Category Archives: Stell Cell Research

Mitophagy in human health, ageing and disease – Nature.com

Palikaras, K., Lionaki, E. & Tavernarakis, N. Mechanisms of mitophagy in cellular homeostasis, physiology and pathology. Nat. Cell Biol. 20, 10131022 (2018).

Article CAS PubMed Google Scholar

Palikaras, K., Lionaki, E. & Tavernarakis, N. Coordination of mitophagy and mitochondrial biogenesis during ageing in C. elegans. Nature 521, 525528 (2015).

Article CAS PubMed Google Scholar

Lpez-Otn, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. Hallmarks of aging: an expanding universe. Cell 186, 243278 (2023).

Article PubMed Google Scholar

Lpez-Otn, C. & Kroemer, G. Hallmarks of health. Cell 184, 3363 (2021).

Article PubMed Google Scholar

Esteban-Martnez, L. et al. Programmed mitophagy is essential for the glycolytic switch during cell differentiation. EMBO J. 36, 16881706 (2017).

Article PubMed PubMed Central Google Scholar

Sandoval, H. et al. Essential role for Nix in autophagic maturation of erythroid cells. Nature 454, 232235 (2008).

Article CAS PubMed PubMed Central Google Scholar

McWilliams, T. G. et al. mito-QC illuminates mitophagy and mitochondrial architecture in vivo. J. Cell Biol. 214, 333345 (2016).

Article CAS PubMed PubMed Central Google Scholar

Sun, N. et al. Measuring in vivo mitophagy. Mol. Cell 60, 685696 (2015).

Article CAS PubMed PubMed Central Google Scholar

Sekine, S. & Youle, R. J. PINK1 import regulation; a fine system to convey mitochondrial stress to the cytosol. BMC Biol. 16, 2 (2018).

Article PubMed PubMed Central Google Scholar

Tereak, P. et al. Regulation of PRKN-independent mitophagy. Autophagy 18, 2439 (2022).

Article PubMed Google Scholar

Lazarou, M. et al. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature 524, 309314 (2015).

Article CAS PubMed PubMed Central Google Scholar

Zhang, T. et al. BNIP3 protein suppresses PINK1 kinase proteolytic cleavage to promote mitophagy. J. Biol. Chem. 291, 2161621629 (2016).

Article CAS PubMed PubMed Central Google Scholar

Lee, Y., Lee, H.-Y., Hanna, R. A. & Gustafsson, . B. Mitochondrial autophagy by Bnip3 involves Drp1-mediated mitochondrial fission and recruitment of parkin in cardiac myocytes. Am. J. Physiol. Heart Circ. Physiol. 301, H1924H1931 (2011).

Article CAS PubMed PubMed Central Google Scholar

Lou, G. et al. Mitophagy and neuroprotection. Trends Mol. Med. 26, 820 (2020).

Article CAS PubMed Google Scholar

Soubannier, V., Rippstein, P., Kaufman, B. A., Shoubridge, E. A. & McBride, H. M. Reconstitution of mitochondria derived vesicle formation demonstrates selective enrichment of oxidized cargo. PloS ONE 7, e52830 (2012).

Article CAS PubMed PubMed Central Google Scholar

Roberts, R. F., Tang, M. Y., Fon, E. A. & Durcan, T. M. Defending the mitochondria: the pathways of mitophagy and mitochondrial-derived vesicles. Int. J. Biochem. Cell Biol. 79, 427436 (2016).

Article CAS PubMed Google Scholar

Soubannier, V. et al. A vesicular transport pathway shuttles cargo from mitochondria to lysosomes. Curr. Biol. 22, 135141 (2012).

Article CAS PubMed Google Scholar

McLelland, G.-L., Soubannier, V., Chen, C. X., McBride, H. M. & Fon, E. A. Parkin and PINK1 function in a vesicular trafficking pathway regulating mitochondrial quality control. EMBO J. 33, 282295 (2014).

CAS PubMed PubMed Central Google Scholar

Nicols-vila, J. A. et al. A network of macrophages supports mitochondrial homeostasis in the heart. Cell 183, 94109 (2020).

Article PubMed Google Scholar

Liang, W. et al. Mitochondria are secreted in extracellular vesicles when lysosomal function is impaired. Nat. Commun. 14, 5031 (2023).

Article CAS PubMed PubMed Central Google Scholar

Rosina, M. et al. Ejection of damaged mitochondria and their removal by macrophages ensure efficient thermogenesis in brown adipose tissue. Cell Metab. 34, 533548 (2022).

Article CAS PubMed PubMed Central Google Scholar

Davis, C. O. et al. Transcellular degradation of axonal mitochondria. Proc. Natl Acad. Sci. USA 111, 96339638 (2014).

Article CAS PubMed PubMed Central Google Scholar

Melentijevic, I. et al. C. elegans neurons jettison protein aggregates and mitochondria under neurotoxic stress. Nature 542, 367371 (2017).

Hao, T. et al. Hypoxia-reprogramed megamitochondrion contacts and engulfs lysosome to mediate mitochondrial self-digestion. Nat. Commun. 14, 4105 (2023).

Article CAS PubMed PubMed Central Google Scholar

Wu, W. et al. FUNDC1 regulates mitochondrial dynamics at the ERmitochondrial contact site under hypoxic conditions. EMBO J. 35, 13681384 (2016).

Article CAS PubMed PubMed Central Google Scholar

Pryde, K. R., Smith, H. L., Chau, K.-Y. & Schapira, A. H. V. PINK1 disables the anti-fission machinery to segregate damaged mitochondria for mitophagy. J. Cell Biol. 213, 163171 (2016).

Article CAS PubMed PubMed Central Google Scholar

Oshima, Y. et al. Parkin-independent mitophagy via Drp1-mediated outer membrane severing and inner membrane ubiquitination. J. Cell Biol. 220, e202006043 (2021).

Article CAS PubMed PubMed Central Google Scholar

Munson, M. J. et al. GAK and PRKCD are positive regulators of PRKN-independent mitophagy. Nat. Commun. 12, 6101 (2021).

Article CAS PubMed PubMed Central Google Scholar

Gegg, M. E. et al. Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/parkin-dependent manner upon induction of mitophagy. Hum. Mol. Genet. 19, 48614870 (2010).

Article CAS PubMed PubMed Central Google Scholar

Ziviani, E. & Whitworth, A. J. How could parkin-mediated ubiquitination of mitofusin promote mitophagy? Autophagy 6, 660662 (2010).

Article PubMed Google Scholar

Palikaras, K. & Tavernarakis, N. Mitochondrial homeostasis: the interplay between mitophagy and mitochondrial biogenesis. Exp. Gerontol. 56, 182188 (2014).

Article CAS PubMed Google Scholar

Cant, C. & Auwerx, J. PGC-1, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr. Opin. Lipidol. 20, 98105 (2009).

Article PubMed PubMed Central Google Scholar

Malik, N. et al. Induction of lysosomal and mitochondrial biogenesis by AMPK phosphorylation of FNIP1. Science 380, eabj5559 (2023).

Article CAS PubMed Google Scholar

Lionaki, E., Markaki, M., Palikaras, K. & Tavernarakis, N. Mitochondria, autophagy and age-associated neurodegenerative diseases: new insights into a complex interplay. Biochim. Biophys. Acta 1847, 14121423 (2015).

Article CAS PubMed Google Scholar

Laker, R. C. et al. Ampk phosphorylation of Ulk1 is required for targeting of mitochondria to lysosomes in exercise-induced mitophagy. Nat. Commun. 8, 548 (2017).

Article PubMed PubMed Central Google Scholar

Iorio, R., Celenza, G. & Petricca, S. Mitophagy: molecular mechanisms, new concepts on parkin activation and the emerging role of AMPK/ULK1 axis. Cells 11, 30 (2021).

Article PubMed PubMed Central Google Scholar

DAmico, D. et al. The RNA-binding protein PUM2 impairs mitochondrial dynamics and mitophagy during aging. Mol. Cell 73, 775787 (2019).

Article PubMed PubMed Central Google Scholar

Shin, H. J. et al. Pink1-mediated chondrocytic mitophagy contributes to cartilage degeneration in osteoarthritis. J. Clin. Med. 8, 1849 (2019).

Article CAS PubMed PubMed Central Google Scholar

Kuroda, Y. et al. Parkin enhances mitochondrial biogenesis in proliferating cells. Hum. Mol. Genet. 15, 883895 (2006).

Article CAS PubMed Google Scholar

Egan, B. & Zierath, J. R. Exercise metabolism and the molecular regulation of skeletal muscle adaptation. Cell Metab. 17, 162184 (2013).

Article CAS PubMed Google Scholar

Gaitanos, G. C., Williams, C., Boobis, L. H. & Brooks, S. Human muscle metabolism during intermittent maximal exercise. J. Appl. Physiol. 75, 712719 (1993).

Article CAS PubMed Google Scholar

Sin, J. et al. Mitophagy is required for mitochondrial biogenesis and myogenic differentiation of C2C12 myoblasts. Autophagy 12, 369380 (2016).

Article CAS PubMed Google Scholar

Hong, X. et al. Mitochondrial dynamics maintain muscle stem cell regenerative competence throughout adult life by regulating metabolism and mitophagy. Cell Stem Cell 29, 12981314 (2022).

CAS Google Scholar

Leduc-Gaudet, J.-P. et al. Parkin overexpression attenuates sepsis-induced muscle wasting. Cells 9, 1454 (2020).

Article CAS PubMed PubMed Central Google Scholar

Leduc-Gaudet, J.-P. et al. Mitochondrial morphology is altered in atrophied skeletal muscle of aged mice. Oncotarget 6, 1792317937 (2015).

Article PubMed PubMed Central Google Scholar

Garca-Prat, L. et al. Autophagy maintains stemness by preventing senescence. Nature 529, 3742 (2016).

Article PubMed Google Scholar

Luan, P. et al. Urolithin A improves muscle function by inducing mitophagy in muscular dystrophy. Sci. Transl. Med. 13, eabb0319 (2021).

Article CAS PubMed Google Scholar

Ryu, D. et al. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat. Med. 22, 879888 (2016).

Article CAS PubMed Google Scholar

Fang, E. F. et al. Tomatidine enhances lifespan and healthspan in C. elegans through mitophagy induction via the SKN-1/Nrf2 pathway. Sci. Rep. 7, 46208 (2017).

Article CAS PubMed PubMed Central Google Scholar

DAmico, D. et al. Urolithin A improves mitochondrial health, reduces cartilage degeneration, and alleviates pain in osteoarthritis. Aging Cell 21, e13662 (2022).

Article PubMed PubMed Central Google Scholar

Choi, S. et al. 31P magnetic resonance spectroscopy assessment of muscle bioenergetics as a predictor of gait speed in the Baltimore Longitudinal Study of Aging. J. Gerontol. A Biol. Sci. Med. Sci. 71, 16381645 (2016).

Link:
Mitophagy in human health, ageing and disease - Nature.com

From immunology to artificial intelligence: revolutionizing latent … – Military Medical Research

World Health Organization. Global tuberculosis report 2022. Geneva: World Health Organization; 2022. https://www.who.int/teams/global-tuberculosis-programme/tb-reports

Bagcchi S. WHOs global tuberculosis report 2022. Lancet Microbe. 2023;4(1):e20.

Article PubMed Google Scholar

Ernst JD. The immunological life cycle of tuberculosis. Nat Rev Immunol. 2012;12(8):58191.

Article CAS PubMed Google Scholar

Lewinsohn DM, Leonard MK, LoBue PA, Cohn DL, Daley CL, Desmond E, et al. Official American Thoracic Society/Infectious Diseases Society of America/Centers for Disease Control and Prevention Clinical Practice Guidelines: diagnosis of tuberculosis in adults and children. Clin Infect Dis. 2017;64(2):e1-33.

Article PubMed Google Scholar

Cohen A, Mathiasen VD, Schon T, Wejse C. The global prevalence of latent tuberculosis: a systematic review and meta-analysis. Eur Respir J. 2019;54(3):1900655.

Article PubMed Google Scholar

Khabibullina NF, Kutuzova DM, Burmistrova IA, Lyadova IV. The biological and clinical aspects of a latent tuberculosis infection. Trop Med Infect Dis. 2022;7(3):48.

Article PubMed PubMed Central Google Scholar

Houben RMGJ, Dodd PJ. The global burden of latent tuberculosis infection: a re-estimation using mathematical modelling. PLoS Med. 2016;13(10):e1002152.

Article PubMed PubMed Central Google Scholar

Jilani TN, Avula A, Zafar Gondal A, Siddiqui AH. Active tuberculosis. In: StatPearls. Treasure Island (FL): StatPearls Publishing LLC; 2023.

Ding C, Hu M, Guo W, Hu W, Li X, Wang S, et al. Prevalence trends of latent tuberculosis infection at the global, regional, and country levels from 19902019. Int J Infect Dis. 2022;122:46.

Article PubMed Google Scholar

Kiazyk S, Ball TB. Latent tuberculosis infection: an overview. Can Commun Dis Rep. 2017;43(34):626.

Article CAS PubMed PubMed Central Google Scholar

Luo Y, Xue Y, Song H, Tang G, Liu W, Bai H, et al. Machine learning based on routine laboratory indicators promoting the discrimination between active tuberculosis and latent tuberculosis infection. J Infect. 2022;84(5):64857.

Article PubMed Google Scholar

Estvez O, Anibarro L, Garet E, Pallares , Barcia L, Calvio L, et al. An RNA-seq based machine learning approach identifies latent tuberculosis patients with an active tuberculosis profile. Front Immunol. 2020;11:1470.

Article PubMed PubMed Central Google Scholar

Gong W, Wu X. Differential diagnosis of latent tuberculosis infection and active tuberculosis: a key to a successful tuberculosis control strategy. Front Microbiol. 2021;12(3126):745592.

Article PubMed PubMed Central Google Scholar

Chee CBE, Reves R, Zhang Y, Belknap R. Latent tuberculosis infection: opportunities and challenges. Respirology. 2018;23(10):893900.

Article PubMed Google Scholar

Hauck FR, Neese BH, Panchal AS, El-Amin W. Identification and management of latent tuberculosis infection. Am Fam Physician. 2009;79(10):87986.

PubMed Google Scholar

Gutti G, Arya K, Singh SK. Latent tuberculosis infection (LTBI) and its potential targets: an investigation into dormant phase pathogens. Mini Rev Med Chem. 2019;19(19):162742.

Article CAS PubMed Google Scholar

Yang Z, Rosenthal M, Rosenberg NA, Talarico S, Zhang L, Marrs C, et al. How dormant is Mycobacterium tuberculosis during latency? A study integrating genomics and molecular epidemiology. Infect Genet Evol. 2011;11(5):11647.

Article PubMed PubMed Central Google Scholar

Gordon SV, Eiglmeier K, Garnier T, Brosch R, Parkhill J, Barrell B, et al. Genomics of Mycobacterium bovis. Tuberculosis. 2001;81(12):15763.

Article CAS PubMed Google Scholar

Chen J, Su X, Zhang Y, Wang S, Shao L, Wu J, et al. Novel recombinant RD2- and RD11-encoded Mycobacterium tuberculosis antigens are potential candidates for diagnosis of tuberculosis infections in BCG-vaccinated individuals. Microbes Infect. 2009;11(1011):87685.

Article CAS PubMed Google Scholar

Meier NR, Jacobsen M, Ottenhoff THM, Ritz N. A systematic review on novel Mycobacterium tuberculosis antigens and their discriminatory potential for the diagnosis of latent and active tuberculosis. Front Immunol. 2018;9:2476.

Article PubMed PubMed Central Google Scholar

Ji P, Fan X, Wu K, Lu S. Research progress on the antigens associated with latent infection of Mycobacterium tuberculosis. Zhonghua Wei Sheng Wu Xue He Mian Yi Xue Za Zhi. 2015;35(1):5964 (in Chinese).

CAS Google Scholar

Zellweger JP, Sotgiu G, Corradi M, Durando P. The diagnosis of latent tuberculosis infection (LTBI): currently available tests, future developments, and perspectives to eliminate tuberculosis (TB). Med Lav. 2020;111(3):17083.

PubMed PubMed Central Google Scholar

Crouser ED, White P, Caceres EG, Julian MW, Papp AC, Locke LW, et al. A novel in vitro human granuloma model of sarcoidosis and latent tuberculosis infection. Am J Respir Cell Mol Biol. 2017;57(4):48798.

Article CAS PubMed PubMed Central Google Scholar

Rosser A, Stover C, Pareek M, Mukamolova GV. Resuscitation-promoting factors are important determinants of the pathophysiology in Mycobacterium tuberculosis infection. Crit Rev Microbiol. 2017;43(5):62130.

Article CAS PubMed Google Scholar

Downing KJ, Mischenko VV, Shleeva MO, Young DI, Young M, Kaprelyants AS, et al. Mutants of Mycobacterium tuberculosis lacking three of the five rpf-like genes are defective for growth in vivo and for resuscitation in vitro. Infect Immun. 2005;73(5):303843.

Article CAS PubMed PubMed Central Google Scholar

Arroyo L, Marn D, Franken KLMC, Ottenhoff THM, Barrera LF. Potential of DosR and Rpf antigens from Mycobacterium tuberculosis to discriminate between latent and active tuberculosis in a tuberculosis endemic population of Medellin Colombia. BMC Infect Dis. 2018;18(1):26.

Article PubMed PubMed Central Google Scholar

Zhu W, Plikaytis BB, Shinnick TM. Resuscitation factors from mycobacteria: homologs of Micrococcus luteus proteins. Tuberculosis. 2003;83(4):2619.

Article PubMed Google Scholar

Cohen-Gonsaud M, Barthe P, Bagnris C, Henderson B, Ward J, Roumestand C, et al. The structure of a resuscitation-promoting factor domain from Mycobacterium tuberculosis shows homology to lysozymes. Nat Struct Mol Biol. 2005;12(3):2703.

Article CAS PubMed Google Scholar

Segueni N, Benmerzoug S, Rose S, Gauthier A, Bourigault ML, Reverchon F, et al. Innate myeloid cell TNFR1 mediates first line defence against primary Mycobacterium tuberculosis infection. Sci Rep. 2016;6:22454.

Article CAS PubMed PubMed Central Google Scholar

Koeken V, Verrall AJ, Netea MG, Hill PC, van Crevel R. Trained innate immunity and resistance to Mycobacterium tuberculosis infection. Clin Microbiol Infect. 2019;25(12):146872.

Article CAS PubMed Google Scholar

Cadena AM, Flynn JL, Fortune SM. The importance of first impressions: early events in Mycobacterium tuberculosis infection influence Outcome. MBio. 2016;7(2):e00342-e416.

Article CAS PubMed PubMed Central Google Scholar

McClean CM, Tobin DM. Macrophage form, function, and phenotype in mycobacterial infection: lessons from tuberculosis and other diseases. Pathog Dis. 2016;74(7):ftw068.

Article PubMed PubMed Central Google Scholar

Hmama Z, Pea-Daz S, Joseph S, Av-Gay Y. Immunoevasion and immunosuppression of the macrophage by Mycobacterium tuberculosis. Immunol Rev. 2015;264(1):22032.

Article CAS PubMed Google Scholar

Middleton AM, Chadwick MV, Nicholson AG, Dewar A, Groger RK, Brown EJ, et al. Interaction of Mycobacterium tuberculosis with human respiratory mucosa. Tuberculosis. 2002;82(23):6978.

Article CAS PubMed Google Scholar

Peyron P, Vaubourgeix J, Poquet Y, Levillain F, Botanch C, Bardou F, et al. Foamy macrophages from tuberculous patients granulomas constitute a nutrient-rich reservoir for M tuberculosis persistence. PLoS Pathog. 2008;4(11):e1000204.

Article PubMed PubMed Central Google Scholar

Mattila JT, Ojo OO, Kepka-Lenhart D, Marino S, Kim JH, Eum SY, et al. Microenvironments in tuberculous granulomas are delineated by distinct populations of macrophage subsets and expression of nitric oxide synthase and arginase isoforms. J Immunol. 2013;191(2):77384.

Article CAS PubMed Google Scholar

El Kasmi KC, Qualls JE, Pesce JT, Smith AM, Thompson RW, Henao-Tamayo M, et al. Toll-like receptor-induced arginase 1 in macrophages thwarts effective immunity against intracellular pathogens. Nat Immunol. 2008;9(12):1399406.

Article PubMed PubMed Central Google Scholar

Duque-Correa MA, Kuhl AA, Rodriguez PC, Zedler U, Schommer-Leitner S, Rao M, et al. Macrophage arginase-1 controls bacterial growth and pathology in hypoxic tuberculosis granulomas. Proc Natl Acad Sci U S A. 2014;111(38):E402432.

Article CAS PubMed PubMed Central Google Scholar

Khan A, Hunter RL, Jagannath C. Emerging role of mesenchymal stem cells during tuberculosis: the fifth element in cell mediated immunity. Tuberculosis. 2016;101S:S45-52.

Article PubMed Google Scholar

Keane J, Balcewicz-Sablinska MK, Remold HG, Chupp GL, Meek BB, Fenton MJ, et al. Infection by Mycobacterium tuberculosis promotes human alveolar macrophage apoptosis. Infect Immun. 1997;65(1):298304.

Article CAS PubMed PubMed Central Google Scholar

Harding JS, Schreiber HA, Sandor M. Granuloma transplantation: an approach to study Mycobacterium-host interactions. Front Microbiol. 2011;2:245.

Article PubMed PubMed Central Google Scholar

Gaffney E, Murphy D, Walsh A, Connolly S, Basdeo SA, Keane J, et al. Defining the role of neutrophils in the lung during infection: implications for tuberculosis disease. Front Immunol. 2022;13:984293.

Article CAS PubMed PubMed Central Google Scholar

Yang CT, Cambier CJ, Davis JM, Hall CJ, Crosier PS, Ramakrishnan L. Neutrophils exert protection in the early tuberculous granuloma by oxidative killing of mycobacteria phagocytosed from infected macrophages. Cell Host Microbe. 2012;12(3):30112.

Article CAS PubMed PubMed Central Google Scholar

Mantegazza AR, Savina A, Vermeulen M, Perez L, Geffner J, Hermine O, et al. NADPH oxidase controls phagosomal pH and antigen cross-presentation in human dendritic cells. Blood. 2008;112(12):471222.

Article CAS PubMed PubMed Central Google Scholar

Barnes PF, Leedom JM, Chan LS, Wong SF, Shah J, Vachon LA, et al. Predictors of short-term prognosis in patients with pulmonary tuberculosis. J Infect Dis. 1988;158(2):36671.

Article CAS PubMed Google Scholar

Dallenga T, Schaible UE. Neutrophils in tuberculosisfirst line of defence or booster of disease and targets for host-directed therapy?. Pathog Dis. 2016;74(3):ftw012.

Article PubMed Google Scholar

Eruslanov EB, Lyadova IV, Kondratieva TK, Majorov KB, Scheglov IV, Orlova MO, et al. Neutrophil responses to Mycobacterium tuberculosis infection in genetically susceptible and resistant mice. Infect Immun. 2005;73(3):174453.

Article CAS PubMed PubMed Central Google Scholar

Divangahi M, Chen M, Gan H, Desjardins D, Hickman TT, Lee DM, et al. Mycobacterium tuberculosis evades macrophage defenses by inhibiting plasma membrane repair. Nat Immunol. 2009;10(8):899906.

Article CAS PubMed PubMed Central Google Scholar

Ahmad S, Amoudy HA, Thole JE, Young DB, Mustafa AS. Identification of a novel protein antigen encoded by a Mycobacterium tuberculosis-specific RD1 region gene. Scand J Immunol. 1999;49(5):51522.

Article CAS PubMed Google Scholar

Albayrak N, Dirix V, Aerts L, Van Praet A, Godefroid A, Dauby N, et al. Differential expression of maturation and activation markers on NK cells in patients with active and latent tuberculosis. J Leukoc Biol. 2022;111(5):103142.

Article CAS PubMed Google Scholar

Mah AY, Cooper MA. Metabolic regulation of natural killer cell IFN-gamma production. Crit Rev Immunol. 2016;36(2):13147.

Article PubMed PubMed Central Google Scholar

Read the rest here:
From immunology to artificial intelligence: revolutionizing latent ... - Military Medical Research

Investing in healthcare where are the opportunities? – Hargreaves Lansdown

Key takeaways

The healthcare sector covers the huge range of activities needed to deliver medical care globally. Things like diagnostic tools and the wide selection of pharmaceuticals and medical devices used to treat patients.

Then there are research organisations and insurers. This universe also includes privately-run care facilities, as well as medical distributors and pharmacies.

Past performance isnt a guide to the future. Source: Refinitiv Eikon, 26 November 2023.

The pandemic has been hugely disruptive for the healthcare sector. It caused significant delays in clinical trials pipelines and an unprecedented backlog in patient care. Made even worse by difficulties in the supply chain.

So maybe its no surprise that the sector underperformed at the peak of the pandemic. But it also let certain players demonstrate excellence in science innovation. They led the world's response to the crisis through the development of new treatments, and most importantly the rollout of vaccines in record time.

In 2022 this let the sector keep generating positive returns, while the wider market retreated as the global economy saw high inflation and rising interest rates.

More recently though, those companies that saw a boost from tackling COVID-19, have struggled to replace the revenue from falling sales of these same products. That's seen a lot of the initial outperformance reverse in 2023.

One of the biggest stories coming from the healthcare industry is the boom in next-generation medicines for combating obesity and diabetes. These have the potential to become the biggest category of pharmaceuticals in medical history.

For now, this space is dominated by a handful of trailblazers, but theres a swarm of competitors waiting in the wings. This reflects the growing burden of chronic diseases. Tackling these conditions makes commercial sense and potentially creates long-term revenue streams.

We also see an opportunity in the evolution of new treatment mechanisms that can help or potentially cure serious diseases. Despite huge advances in the standard of care, cancer is still responsible for nearly one in six deaths worldwide and is by far still the busiest area for pharmaceutical research.

Immunotherapy, where the body's own immune system is honed to fight cancerous cells has been a key driver of improved survival rates. But there's room to improve its efficacy and increase the number of people and cancers it can be used on.

Gene therapy and stem cell treatments are other breakthroughs keeping the white coats busy, and they could also have use cases in other disease areas.

But it's not all about new products.

Pursuing approvals in new territories, patient populations, and conditions for medicines that already exist in the market makes perfect sense. This is a concept thats become known as a pipeline in a product.

Information and communication technology could also have a transformational effect on healthcare delivery.

The adoption of telemedicine was accelerated by the need for social distancing during the pandemic. It's now relatively common to have a consultation by video call, and we expect this will stay integral to health services going forward.

Healthcare hasn't escaped the hype around artificial intelligence either. There are opportunities in drug discovery, diagnostics and the delivery of patient care, but this is still early days.

Drug development is lengthy and expensive, with a low success rate. Companies that focus on these need to spend a lot of cash, with no guarantee of a return.

And it's important to keep refilling the hopper as eventually the exclusivity you get for taking the initial risk lapses. This paves the way for generic competitors to enter the market. And there's always the chance competitors will develop more effective treatments.

In the near term, pressure on government finances could keep a lid on the level of expenditure committed to the healthcare sector.

The sector is dependent on the availability of highly-skilled doctors, nurses and research scientists. This can hamper the development and delivery of products and services as well as push up costs.

Healthcare is highly regulated and a crucial political battleground. There's growing pressure to bring down prices. Consolidation in the industry is attracting the eye of the competition authorities. And there arent guarantees that certain medicines will be reimbursed by state or private health insurers.

Safety of clinical trials and marketed medicines is another risk to watch. It can take years after a product launch for side effects to become apparent, and this leaves the sector open to legal claims which can be expensive.

While COVID-19 seems to be well managed for now, we can't rule out the emergence of aggressive or vaccine-resistant strains, which could bring huge disruption to the sector.

The healthcare sector has an impressive long-term growth record. A number of drivers are likely to prolong this trend, not least being population growth and increasing life expectancy.

Meanwhile, there's a need to improve patient access and bridge the gap in healthcare coverage between developed and developing nations.

It's also a sector that has significant barriers to entry, which we see as a key attraction for investors.

In recent years, many of the major breakthroughs in medicine have been made by smaller biotech companies. The winners among them have proved very rewarding for investors. But the challenging economic environment has seen the falling appetites to fund these riskier businesses.

We favour companies with good cash flows and strong balance sheets. This lets them build more diverse development pipelines. And also create the infrastructure needed to commercialise their science, meaning they get more of the profits.

It also allows them to take advantage of the funding gap for smaller industry participants, by cherry-picking companies and assets in spaces they think are attractive.

It's certainly an exciting space to be in, but also a complex one. Its important to make sure you understand what youre investing in before diving in and remember, there are no guarantees.

Investments and any income from them can fall as well as rise in value, so you could get back less than you invest. This isnt personal advice if youre not sure whats right for you, seek advice.

To make sure you dont miss out on our latest share sector reviews, sign up to our Share Insight email. Well send you our latest share research and articles every week, direct to your inbox.

This article is not advice or a recommendation to buy, sell or hold any investment. No view is given on the present or future value or price of any investment, and investors should form their own view on any proposed investment. This article has not been prepared in accordance with legal requirements designed to promote the independence of investment research and is considered a marketing communication. Non-independent research is not subject to FCA rules prohibiting dealing ahead of research, however HL has put controls in place (including dealing restrictions, physical and information barriers) to manage potential conflicts of interest presented by such dealing. Please see our full non-independent research disclosure for more information.

Sign up to receive weekly shares content from HL.

Please correct the following errors before you continue:

Hargreaves Lansdown PLC group companies will usually send you further information by post and/or email about our products and services. If you would prefer not to receive this, please do let us know. We will not sell or trade your personal data.

Excerpt from:
Investing in healthcare where are the opportunities? - Hargreaves Lansdown

MetroHealth, Case Western Reserve University Cancer … – Newsroom MetroHealth

Dr. Wang Headshot

MetroHealth and Case Western Reserve University (CWRU) cancer researchers have solved a mystery surrounding a receptor protein that can suppress cancer or make it grow and spread. Their findings, detailing how and why the EphA2 receptor plays the roles of both cancer hero and villain, will be published in the journal Science November 16.

The team of researchers was led by Bingcheng Wang, PhD, Director of the MetroHealth Division of Cancer Biology and MetroHealth Research Institute Director of Basic Sciences.

Discoveries like this make it possible to treat cancer, said Dr. Wang, who also is the John A. and Josephine B. Wootton Endowed Chair of Research and professor at the Case Western Reserve School of Medicine and a member of the Case Comprehensive Cancer Center. As a cancer researcher, there is no greater accomplishment. Being asked to share this work with the scientific community through the prestigious journal Science is an honor. But the greatest reward is to know that we are making strides that will have a real impact on our own patients and others throughout world.

Dr. Wang, who has been studying the EphA2 receptor for 25 years, is recognized as a pioneer in the field. His lab has made several key discoveries around the receptor, which is overexpressed in solid tumors like prostate, breast, colon and lung cancers as well as the aggressive brain tumor glioblastoma.

In two landmark studies published in Nature Cell Biology in 2000 and 2001, Dr. Wangs lab was the first to make the groundbreaking discoveries that the EphA2 can suppress malignant behaviors of cancer cells. In 2009, his team reported in Cancer Cell that the same receptor also can have the opposite function after being modified by tumor-promoting proteins. The modified EphA2 causes cancer cells to proliferate, maintain stem cell properties and metastasize to other parts of the body.

Now, after years of investigation, the researchers have figured out how EphA2 plays these dual, opposing roles in cancer. Using a cutting-edge spectroscopic platform (PIE-FCCS) that allows molecular analysis on live cells, they saw that EphA2 is automatically assembled into small clusters on live cells through two different types of interactions among adjacent EphA2 molecules that glue them together. One interaction contributes to the hero role and the other triggers the villain side of the molecule.

The first author of the paper is Dr. Xiaojun (Roger) Shi, a postdoctoral scholar at the CWRU School of Medicine and a current trainee with the Cancer Biology Training Program of the National Cancer Institute. Roger made the discovery by combining his expertise in molecular imaging during doctoral thesis work and mastery of experimental cancer biology gained in the Wang lab.

As the lead contact author, Dr. Wang shares the findings in the Science article Time-Resolved Live Cell Spectroscopy Reveals EphA2 Multimeric Assembly. A large multidisciplinary team contributed to the work. Dimitar B. Nikolov, of Memorial Sloan Kettering Cancer Center, and Adam W. Smith, of Texas Tech University, are co-corresponding authors of the paper. Khalid Sossey-Alaoui, of MetroHealth and CWRU; Matthias Buck, of CWRU; Ben Brown and Jens Meiler, of Vanderbilt University; and Dolores Hambardzumyan, of Icahn School of Medicine at Mount Sinai, are among the co-authors who contributed to the work. The paper will be published online by the journal Science on Thursday, November 16.

As the inaugural Director of the Division of Cancer Biology in the Department of Medicine, Dr. Wang has played a significant role in MetroHealths strategic vision for research, successfully recruiting several nationally recognized cancer researchers. In 2021, he led the formation of a new Cancer Research Team, funded through millions of dollars in support and grants, to focus on ending the racial, ethnic, social and economic inequities that impact cancer diagnosis and treatment.

We know that many types of cancer disproportionately affect people of color, said MetroHealth President & CEO Airica Steed, Ed.D, RN, MBA, FACHE. This is why we are hyperfocused on eradicating health disparities and will continue to support the cutting-edge research that leads to these discoveries, so eventually all patients who face a diagnosis of cancer can hope for a long life, regardless of their cultural background, where they live or how much money they make.

John Chae, MD, MetroHealth Senior Vice President, Chief Academic Officer, said Dr. Wangs discoveries and other pivotal research being done at MetroHealth are reinforcing the Systems reputation as a world-class research institution.

This is the sort of foundational research that life-saving therapies are built upon, said Dr. Chae, who also is Senior Associate Dean for Medical Affairs at the CWRU School of Medicine. We are fortunate to have internationally respected researchers like Dr. Wang and the incredible team he has assembled. We will go on supporting this research and proving that some of the very best science in the world is being done in Cleveland at The MetroHealth System.

See more here:
MetroHealth, Case Western Reserve University Cancer ... - Newsroom MetroHealth

3D Cell Culture Market to grow by USD 1.28 billion from 2021 to … – PR Newswire

NEW YORK, Nov. 13, 2023 /PRNewswire/ -- The3D cell culture market size is expected to grow by USD 1.28 billion from 2021 to 2026. In addition, the momentum of the market will be progressing at a CAGR of15.69% during the forecast period, according to Technavio Research.The market is segmented by application (Cancer and stem cell research, Drug discovery and toxicology testing, and Tissue engineering and regenerative medicine) and geography (North America, Europe, Asia, and Rest of World (ROW)).The 3D cell culture market share growth by cancer and stem cell research segment will be significant during the forecast period.The rising prevalence of cancer and significant funding for cancer research are significant factors that are anticipated to drive the growth of the segment in focus during the forecast period.This report offers an up-to-date analysis of the current market scenario, the latest trends and drivers, and the overall market environment. Read FREE PDF Sample Report

Company Profile:

3D Biotek LLC, BICO Group AB, CN Bio Innovations Ltd., Corning Inc., Elveflow, Emulate Inc., Greiner Bio-One International GmbH, Hamilton Bonaduz AG, InSphero AG, Lonza Group Ltd., Merck KGaA, PromoCell GmbH, QGel SA, REPROCELL Inc., Synthecon Inc., SynVivoInc., Tecan Group Ltd., Thermo Fisher Scientific Inc., TissUse GmbH, and MIMETAS BV

3D Biotek LLC -The company offers 3D Cell Culture products such as 3D cell culture devices.

To gain access to more vendor profiles available withTechnavio, buy the report

Learn about the contribution of each segment summarized in conciseinfographics and thorough descriptions. View a FREE PDF Sample Report

3D Cell Culture Market: Geographical Analysis

North Americais estimated toaccount for41%of the global market duringthe forecast period. The primary markets for 3D cell culture in North America are the US and Canada. In this region, market growth is expected to outpace that in Europe and the Rest of the World (ROW). This accelerated growth can be attributed to substantial investments in new manufacturing facilities made by major companies like Becton, Dickinson, and Company, Corning Incorporated, and Thermo Fisher Scientific Inc. Such investments are set to drive the expansion of the 3D cell culture market in North America during the forecast period.

3D Cell Culture Market: Driver & Trend:

The increase in infectious diseases is notably driving the market growth.

Identify key trends, drivers, and challenges in the market. Download FREE sample to gain access to this information.

What are the key data covered in this 3D cell culture market report?

Related Reports:

The GlobalCell Culture Marketsize is estimated togrowat aCAGR of 11.3%between 2022 and 2027. The size of the market is forecasted to increase byUSD 17.74 billion.

The cell culture consumables market size is estimated togrowat a CAGR of 22.3%between 2022 and 2027. The size of the cell culture market is forecast to increase byUSD 23,729.7 million.

ToC:

Executive Summary

Market Landscape

Market Sizing

Historic Market Sizes

Five Forces Analysis

Market Segmentation by Application

Market Segmentation by Geography

Customer Landscape

Geographic Landscape

Drivers,Challenges, &Trends

Company Landscape

Company Analysis

Appendix

About Technavio

Technavio is a leading global technology research and advisory company. Their research and analysis focus on emerging market trends and provide actionable insights to help businesses identify market opportunities and develop effective strategies to optimize their market positions.With over 500 specialized analysts, Technavio's report library consists of more than 17,000 reports and counting, covering 800 technologies, spanning across 50 countries. Their client base consists of enterprises of all sizes, including more than 100 Fortune 500 companies. This growing client base relies on Technavio's comprehensive coverage, extensive research, and actionable market insights to identify opportunities in existing and potential markets and assess their competitive positions within changing market scenarios.

Contacts

Technavio Research Jesse Maida Media & Marketing Executive US: +1 844 364 1100 UK: +44 203 893 3200 Email:[emailprotected] Website:www.technavio.com

SOURCE Technavio

See the rest here:
3D Cell Culture Market to grow by USD 1.28 billion from 2021 to ... - PR Newswire

New study reveals the critical role of microglia in human brain … – EurekAlert

image:

Super-resolution image of human stem cell-derived Microglia cells with labeled mitochondria (yellow), nucleus (magenta), and actin filaments (cyan). These Microglia cells help in the maturation of neurons in human brain organoid models. Photo credit: A*STAR's SIgN

Credit: A*STAR's SIgN

An international team of scientists has uncovered the vital role of microglia, the immune cells in the brain that acts as its dedicated defense team, in early human brain development. By incorporating microglia into lab-grown brain organoids, scientists were able mimic the complex environment within the developing human brain to understand how microglia influence brain cell growth and development. This research represents a significant leap forward in the development of human brain organoids and has the potential to significantly impact our understanding of brain development and disorders. The study, iPS-cell-derived microglia promote brain organoid maturation via cholesterol transfer was published in Nature on 1 November 2023.

To investigate microglia's crucial role in early human brain development, scientists from A*STAR's Singapore Immunology Network (SIgN) led by Professor Florent Ginhoux, utilised cutting-edge technology to create brain-like structures called organoids, also known as mini-brains in the laboratory. These brain organoids closely resemble the development of the human brain. However, previous models were lacking in microglia, a key component of early brain development.

To bridge this gap, A*STAR researchers designed a unique protocol to introduce microglia-like cells generated from the same human stem cells used to create the brain organoids. These introduced cells not only behaved like real microglia but also influenced the development of other brain cells within the organoids.

A*STAR's Institute of Molecular and Cell Biology (IMCB)'s Dr Radoslaw Sobota and his team at the SingMass National Laboratory for Mass Spectrometry applied cutting edge quantitative proteomics approach to uncover changes in protein. Their analysis provided crucial insights into the protein composition of the organoids, further confirming the studys findings.

What sets this study apart is the discovery of a unique pathway through which microglia interact with other brain cells. The study found that microglia play a crucial role in regulating cholesterol levels in the brain.The microglia-like cells were found to contain lipid droplets containing cholesterol, which were released and taken up by other developing brain cells in the organoids. This cholesterol exchange was shown to significantly enhance the growth and development of these brain cells, especially their progenitors.

Cholesterol, makes up about 25% of the body's total cholesterol content, is abundantly present in the brain and is essential for the structure and function of neurons. Abnormal cholesterol metabolism has been linked to various neurological disorders, including Alzheimer's and Parkinson's Disease.

To investigate the roles of lipids in brain development and disease, researchers from the Department of Biochemistry at the Yong Loo Lin School of Medicine (NUS Medicine), led by Professor Markus Wenk, took on the crucial task of data acquisition, particularly in the field of lipidomics to draw valuable insights into the lipid composition and dynamics within the brain organoids containing microglia.

Using this information, another team from the Department of Microbiology and Immunology at NUS Medicine and led by Associate Professor Veronique Angeli, found that cholesterol affects the growth and development of young brain cells in human brain models. Microglia use a specific protein to release cholesterol, and when this process is blocked, it causes the organoid cells to grow more, leading to larger brain models. It has always been known that the microglia is key to brain development, however their precise role remains poorly understood. This finding from our team at the Department of Microbiology and Immunology is particularly impactful because we finally understand how cholesterol is transported. Our next focus will be finding out how we can regulate cholesterol release to optimise brain development and slow down, or prevent, the onset of neurological conditions, added Assoc Prof Veronique, who is also Director of the Immunology Translational Research Programme at NUS Medicine.

Moreover, Dr Olivier Cexus from the University of Surrey and formely at A*STAR, progressively deciphered the complex molecular interactions within the brain organoids using proteomic and lipidomic analysis. This provided valuable insights into the metabolic cross-talks involved in brain development and potential implications for diseases.

Together, these collective efforts were instrumental in deepening our understanding of the roles of microglia and the molecular components within brain organoids and its implications for human health.

Prof Florent Ginhoux, Senior Principal Investigator at A*STARs SIgN and Senior author of the study said, "Understanding the complex roles of microglia in brain development and function is an active area of research. Our findings not only advance our understanding of human brain development but also have the potential to impact our knowledge of brain disorders. This opens up new possibilities for future research into neurodevelopmental conditions and potential therapies."

Co-author of the study, Professor Jerry Chan, Senior Consultant, Department of Reproductive Medicine, KK Womens and Childrens Hospital, and Senior National Medical Research Council Clinician Scientist, added, There is currently a lack of tools to study how microglia interacts with the developing brain. This has hampered the understanding of microglia-associated diseases that play an important role during the early development of conditions such as autism, schizophrenia, and neurodegenerative diseases such as Alzheimers and Parkinsons disease.

The development of these novel microglia-associated brain organoids with same-donor pluripotent stem cells gives us an opportunity to study the complex interactions between microglia and neurons during early brain development. Consequentially, this may enable us to study the role of microglia in the setting of diseases and suggest ways to develop new therapies in time.

iPS-cell-derived microglia promote brain organoid maturation via cholesterol transfer

1-Nov-2023

Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.

Read the original post:
New study reveals the critical role of microglia in human brain ... - EurekAlert

Researchers chart the contents of human bone marrow – Science Daily

A team at Weill Cornell Medicine has mapped the location and spatial features of blood-forming cells within human bone marrow. Their findings confirm hypotheses about the anatomy of this tissue and provide a powerful new means to study diseases, ranging from noncancerous conditions, such as sickle cell anemia, to malignant conditions, such as acute leukemia, that affect bone marrow.

For the research described Sept. 29 in Blood, the investigators retrieved deidentified archival bone marrow samples from 29 patients at NewYork-Presbyterian/Weill Cornell Medical Center, generating a vast amount of data about the spatial relationships among their contents.

Creating images of bone marrow has been difficult historically, according to senior author Dr. Sanjay Patel, director of the Multiparametric In Situ Imaging (MISI) Laboratory in the Department of Pathology and Laboratory Medicine and an assistant professor of pathology and laboratory medicine at Weill Cornell Medicine. He and his colleagues overcame these challenges by devising a method for visualizing whole pieces of the tissue, then analyzing them with artificial intelligence (AI).

"We have been able to apply our approach to archival samples in a way that wasn't possible before," said Dr. Patel, who is also a hematopathologist at NewYork-Presbyterian/Weill Cornell Medical Center and a member of the Sandra and Edward Meyer Cancer Center at Weill Cornell Medicine. He noted that they succeeded in identifying and determining the positions of about 1.5 million cells in all.

Visualizing the Elusive Birthplace of Blood

Our blood cells get their start in the bone marrow, where stem cells produce the progenitors that in turn generate red and white blood cells, as well as the wound-sealing fragments known as platelets. Errors in these processes can give rise to acquired diseases including cancers, such as leukemia, lymphoma, and multiple myeloma, and those, such as sickle cell anemia, present from birth.

Studying the birth of blood cells within their native environment in human tissues, however, has proven challenging. What's more, when bone marrow samples are collected, the preservation technique can degrade some nucleic acids and proteins within the cells they contain. And, to avoid bias, researchers need to capture images of an entire piece of tissue, generating a daunting amount of data.

Dr. Patel's team came up with a series of solutions. They started by gathering samples from the tissue archive within Weill Cornell Medicine's Department of Pathology and Laboratory Medicine. These one-to-two-centimeter-long pieces of tissue came from patients who had received biopsies, but who had turned out to be disease free. Researchers in the MISI lab tested a variety of immune proteins known as antibodies, selecting from a catalog of thoroughly-vetted markers used in routine clinical diagnostics, to see which most effectively tagged the contents of bone marrow to make them visible with their fluorescence-based imaging instrumentation.

Their collaborators at BostonGene Corporation, a medical bioinformatics company, then used AI to analyze the resulting images, picking out individual cells, such as stem cells and the platelet-producing megakaryocytes, as well as bone, fat and blood vessels. This technology allowed the team to wrangle an otherwise unmanageable amount of information into a sophisticated analysis, according to Dr. Patel.

A New Way to Investigate Diseases

Previous studies have suggested that, during normal blood cell development, stem and progenitor cells inhabit certain locations, near bone and blood vessels, where surrounding cells create environments critical for their normal function. More recently, some research has suggested that these cells also gather around megakaryocytes, large cells that give rise to platelets. The team's analysis confirmed these patterns, including for megakaryocytes, in human samples. However, when they took patients' age into account, they found the cells were no longer as closely associated with megakaryocytes, which also tended to be smaller in older patients.

While these findings contribute to scientists' understanding of normal bone marrow, Dr. Patel sees the new method's greatest potential in investigating diseases, particularly along the course of their evolution. For a few conditions, such as acute myeloid leukemia, researchers already have evidence that the spatial arrangement of stem and progenitor cells may be disrupted. This new method could open the door to studies that specifically explore such changes -- and to those testing new treatments and evaluating existing ones, according to Dr. Patel.

"I hope our work unlocks the imagination of people who study diseases related to the bone marrow," he said.

Read the original:
Researchers chart the contents of human bone marrow - Science Daily

Anti-aging molecule extends lifespan by improving cellular health – Earth.com

Researchers at the Buck Institute have made a significant breakthrough in the field of aging and disease with the discovery of a new drug-like molecule.

This molecule, known as MIC (Mitophagy-inducing compound), has been shown to extend lifespan and improve health in a variety of ways.

MIC operates by promoting healthy mitochondria through a process called mitophagy, which removes and recycles damaged mitochondria.

Mitochondria are crucial organelles in cells that produce energy, and their health is directly linked to overall cellular health and longevity.

The study demonstrated that this molecule extended the lifespan of C. elegans, a nematode worm frequently used in aging research.

MIC also improved mitochondrial function in mouse muscle cells and showed promise in ameliorating pathology in neurodegenerative disease models.

Mitochondrial dysfunction is known to play a role in various age-related diseases, including neurodegenerative disorders like Parkinsons and Alzheimers, cardiovascular diseases, metabolic disorders, muscle wasting, and cancer progression.

Despite the potential of treatments targeting mitochondrial dysfunction, none have been approved for human use to date.

The mitophagy-inducing compound is a coumarin, a type of naturally bioactive compound found in many plants and certain types of cinnamon.

Coumarins are known for their diverse health benefits, including anticoagulant, antibacterial, antifungal, antiviral, anticancer, antihyperglycemic properties, and neuroprotective effects.

The discovery of the effects of MIC originated from a study on Parkinsons disease. A team of experts including Dr. Julie Andersen and Dr. Shankar Chinta were examining known enhancers of mitophagy in a mouse model.

The mitophagy-inducing compound emerged as a significant find in their research. Instead of immediately testing MIC in mice, the researchers opted to study its impact on overall aging and its mechanism of action using the C. elegans model.

This approach led to the discovery that MIC belongs to a different class of molecules that enhance the expression of a key protein in autophagy and lysosomal functions (TFEB).

The study, led by Dr. Andersen and research scientist Dr. Manish Chamoli, revealed that MIC activates the transcription factor TFEB, a master regulator of genes involved in autophagy and lysosomal functions. Autophagy is an intracellular recycling process vital for cellular health.

The research findings are significant as they show MICs potential in not only extending lifespan but also preventing mitochondrial dysfunction in mammalian cells, offering new avenues for treating various age-related diseases.

Theres a bottleneck in efforts to develop potential therapeutics in the field of geroscience, and the bottleneck is that we dont have enough molecules in the pipeline, said study senior co-author Dr. Gordon Lithgow.

MIC is a great candidate to bring forward given its therapeutic effect across multiple models and the fact that it is a naturally occurring molecule.

Anti-aging strategies encompass a variety of practices and research areas focused on slowing down or reversing the aging process. Here are some key areas:

This includes a balanced diet rich in antioxidants, regular physical activity, adequate sleep, and stress management. Avoiding smoking and excessive alcohol consumption also plays a critical role.

Using sunscreen to protect the skin from UV damage, along with regular use of moisturizers and anti-aging products like retinoids and peptides, can help maintain skin health.

These include hormone replacement therapies, cosmetic procedures like Botox or fillers, and plastic surgery. These methods should be approached cautiously and under medical supervision.

Some people use supplements like omega-3 fatty acids, vitamin D, coenzyme Q10, and others believed to have anti-aging effects. However, their effectiveness can vary and should be used judiciously.

Areas like telomere therapy, stem cell research, and gene editing are being explored for potential anti-aging benefits. While promising, many of these are still in the experimental stages.

Maintaining mental health and active social life is essential. Activities that stimulate the mind, like puzzles, reading, and learning new skills, along with regular social interaction, can contribute to longevity and quality of life.

Regular visits to healthcare professionals for check-ups can help in early detection and management of age-related diseases.

Certain foods are known for their potential anti-aging benefits, mainly due to their high antioxidant content and other beneficial nutrients. Heres a list of some of these foods:

Incorporating these foods into a balanced diet can contribute to overall health and potentially slow some aspects of the aging process. Its also important to maintain a diverse diet and consult with a healthcare professional, especially when making significant dietary changes.

The research is published in the journal Nature Aging.

Like what you read? Subscribe to our newsletter for engaging articles, exclusive content, and the latest updates.

-

Check us out on EarthSnap, a free app brought to you by Eric Ralls and Earth.com.

Continue reading here:
Anti-aging molecule extends lifespan by improving cellular health - Earth.com

Pancreas gene finding gives new insights into human development … – EurekAlert

Understanding how the human pancreas develops is crucial to allow scientists to make insulin producingbeta cells in the quest to cure Type 1 diabetes. Now, scientists have made a unique and surprising discovery - a gene that is essential for making the pancreas in humans is not present in almost all other animals.

Beta cells within the pancreas produce insulin that regulate blood sugar. Every mammal needs the pancreatic beta-cells to survive. In established Type 1 diabetes there are no, or very few, working beta-cells.

The new finding, published in Nature Genetics, challenges assumptions about how the regulation of development evolves. Until now, scientists had assumed that genes essential for development of key organs and functions were highly conserved through evolution, meaning the genetic pathway remains the same between different species, from fish to humans. However, the gene, called ZNF808, is only found in humans, other apes such as chimpanzees and gorillas, and in some monkeys, such as macaques.

This Wellcome Trust-funded research was carried out by researchers at the University of Exeter Medical School, the University of Cambridge and the University of Helsinki in Finland. The study shows just how different humans can be to other animals often used in research, such as mice, emphasising the importance of studying the human pancreas.

Lead author Dr Elisa De Franco, of the University of Exeter Medical School, said: Our finding is really surprising this is the only example we know of where a gene that is fundamental to the development of an organ in humans and primates is not present in other animals. Youd expect a gene only found in primates to regulate a feature that is specific to primates, such as brain size, but it is not the case for this gene, which instead is involved in development of an organ shared by all vertebrates! We think this shows that there must have been an evolutionary shift in higher primates to serve a purpose.

Senior author Professor Andrew Hattersley, of the University of Exeter Medical School, said: One hypothesis that we are exploring is that the evolutionary benefit is to the pancreas in the fetus. Human babies are born through the pelvis, so they cannot stay in the uterus for a longtime as they would grow too large for birth. Instead to cope with being born early and needing to survive without continual feeding they need to be born with more fat than any other animal. This fat is laid down when the fetus pancreas produces more insulin. Our research has shown that human fetuses have more insulin-related growth than other animals.

Dr Nick Owens, of the University of Exeter Medical School, remarked This research really emphasises the importance of studying the human pancreas in order to understand and find new treatments for diabetes. Animal research is important, but it can only tell us so much. We know there are fundamental differences between humans and other animals, such as mice which are often the subject of research in this field. The human pancreas is different in how it looks, works and develops. Our genetic finding could help us understand why thats the case.

ZNF808 belongs to a family of recently evolved proteins which bind and switch off specific regions of the DNA which have also developed recently in evolutionary terms. These DNA regions were among the regions considered junk DNA with no meaningful purpose for decades, but new technology have recently allowed us to discover their functions. Our findings confirm that these regions of our DNA are playing important roles during human development.

Dr Michael Imbeault, from the University of Cambridge, said These findings show that genes like ZNF808, even if relatively recent in evolution, can have a crucial role in human development. ZNF808 is a member of the largest, but also least studied family of proteins that regulate our genome. There are hundreds of genes like ZNF808 in our DNA, many primate or even human specific, and our results demonstrate how these can be key players in human health..

The identification of ZNF808 as being involved in human pancreas development occurred after researchers at the University of Exeter examined genetic samples from patients recruited across the world who were born without a pancreas and found that they all had genetic changes resulting in loss of ZNF808. They then teamed up with colleagues at the University of Cambridge and Helsinki University to study the effect of ZNF808 loss using stem cells in the lab. The results showed that ZNF808 plays an important function early during human development when cells need to decide whether to become pancreas or liver.

Among those who shared their genetic samples was Tania Bashir, aged 12, from Luton. Her father Imran Bashir welcomed the Exeter teams progress. Having an answer to why this happened is important. Weve always wanted to know now we do. The next important step is to understand what this means to the future of science. My dream is that one day, scientists will be able to genetically modify a stem cell and grow a human pancreas, and implant that into Tania, and potentially cure her. I dont know if that will ever be possible, but I do know that this understanding is a crucial step forward.

Professor Timo Otonkoski from University of Helsinki remarked The input of people born without a pancreas was fundamental to this discovery. Nobody would have ever thought that ZNF808 played a role in pancreatic development if we hadnt found the changes in this gene in these patients. The ultimate goal of our research is for this knowledge to be translated into being able to manipulate stem cells to produce beta cells that can produce insulin in the laboratory. That could be the key to curing type 1 diabetes. Our finding is a significant step in understanding what makes the human pancreas unique, which could help progress this area.

The research was supported by the Wellcome Trust, Diabetes UK, and by the Exeter NIHR Biomedical Research Centre. The paper is entitled Primate-specific ZNF808 is essential for pancreatic development in humans and is published in Nature Genetics.

Tanias story

Tania Bashir, Twin 2, weighed just 1.1kg when she was born, via emergency caesarean section, five weeks premature, without a pancreas.

Her mother Saiqa said: From week 20 onwards the weekly scans were stressful. We were told there was a high chance that the smaller twin wouldnt make it, so we kept the fact we had a twin a secret from friends, family and even her other three siblings.

Tanias father Imran, a chartered hardware engineer in Luton, recalled: Tania weighed about as much as a bag of sugar; you could quite easily fit her in the palm of your hand They immediately realised she had neonatal diabetes, but she was also not growing or gaining weight. It took eight weeks of investigations, tests and scans to figure out she had no pancreas. Our lives have never been the same since.

As well as producing no insulin to control her blood sugar, Tania, now 12, does not produce the enzymes that break down fats, proteins and carbohydrates into smaller molecules such as triglycerides, amino acids, and sugars so they can pass through the intestine into the bloodstream. Today, with the support of her parents, she lives a relatively normal life, despite still needing a special liquid feed via a tube at night and permanently using an insulin pump. But her dad recalls the dark days of fear and uncertainty when she was small.

First, we were told she wouldnt survive till birth, then that she wouldnt survive the next few weeks I remember consciously thinking that I didnt want to get too attached, because one of us would have to be strong when she died. In the end, we stopped asking. You normally look to the medical professionals for answers, but because the condition was so rare, there just is not the experience in the UK or across the world. We were learning along with the medical professionals, pushing each other to find better solutions for Tania. We are really lucky to have a fantastic team at the Luton and Dunstable hospital.

Imran found a small network of families globally via Facebook, which provided some shared experience. When Tania was six months old, the family was connected to the research team at the University of Exeter, who specialise in genetic causes of diabetes. They visited the lab and Imran said: I remember thinking, I like what theyre trying to do here we could get an answer.

A decade later, through sequencing all the genes in Tanias DNA (a technique called whole exome sequencing) the Exeter team has identified a gene which is crucial to the development of the human pancreas and is only present in humans and some monkeys, but not in other mammals. Tanias genetic sample was one of just 13 of children born without a pancreas to enable this discovery.

Imran welcomed the progress. Having an answer is important. It draws a line under the question of why, but the journey is far from over. Unlike people with type 1 diabetes, Tanias immune system didnt attack her pancreas so a pancreas could function in her body. I believe that it might be possible to use this research to modify stem cells and grow a pancreas using Tanias own cells, which could be implanted into her. I know it sounds like science fiction, but 40 years ago, there was no such thing as the internet. Now we can share moments instantly across the world. Theres some amazing scientific progress going on in the world, and the work done by Exeter has brought us one step closer to making my dream possible.

Dr Elisa de Franco, of the University of Exeter Medical School, said: Our findings really show the importance of studying the DNA of people with rare diseases to understand how organs develop and function. We are immensely grateful to people like Tania and her family, without them none of this would be possible.

Case study

People

Primate-specific ZNF808 is essential for pancreatic development in humans

Here is the original post:
Pancreas gene finding gives new insights into human development ... - EurekAlert

Study Provides Clues to Developing Better Treatments for Lung … – Duke University School of Medicine

Scientists and clinicians at the Duke University School of Medicine have discovered new details about how lung tissue heals after injury caused by toxins such as air pollution or cigarette smoke.

The researchers found that a cascade of interacting steps involving two different cell types is crucial for healing. An imbalance in these steps can lead to damage that resembles emphysema or lung fibrosis, the study found.

The study, published November 2, 2023, in the journal Cell Stem Cell, paves the way for future investigations to identify possible new treatments to prevent or reverse these diseases.

"A long-standing question in the field of wound healing is how our body organs know to regrow and build the same structure after a wound," said co-senior author Purushothama Rao Tata, PhD, assistant professor of cell biology and medicine, and co-director of the Duke Regeneration Center. The study's other senior author was Aleksandra Tata, PhD, assistant research professor of cell biology.

Tata explained that lung tissue is like a big balloon draped by a structure akin to a fishnet: the extracellular matrix scaffold, which creates multiple compartments with strong, flexible walls that expand and contract as we breathe. This study focused on how the lungs rebuild this scaffold after injury.

To study this question, the scientists used a variety of methods, including single cell transcriptome analysis and other computational tools, to build "time-lapse molecular circuits" to reconstruct wound repair in mouse lungs.

"We refer to it as molecular circuits because we are not looking at one or two genes, but a collection of genes associated with a particular cell state or phenotype," Tata said. "These are like electrical circuits that all come together to switch on a light, for example. All of these genes together exert a collective function."

"Disruption of these circuits revealed key druggable molecules to target two currently incurable lung diseases emphysema and fibrosis," he said. "These diseases are like two sides of the same coin. In a lung with emphysema, we lose the walls of the scaffold. In the case of fibrosis, the wall thickness increases so they are no longer flexible."

The study revealed that after a healing "program" is activated, a cascade of events ensues, involving both epithelial cells (cells that line the lungs) and mesenchymal cells (support cells).

The researchers outlined three crucial steps or "transitional states" that happen during this process. If certain transitional states involving epithelial cells persist too long, the result is fibrosis (buildup of scar tissue). "If there is a blockade in the transition of these cell states, the result is loss of tissue that resembles emphysema," Tata said.

In a preview article highlighting the work, scientists not affiliated with the Duke study pointed out that one of the intermediate cell states identified as crucial in the healing process has previously been termed a "bad actor" in lung fibrosis research. Two drugs currently approved for fibrosis (nintedanib and pirfenodone) actually kill this cell state, Tata said. "Our study shows that treating with these drugs may actually be a bad thing," he said.

Other authors of the study are: Arvind Konkimalla, MD, PhD, currently a resident at Duke University Hospital; postdoctoral associate Satoshi Konishi, MD, PhD; laboratory research analyst Lauren Macadlo; PhD candidates Jeremy Morowitz and Zachary Farino; postdoctoral fellow Naoya Miyashita, PhD; and bioinformatician Pankaj Agarwal, all in the Duke Department of Cell Biology; Department of Pediatrics postdoctoral research associate Lea El Haddad, MD, PhD; Mai K. ElMallah, MD, associate professor of pediatrics; Christina E. Barkauskas, MD, associate professor of medicine; and Tomokazu Souma, MD, PhD, assistant professor in medicine; and Yoshihiko Kobayashi, PhD, now an assistant professor at Kyoto University in Japan.

Read more:
Study Provides Clues to Developing Better Treatments for Lung ... - Duke University School of Medicine