoquinone to improve skeletal aging.”

    Methylene blue (MB) is a well-established antioxidant that has been shown to improve mitochondrial function in both in vitro and in vivo settings. Mitoquinone (MitoQ) is a selective antioxidant that specifically targets mitochondria and effectively reduces the accumulation of reactive oxygen species. In this new study, researchers Sher Bahadur Poudel, Dorra Frikha-Benayed, Ryan R. Ruff, Gozde Yildirim, Manisha Dixit, Ron Korstanje, Laura Robinson, Richard A. Miller, David E. Harrison, John R. Strong, Mitchell B. Schaffler, and Shoshana Yakar from New York University College of Dentistry, City College of New York, The Jackson Laboratory, University of Michigan, South Texas Veterans Health Care System, and The University of Texas Health Science Center investigated the effect of long-term administration of MB or MitoQ on skeletal morphology during the aging process.

    “[...] we administered MB to aged (18 months old) female C57BL/J6 mice, as well as to adult male and female mice with a genetically diverse background (UM-HET3). Additionally, we used MitoQ as an alternative approach to target mitochondrial oxidative stress during aging in adult female and male UM-HET3 mice.”

    Although the researchers observed some beneficial effects of MB and MitoQ in vitro, the administration of these compounds in vivo did not alter the progression of age-induced bone loss. Specifically, treating 18-month-old female mice with MB for 6 or 12 months did not have an effect on age-related bone loss. Similarly, long-term treatment with MB from 7 to 22 months or with MitoQ from 4 to 22 months of age did not affect the morphology of cortical bone at the mid-diaphysis of the femur, trabecular bone at the distal-metaphysis of the femur, or trabecular bone at the lumbar vertebra-5 in UM-HET3 mice.

    “Based on our findings, it appears that long-term treatment with MB or MitoQ alone, as a means to reduce skeletal oxidative stress, is insufficient to inhibit age-associated bone loss. This supports the notion that interventions solely with antioxidants may not provide adequate protection against skeletal aging.”

    DOI - https://doi.org/10.18632/aging.205147

    Corresponding author - Shoshana Yakar - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205147

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    [email protected]

  • BUFFALO, NY- April 1, 2024 – A new #research paper was #published on the #cover of Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 6, entitled, “Altered brain morphology and functional connectivity in postmenopausal women: automatic segmentation of whole-brain and thalamic subnuclei and resting-state fMRI.”

    The transition to menopause is associated with various physiological changes, including alterations in brain structure and function. However, menopause-related structural and functional changes are poorly understood. In this new study, researchers Gwang-Won Kim, Kwangsung Park, Yun-Hyeon Kim, and Gwang-Woo Jeong from Chonnam National University not only compared the brain volume changes between premenopausal and postmenopausal women, but also evaluated the functional connectivity between the targeted brain regions associated with structural atrophy in postmenopausal women.

    “To the best of our knowledge, no comparative neuroimaging study on alterations in the brain volume and functional connectivity, especially focusing on the thalamic subnuclei in premenopausal vs. postmenopausal women has been reported.”

    Each of the 21 premenopausal and postmenopausal women underwent magnetic resonance imaging (MRI). T1-weighted MRI and resting-state functional MRI data were used to compare the brain volume and seed-based functional connectivity, respectively. In statistical analysis, multivariate analysis of variance, with age and whole brain volume as covariates, was used to evaluate surface areas and subcortical volumes between the two groups.

    Postmenopausal women showed significantly smaller cortical surface, especially in the left medial orbitofrontal cortex (mOFC), right superior temporal cortex, and right lateral orbitofrontal cortex, compared to premenopausal women (p < 0.05, Bonferroni-corrected) as well as significantly decreased functional connectivity between the left mOFC and the right thalamus was observed (p < 0.005, Monte-Carlo corrected). Although postmenopausal women did not show volume atrophy in the right thalamus, the volume of the right pulvinar anterior, which is one of the distinguished thalamic subnuclei, was significantly decreased (p < 0.05, Bonferroni-corrected).

    “Postmenopausal women showed significantly lower left mOFC, right lOFC, and right STC surface areas, reduced right PuA volume, and decreased left mOFC-right thalamus functional connectivity compared to premenopausal women. If replicated in an independent sample, these findings will be helpful for understanding the effects of menopause on the altered brain volume and functional connectivity in postmenopausal women.”

    DOI - https://doi.org/10.18632/aging.205662

    Corresponding author - Gwang-Woo Jeong - [email protected]

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​.

    [email protected]

  • BUFFALO, NY- March 26, 2024 – A new #researchpaper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 5, entitled, “Chlorogenic acid prevents ovariectomized-induced bone loss by facilitating osteoblast functions and suppressing osteoclast formation.”

    Osteoporosis is a common bone disease in aging populations, principally in postmenopausal women. Anti-resorptive and anabolic drugs have been applied to prevent and cure osteoporosis and are associated with different adverse effects. Du-Zhong is usually applied in Traditional Chinese Medicine to strengthen bone, regulate bone metabolism, and treat osteoporosis. Chlorogenic acid is a major polyphenol in Du-Zhong.

    In this new study, researchers Chien-Yi Ho, Chih-Hsin Tang, Trung-Loc Ho, Wen-Ling Wang, and Chun-Hsu Yao from China Medical University, China Medical University Hospital and Asia University found chlorogenic acid to enhance osteoblast proliferation and differentiation. Chlorogenic acid also inhibited RANKL-induced osteoclastogenesis. Notably, ovariectomy significantly decreased bone volume and mechanical properties in the ovariectomized (OVX) rats. Administration of chlorogenic acid antagonized OVX-induced bone loss.

    “Taken together, chlorogenic acid seems to be a hopeful molecule for the development of novel anti-osteoporosis treatment.”

    DOI - https://doi.org/10.18632/aging.205635

    Corresponding authors - Wen-Ling Wang - [email protected], and Chun-Hsu Yao - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205635

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, chlorogenic acid, osteoporosis, ovariectomized, osteoclast, osteoblast

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Aging is indexed by PubMed/Medline (abbreviated as “Aging (Albany NY)”), PubMed Central, Web of Science: Science Citation Index Expanded (abbreviated as “Aging‐US” and listed in the Cell Biology and Geriatrics & Gerontology categories), Scopus (abbreviated as “Aging” and listed in the Cell Biology and Aging categories), Biological Abstracts, BIOSIS Previews, EMBASE, META (Chan Zuckerberg Initiative) (2018-2022), and Dimensions (Digital Science).

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    Media Contact
    18009220957
    [email protected]

  • The human brain is a complex organ, and its aging process is influenced by a plethora of factors, both genetic and environmental. Aging-related changes in the brain can lead to cognitive decline and susceptibility to neurodegenerative diseases. Therefore, understanding the molecular mechanisms underlying these changes is crucial for developing therapeutic strategies to delay or prevent age-related cognitive decline.

    Over the past few years, a myriad of scientific studies have been conducted to understand the intricate relationship between our genes and the aging process. In a new study, researchers Joseph A. Zarrella and Amy Tsurumi from Harvard T.H. Chan School of Public Health, Massachusetts General Hospital, Harvard Medical School, and Shriner’s Hospitals for Children-Boston explored the concept of genome brain age prediction, a groundbreaking area of study that employs advanced bioinformatics tools to analyze changes in gene expression associated with aging. On February 28, 2024, their research paper was published and chosen as the cover paper for Aging’s Volume 16, Issue 5, entitled, “Genome-wide transcriptome profiling and development of age prediction models in the human brain.”

    “[…] we aimed to profile transcriptome changes in the aging PFC [prefrontal cortex] overall and compare females and males, and develop prediction models for age.”

    Full blog - https://aging-us.org/2024/03/predicting-brain-age-with-machine-learning-and-transcriptome-profiling/

    Paper DOI - https://doi.org/10.18632/aging.205609

    Corresponding author - Amy Tsurumi - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205609

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, machine learning, prediction model, biomarker, transcriptome

    About Aging-US

    Launched in 2009, Aging-US publishes papers of general interest and biological significance in all fields of aging research and age-related diseases, including cancer—and now, with a special focus on COVID-19 vulnerability as an age-dependent syndrome. Topics in Aging-US go beyond traditional gerontology, including, but not limited to, cellular and molecular biology, human age-related diseases, pathology in model organisms, signal transduction pathways (e.g., p53, sirtuins, and PI-3K/AKT/mTOR, among others), and approaches to modulating these signaling pathways.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    Media Contact
    18009220957
    [email protected]

  • BUFFALO, NY- March 20, 2024 – A new #research paper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 5, entitled, “FoxO6-mediated ApoC3 upregulation promotes hepatic steatosis and hyperlipidemia in aged rats fed a high-fat diet.”

    FoxO6, an identified factor, induces hyperlipidemia and hepatic steatosis during aging by activating hepatic lipoprotein secretion and lipogenesis leading to increased ApoC3 concentrations in the bloodstream. However, the intricate mechanisms underlying hepatic steatosis induced by elevated FoxO6 under hyperglycemic conditions remain intricate and require further elucidation.

    In this new study, researchers Dae Hyun Kim, Seulah Lee, Sang Gyun Noh, Jaewon Lee, and Hae Young Chung from Pusan National University aimed to delineate the regulatory pathway involving ApoC3 controlled by FoxO6 and its resultant functional impacts.

    “[...] we employed a spectrum of models including liver cell cultures, aged rats subjected to HFD, transgenic mice overexpressing FoxO6 (FoxO6-Tg), and FoxO6 knockout mice (FoxO6-KO).”

    Their findings indicate that FoxO6 triggered ApoC3-driven lipid accumulation in the livers of aged rats on an HFD and in FoxO6-Tg, consequently leading to hepatic steatosis and hyperglycemia. Conversely, the absence of FoxO6 attenuated the expression of genes involved in lipogenesis, resulting in diminished hepatic lipid accumulation and mitigated hyperlipidemia in murine models. Additionally, the upregulation of FoxO6 due to elevated glucose levels led to increased ApoC3 expression, consequently instigating cellular triglyceride mediated lipid accumulation. The transcriptional activation of FoxO6 induced by both the HFD and high glucose levels resulted in hepatic steatosis by upregulating ApoC3 and genes associated with gluconeogenesis in aged rats and liver cell cultures.

    “Our conclusions indicate that the upregulation of ApoC3 by FoxO6 promotes the development of hyperlipidemia, hyperglycemia, and hepatic steatosis in vivo, and in vitro. Taken together, our findings underscore the significance of FoxO6 in driving hyperlipidemia and hepatic steatosis specifically under hyperglycemic states by enhancing the expression of ApoC3 in aged rats.”

    DOI - https://doi.org/10.18632/aging.205610

    Corresponding author - Hae Young Chung - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205610

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, HFD-feeding aging, forkhead transcription factor O6, ApoC3, lipid accumulation, hepatic steatosis

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    [email protected]

  • BUFFALO, NY- March 19, 2024 – A new #research paper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 5, entitled, “PR55α-controlled protein phosphatase 2A inhibits p16 expression and blocks cellular senescence induction by γ-irradiation.”

    Cellular senescence is a permanent cell cycle arrest that can be triggered by both internal and external genotoxic stressors, such as telomere dysfunction and DNA damage. The execution of senescence is mainly by two pathways, p16/RB and p53/p21, which lead to CDK4/6 inhibition and RB activation to block cell cycle progression. While the regulation of p53/p21 signaling in response to DNA damage and other insults is well-defined, the regulation of the p16/RB pathway in response to various stressors remains poorly understood.

    In this new study, researchers Chitra Palanivel, Lepakshe S. V. Madduri, Ashley L. Hein, Christopher B. Jenkins, Brendan T. Graff, Alison L. Camero, Sumin Zhou, Charles A. Enke, Michel M. Ouellette, and Ying Yan from the University of Nebraska Medical Center report a novel function of PR55α, a regulatory subunit of PP2A Ser/Thr phosphatase, as a potent inhibitor of p16 expression and senescence induction by ionizing radiation (IR), such as γ-rays.

    “During natural aging, there is a gradual accumulation of p16-expressing senescent cells in tissues [76]. To investigate the significance of PR55α in this up-regulation of p16, we compared levels of the p16 and PR55α proteins in a panel of normal tissue specimens derived from young (≤43 y/o) and old (≥68 y/o) donors.”

    The results show that ectopic PR55α expression in normal pancreatic cells inhibits p16 transcription, increases RB phosphorylation, and blocks IR-induced senescence. Conversely, PR55α-knockdown by shRNA in pancreatic cancer cells elevates p16 transcription, reduces RB phosphorylation, and triggers senescence induction after IR. Furthermore, this PR55α function in the regulation of p16 and senescence is p53-independent because it was unaffected by the mutational status of p53. Moreover, PR55α only affects p16 expression but not p14 (ARF) expression, which is also transcribed from the same CDKN2A locus but from an alternative promoter. In normal human tissues, levels of p16 and PR55α proteins were inversely correlated and mutually exclusive.

    “Collectively, these results describe a novel function of PR55α/PP2A in blocking p16/RB signaling and IR-induced cellular senescence.”

    DOI - https://doi.org/10.18632/aging.205619

    Corresponding authors - Michel M. Ouellette - [email protected], and Ying Yan - [email protected]

    About Aging-US:

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Aging is indexed by PubMed/Medline (abbreviated as “Aging (Albany NY)”), PubMed Central, Web of Science: Science Citation Index Expanded (abbreviated as “Aging‐US” and listed in the Cell Biology and Geriatrics & Gerontology categories), Scopus (abbreviated as “Aging” and listed in the Cell Biology and Aging categories), Biological Abstracts, BIOSIS Previews, EMBASE, META (Chan Zuckerberg Initiative) (2018-2022), and Dimensions (Digital Science).

    Please visit our website at https://www.Aging-US.com​​.

    [email protected]

  • BUFFALO, NY- March 15, 2024 – A new #research paper was #published on the #cover of Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 5, entitled, “Genome-wide transcriptome profiling and development of age prediction models in the human brain.”

    Aging-related transcriptome changes in various regions of the healthy human brain have been explored in previous works, however, a study to develop prediction models for age based on the expression levels of specific panels of transcripts is lacking. Moreover, studies that have assessed sexually dimorphic gene activities in the aging brain have reported discrepant results, suggesting that additional studies would be advantageous. The prefrontal cortex (PFC) region was previously shown to have a particularly large number of significant transcriptome alterations during healthy aging in a study that compared different regions in the human brain.

    In this new study, researchers Joseph A. Zarrella and Amy Tsurumi from the Harvard T.H. Chan School of Public Health, Massachusetts General Hospital, Harvard Medical School, and Shriner's Hospitals for Children-Boston aimed to profile PFC transcriptome changes during healthy human aging overall and comparing potential differences between female and male samples, as well as developing chronological age prediction models by various methods.

    “We harmonized neuropathologically normal PFC transcriptome datasets obtained from the Gene Expression Omnibus (GEO) repository, ranging in age from 21 to 105 years, and found a large number of differentially regulated transcripts in the old and elderly, compared to young samples overall, and compared female and male-specific expression alterations.”

    The team assessed the genes that were associated with age by employing ontology, pathway, and network analyses. Furthermore, they applied various established (least absolute shrinkage and selection operator (Lasso) and Elastic Net (EN)) and recent (eXtreme Gradient Boosting (XGBoost) and Light Gradient Boosting Machine (LightGBM)) machine learning algorithms to develop accurate prediction models for chronological age and validated them. Studies to further validate these models in other large populations and molecular studies to elucidate the potential mechanisms by which the transcripts identified may be related to aging phenotypes would be advantageous.

    “Our results support the notions that specific gene expression changes in the PFC are highly correlated with age, that some transcripts show female and male-specific differences, and that machine learning algorithms are useful tools for developing prediction models for age based on transcriptome information.”

    DOI - https://doi.org/10.18632/aging.205609

    Corresponding author - Amy Tsurumi - [email protected]

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    About Aging-US

    Launched in 2009, Aging-US publishes papers of general interest and biological significance in all fields of aging research and age-related diseases, including cancer—and now, with a special focus on COVID-19 vulnerability as an age-dependent syndrome. Topics in Aging-US go beyond traditional gerontology, including, but not limited to, cellular and molecular biology, human age-related diseases, pathology in model organisms, signal transduction pathways (e.g., p53, sirtuins, and PI-3K/AKT/mTOR, among others), and approaches to modulating these signaling pathways.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    [email protected]

  • Dr. Azra Frkatović-Hodžić from Genos Glycoscience Research Laboratory in Zagreb, Croatia, discusses a #research paper she co-authored that was #published by Aging (Aging-US) in Volume 15, Issue 24, entitled, “Mapping of the gene network that regulates glycan clock of ageing.”

    DOI - https://doi.org/10.18632/aging.205106

    Corresponding authors - Azra Frkatović-Hodžić - [email protected], and Gordan Lauc - [email protected]

    Video - https://www.youtube.com/watch?v=5ExLCMDhpdE

    Video transcription - https://aging-us.net/2024/03/13/behind-the-study-mapping-of-gene-network-that-regulates-glycan-clock-of-aging/

    Abstract

    Glycans are an essential structural component of immunoglobulin G (IgG) that modulate its structure and function. However, regulatory mechanisms behind this complex posttranslational modification are not well known. Previous genome-wide association studies (GWAS) identified 29 genomic regions involved in regulation of IgG glycosylation, but only a few were functionally validated. One of the key functional features of IgG glycosylation is the addition of galactose (galactosylation), a trait which was shown to be associated with ageing. We performed GWAS of IgG galactosylation (N=13,705) and identified 16 significantly associated loci, indicating that IgG galactosylation is regulated by a complex network of genes that extends beyond the galactosyltransferase enzyme that adds galactose to IgG glycans. Gene prioritization identified 37 candidate genes. Using a recently developed CRISPR/dCas9 system we manipulated gene expression of candidate genes in the in vitro IgG expression system. Upregulation of three genes, EEF1A1, MANBA and TNFRSF13B, changed the IgG glycome composition, which confirmed that these three genes are involved in IgG galactosylation in this in vitro expression system.

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205106

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, genome-wide association study, glycosylation, glycan clock, immunoglobulin G, CRISPR/dCas9

    About Aging-US

    Launched in 2009, Aging-US publishes papers of general interest and biological significance in all fields of aging research and age-related diseases, including cancer—and now, with a special focus on COVID-19 vulnerability as an age-dependent syndrome. Topics in Aging-US go beyond traditional gerontology, including, but not limited to, cellular and molecular biology, human age-related diseases, pathology in model organisms, signal transduction pathways (e.g., p53, sirtuins, and PI-3K/AKT/mTOR, among others), and approaches to modulating these signaling pathways.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    SoundCloud - https://soundcloud.com/Aging-Us
    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/

    Media Contact
    18009220957
    [email protected]

  • BUFFALO, NY- March 13, 2024 – A new #research paper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 4, entitled, “Single-Cell RNA-seq reveals transcriptomic modulation of Alzheimer’s disease by activated protein C.”

    Single-Cell RNA sequencing reveals changes in cell population in Alzheimer’s disease (AD) model 5xFAD (5x Familial AD mutation) versus wild type (WT) mice. In this new study, researchers Mohammad Kasim Fatmi, Hao Wang, Lily Slotabec, Changhong Wen, Blaise Seale, Bi Zhao, and Ji Li from the University of South Florida, University of Mississippi Medical Center and the G.V. (Sonny) Montgomery VA Medical Center used single-cell RNA sequencing and bioinformatic analysis to analyze the effects of APC [Activated Protein C] treatment on AD transgenic mice.

    “In our investigation, we utilized transgenic mice that contain expression for five major amyloid pathologies that allow for rapid progression of AD and Aβ deposition known as 5xFAD mice.”

    The returned sequencing data was processed through the 10x Genomics CellRanger platform to perform alignment and form corresponding matrix to perform bioinformatic analysis. Alterations in glial cells occurred in 5xFAD versus WT, especially increases in microglia proliferation were profound in 5xFAD. Differential expression testing of glial cells in 5xFAD versus WT revealed gene regulation. Globally, the critical genes implicated in AD progression are upregulated such as Apoe, Ctsb, Trem2, and Tyrobp.

    Using this differential expression data, GO term enrichment was completed to observe possible biological processes impacted by AD progression. Utilizing anti-inflammatory and cyto-protective recombinant Activated Protein C (APC), the researchers uncovered inflammatory processes to be downregulated by APC treatment in addition to recuperation of nervous system processes. Moreover, animal studies demonstrated that administration of recombinant APC significantly attenuated Aβ burden and improved cognitive function of 5xFAD mice.

    “The downregulation of highly expressed AD biomarkers in 5xFAD could provide insight into the mechanisms by which APC administration benefits AD.”

    DOI - https://doi.org/10.18632/aging.205624

    Corresponding authors - Bi Zhao - [email protected], and Ji Li - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205624

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, APC, Alzheimer’s disease, inflammation

    About Aging-US

    Launched in 2009, Aging-US publishes papers of general interest and biological significance in all fields of aging research and age-related diseases, including cancer—and now, with a special focus on COVID-19 vulnerability as an age-dependent syndrome. Topics in Aging-US go beyond traditional gerontology, including, but not limited to, cellular and molecular biology, human age-related diseases, pathology in model organisms, signal transduction pathways (e.g., p53, sirtuins, and PI-3K/AKT/mTOR, among others), and approaches to modulating these signaling pathways.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    Media Contact
    18009220957
    [email protected]

  • BUFFALO, NY- March 12, 2024 – A new #research paper was #published in Aging (listed by MEDLINE/PubMed as "Aging (Albany NY)" and "Aging-US" by Web of Science) Volume 16, Issue 4, entitled, “Associations of prenatal one-carbon metabolism nutrients and metals with epigenetic aging biomarkers at birth and in childhood in a US cohort.”

    Epigenetic gestational age acceleration (EGAA) at birth and epigenetic age acceleration (EAA) in childhood may be biomarkers of the intrauterine environment. In this new study, researchers Anne K. Bozack, Sheryl L. Rifas-Shiman, Andrea A. Baccarelli, Robert O. Wright, Diane R. Gold, Emily Oken, Marie-France Hivert, and Andres Cardenas from Stanford University School of Medicine, Harvard Medical School, Harvard T.H. Chan School of Public Health, Columbia University, and Icahn School of Medicine at Mount Sinai investigated the extent to which first-trimester folate, B12, 5 essential and 7 non-essential metals in maternal circulation are associated with EGAA and EAA in early life.

    “[...] we hypothesized that OCM [one-carbon metabolism] nutrients and essential metals would be positively associated with EGAA and non-essential metals would be negatively associated with EGAA. We also investigated nonlinear associations and associations with mixtures of micronutrients and metals.”

    Bohlin EGAA and Horvath pan-tissue and skin and blood EAA were calculated using DNA methylation measured in cord blood (N=351) and mid-childhood blood (N=326; median age = 7.7 years) in the Project Viva pre-birth cohort. A one standard deviation increase in individual essential metals (copper, manganese, and zinc) was associated with 0.94-1.2 weeks lower Horvath EAA at birth, and patterns of exposures identified by exploratory factor analysis suggested that a common source of essential metals was associated with Horvath EAA. The researchers also observed evidence of nonlinear associations of zinc with Bohlin EGAA, magnesium and lead with Horvath EAA, and cesium with skin and blood EAA at birth. Overall, associations at birth did not persist in mid-childhood; however, arsenic was associated with greater EAA at birth and in childhood.

    “Prenatal metals, including essential metals and arsenic, are associated with epigenetic aging in early life, which might be associated with future health.”

    DOI - https://doi.org/10.18632/aging.205602

    Corresponding author - Andres Cardenas - [email protected]

    Sign up for free Altmetric alerts about this article -
    https://aging.altmetric.com/details/email_updates?id=10.18632%2Faging.205602

    Subscribe for free publication alerts from Aging - https://www.aging-us.com/subscribe-to-toc-alerts

    Keywords - aging, epigenetic age acceleration, metals, folate, B12, prenatal exposures

    About Aging-US

    Aging publishes research papers in all fields of aging research including but not limited, aging from yeast to mammals, cellular senescence, age-related diseases such as cancer and Alzheimer’s diseases and their prevention and treatment, anti-aging strategies and drug development and especially the role of signal transduction pathways such as mTOR in aging and potential approaches to modulate these signaling pathways to extend lifespan. The journal aims to promote treatment of age-related diseases by slowing down aging, validation of anti-aging drugs by treating age-related diseases, prevention of cancer by inhibiting aging. Cancer and COVID-19 are age-related diseases.

    Please visit our website at https://www.Aging-US.com​​ and connect with us:

    Facebook - https://www.facebook.com/AgingUS/
    X - https://twitter.com/AgingJrnl
    Instagram - https://www.instagram.com/agingjrnl/
    YouTube - https://www.youtube.com/@AgingJournal
    LinkedIn - https://www.linkedin.com/company/aging/
    Pinterest - https://www.pinterest.com/AgingUS/
    Spotify - https://open.spotify.com/show/1X4HQQgegjReaf6Mozn6Mc

    [email protected]