1
|
Gonul CP, Kiser C, Yaka EC, Oz D, Hunerli D, Yerlikaya D, Olcum M, Keskinoglu P, Yener G, Genc S. Microglia-like cells from patient monocytes demonstrate increased phagocytic activity in probable Alzheimer's disease. Mol Cell Neurosci 2024:103990. [PMID: 39732446 DOI: 10.1016/j.mcn.2024.103990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by the accumulation of amyloid plaques, phosphorylated tau tangles and microglia toxicity, resulting in neuronal death and cognitive decline. Since microglia are recognized as one of the key players in the disease, it is crucial to understand how microglia operate in disease conditions and incorporate them into models. The studies on human microglia functions are thought to reflect the post-symptomatic stage of the disease. Recently developed methods involve induced microglia-like cells (iMGs) generated from patients' blood monocytes or induced pluripotent stem cells (iPSCs) as an alternative to studying the microglia cells in vitro. In this research, we aimed to investigate the phenotype and inflammatory responses of iMGs from AD patients. Monocytes derived from blood using density gradient centrifugation were differentiated into iMGs using a cytokine cocktail, including granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-34 (IL-34). After differentiation, cells were assessed by morphological analysis and a microglia surface marker, TMEM119. We used stimulants, lipopolysaccharide (LPS) and beta-amyloid, to examine iMGs' functions. Results showed that iMGs derived from AD patients exhibited increased secretion of pro-inflammatory cytokines upon LPS stimulation. Furthermore, their phagocytic ability was also heightened in stimulated and unstimulated conditions, with cells derived from patients showing increased phagocytic activity compared to healthy controls. Overall, these findings suggest that iMGs derived from patients using the direct conversion method possess characteristics of human microglia, making them an easy and promising model for studying microglia function in AD.
Collapse
Affiliation(s)
- Ceren Perihan Gonul
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Cagla Kiser
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Emis Cansu Yaka
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye; Department of Neurology, İzmir City Hospital, Izmir, Türkiye
| | - Didem Oz
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye; Department of Neurology, Dokuz Eylul University Hospital, Izmir, Türkiye; Global Brain Health Institute, University of California, San Francisco, USA
| | - Duygu Hunerli
- Department of Biostatistics and Medical Informatics, Basic Medical Sciences, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Deniz Yerlikaya
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye
| | - Melis Olcum
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye
| | - Pembe Keskinoglu
- Department of Biostatistics and Medical Informatics, Basic Medical Sciences, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Gorsev Yener
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Department of Neurology, Dokuz Eylul University Hospital, Izmir, Türkiye
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye; Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye.
| |
Collapse
|
2
|
Kloske CM, Mahinrad S, Barnum CJ, Batista AF, Bradshaw EM, Butts B, Carrillo MC, Chakrabarty P, Chen X, Craft S, Da Mesquita S, Dabin LC, Devanand D, Duran-Laforet V, Elyaman W, Evans EE, Fitzgerald-Bocarsly P, Foley KE, Harms AS, Heneka MT, Hong S, Huang YWA, Jackvony S, Lai L, Guen YL, Lemere CA, Liddelow SA, Martin-Peña A, Orr AG, Quintana FJ, Ramey GD, Rexach JE, Rizzo SJS, Sexton C, Tang AS, Torrellas JG, Tsai AP, van Olst L, Walker KA, Wharton W, Tansey MG, Wilcock DM. Advancements in Immunity and Dementia Research: Highlights from the 2023 AAIC Advancements: Immunity Conference. Alzheimers Dement 2024. [PMID: 39692624 DOI: 10.1002/alz.14291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/23/2024] [Accepted: 09/07/2024] [Indexed: 12/19/2024]
Abstract
The immune system is a key player in the onset and progression of neurodegenerative disorders. While brain resident immune cell-mediated neuroinflammation and peripheral immune cell (eg, T cell) infiltration into the brain have been shown to significantly contribute to Alzheimer's disease (AD) pathology, the nature and extent of immune responses in the brain in the context of AD and related dementias (ADRD) remain unclear. Furthermore, the roles of the peripheral immune system in driving ADRD pathology remain incompletely elucidated. In March of 2023, the Alzheimer's Association convened the Alzheimer's Association International Conference (AAIC), Advancements: Immunity, to discuss the roles of the immune system in ADRD. A wide range of topics were discussed, such as animal models that replicate human pathology, immune-related biomarkers and clinical trials, and lessons from other fields describing immune responses in neurodegeneration. This manuscript presents highlights from the conference and outlines avenues for future research on the roles of immunity in neurodegenerative disorders. HIGHLIGHTS: The immune system plays a central role in the pathogenesis of Alzheimer's disease. The immune system exerts numerous effects throughout the brain on amyloid-beta, tau, and other pathways. The 2023 AAIC, Advancements: Immunity, encouraged discussions and collaborations on understanding the role of the immune system.
Collapse
Affiliation(s)
| | | | | | - Andre F Batista
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth M Bradshaw
- Department of Neurology, The Carol and Gene Ludwig Center for Research on Neurodegeneration, Division of Translational Neurobiology, Columbia University, New York, New York, USA
| | | | | | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Diseases, University of Florida, Gainesville, Florida, USA
| | - Xiaoying Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne Craft
- Alzheimer's Disease Research Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Luke C Dabin
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Violeta Duran-Laforet
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Wassim Elyaman
- Department of Neurology, Division of Translational Neurobiology, Columbia University Irving Medical Center, New York, New York, USA
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, USA
| | | | | | - Kate E Foley
- Department of Neurology, Stark Neurosciences Research Institute, Indiana University, Indianapolis, Indiana, USA
| | - Ashley S Harms
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, Massachusetts, USA
| | - Soyon Hong
- UK Dementia Research Institute at University College London, Institute of Neurology, London, UK
| | | | - Stephanie Jackvony
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Neuroscience Graduate Program, Weill Cornell Medicine, New York, New York, USA
| | - Laijun Lai
- University of Connecticut, Storrs, Connecticut, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, California, USA
- Department of Medicine, Quantitative Sciences Unit, Stanford University School of Medicine, Palo Alto, California, USA
| | - Cynthia A Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Departments of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Shane A Liddelow
- Departments of Neuroscience & Physiology and Ophthalmology, Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - Alfonso Martin-Peña
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Anna G Orr
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Neuroscience Graduate Program, Weill Cornell Medicine, New York, New York, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Gene Lay Institute for Immunology and Inflammation, Boston, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Grace D Ramey
- Biological and Medical Informatics PhD Program, University of California San Francisco, San Francisco, California, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California, USA
| | - Jessica E Rexach
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Stacey J S Rizzo
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Alice S Tang
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, California, USA
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, San Francisco, California, USA
| | - Jose G Torrellas
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida, USA
| | - Andy P Tsai
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | - Lynn van Olst
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA) Intramural Research Program, Baltimore, Maryland, USA
| | | | - Malú Gámez Tansey
- Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, Gainesville, Florida, USA
| | - Donna M Wilcock
- Department of Neurology, Stark Neurosciences Research Institute, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
3
|
Carling GK, Fan L, Foxe NR, Norman K, Wong MY, Zhu D, Corona C, Razzoli A, Yu F, Yarahmady A, Ye P, Chen H, Huang Y, Amin S, Sereda R, Lopez-Lee C, Zacharioudakis E, Chen X, Xu J, Cheng F, Gavathiotis E, Cuervo AM, Holtzman DM, Mok SA, Sinha SC, Sidoli S, Ratan RR, Luo W, Gong S, Gan L. Alzheimer's disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model. Neuron 2024; 112:3877-3896.e8. [PMID: 39353433 PMCID: PMC11624100 DOI: 10.1016/j.neuron.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/02/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
The strongest risk factors for late-onset sporadic Alzheimer's disease (AD) include the ε4 allele of apolipoprotein E (APOE), the R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2), and female sex. Here, we combine APOE4 and TREM2R47H (R47H) in female P301S tauopathy mice to identify the pathways activated when AD risk is the strongest, thereby highlighting detrimental disease mechanisms. We find that R47H induces neurodegeneration in 9- to 10-month-old female APOE4 tauopathy mice. The combination of APOE4 and R47H (APOE4-R47H) worsened hyperphosphorylated tau pathology in the frontal cortex and amplified tauopathy-induced microglial cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling and downstream interferon response. APOE4-R47H microglia displayed cGAS- and BAX-dependent upregulation of senescence, showing association between neurotoxic signatures and implicating mitochondrial permeabilization in pathogenesis. By uncovering pathways enhanced by the strongest AD risk factors, our study points to cGAS-STING signaling and associated microglial senescence as potential drivers of AD risk.
Collapse
Affiliation(s)
- Gillian K Carling
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nessa R Foxe
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kendra Norman
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Daphne Zhu
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Carlo Corona
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, NY 10605, USA
| | - Agnese Razzoli
- Transfusion Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia 42122, Italy; Clinical and Experimental PhD Program, University of Modena and Reggio Emilia, Modena 41121, Italy
| | - Fangmin Yu
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Allan Yarahmady
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Pearly Ye
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hao Chen
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yige Huang
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Biochemistry, Structural Biology, Cell Biology, Developmental Biology, and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rebecca Sereda
- Department of Developmental and Molecular Biology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Emmanouil Zacharioudakis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Xiaoying Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center and Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center and Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Evripidis Gavathiotis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sue-Ann Mok
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Subhash C Sinha
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Simone Sidoli
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Rajiv R Ratan
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, NY 10605, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shiaoching Gong
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
4
|
Theme 3 In Vitro Experimental Models. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:122-135. [PMID: 39508666 DOI: 10.1080/21678421.2024.2403300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
|
5
|
Furman S, Green K, Lane TE. COVID-19 and the impact on Alzheimer's disease pathology. J Neurochem 2024; 168:3415-3429. [PMID: 37850241 PMCID: PMC11024062 DOI: 10.1111/jnc.15985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has rapidly escalated into a global pandemic that primarily affects older and immunocompromised individuals due to underlying clinical conditions and suppressed immune responses. Furthermore, COVID-19 patients exhibit a spectrum of neurological symptoms, indicating that COVID-19 can affect the brain in a variety of manners. Many studies, past and recent, suggest a connection between viral infections and an increased risk of neurodegeneration, raising concerns about the neurological effects of COVID-19 and the possibility that it may contribute to Alzheimer's disease (AD) onset or worsen already existing AD pathology through inflammatory processes given that both COVID-19 and AD share pathological features and risk factors. This leads us to question whether COVID-19 is a risk factor for AD and how these two conditions might influence each other. Considering the extensive reach of the COVID-19 pandemic and the devastating impact of the ongoing AD pandemic, their combined effects could have significant public health consequences worldwide.
Collapse
Affiliation(s)
- Susana Furman
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Kim Green
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA
- Center for Virus Research, University of California, Irvine 92697, USA
| |
Collapse
|
6
|
Wu YC, Lehtonen Š, Trontti K, Kauppinen R, Kettunen P, Leinonen V, Laakso M, Kuusisto J, Hiltunen M, Hovatta I, Freude K, Dhungana H, Koistinaho J, Rolova T. Human iPSC-derived pericyte-like cells carrying APP Swedish mutation overproduce beta-amyloid and induce cerebral amyloid angiopathy-like changes. Fluids Barriers CNS 2024; 21:78. [PMID: 39334385 PMCID: PMC11438249 DOI: 10.1186/s12987-024-00576-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Patients with Alzheimer's disease (AD) frequently present with cerebral amyloid angiopathy (CAA), characterized by the accumulation of beta-amyloid (Aβ) within the cerebral blood vessels, leading to cerebrovascular dysfunction. Pericytes, which wrap around vascular capillaries, are crucial for regulating cerebral blood flow, angiogenesis, and vessel stability. Despite the known impact of vascular dysfunction on the progression of neurodegenerative diseases, the specific role of pericytes in AD pathology remains to be elucidated. METHODS To explore this, we generated pericyte-like cells from human induced pluripotent stem cells (iPSCs) harboring the Swedish mutation in the amyloid precursor protein (APPswe) along with cells from healthy controls. We initially verified the expression of classic pericyte markers in these cells. Subsequent functional assessments, including permeability, tube formation, and contraction assays, were conducted to evaluate the functionality of both the APPswe and control cells. Additionally, bulk RNA sequencing was utilized to compare the transcriptional profiles between the two groups. RESULTS Our study reveals that iPSC-derived pericyte-like cells (iPLCs) can produce Aβ peptides. Notably, cells with the APPswe mutation secreted Aβ1-42 at levels ten-fold higher than those of control cells. The APPswe iPLCs also demonstrated a reduced ability to support angiogenesis and maintain barrier integrity, exhibited a prolonged contractile response, and produced elevated levels of pro-inflammatory cytokines following inflammatory stimulation. These functional changes in APPswe iPLCs correspond with transcriptional upregulation in genes related to actin cytoskeleton and extracellular matrix organization. CONCLUSIONS Our findings indicate that the APPswe mutation in iPLCs mimics several aspects of CAA pathology in vitro, suggesting that our iPSC-based vascular cell model could serve as an effective platform for drug discovery aimed to ameliorate vascular dysfunction in AD.
Collapse
Affiliation(s)
- Ying-Chieh Wu
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland.
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland.
| | - Kalevi Trontti
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland
| | - Riitta Kauppinen
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland
| | - Pinja Kettunen
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland
| | - Ville Leinonen
- NeuroCenter, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Iiris Hovatta
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Hiramani Dhungana
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland
| | - Jari Koistinaho
- Helsinki Institute of Life Science, University of Helsinki, 00014, Helsinki, Finland.
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, 00014, Helsinki, Finland.
| | - Taisia Rolova
- Neuroscience Center, University of Helsinki, 00014, Helsinki, Finland.
| |
Collapse
|
7
|
Kuhn MK, Proctor EA. Microglial Drivers of Alzheimer's Disease Pathology: An Evolution of Diverse Participating States. Proteins 2024. [PMID: 39219300 DOI: 10.1002/prot.26723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune-competent cells of the brain, become dysfunctional in Alzheimer's disease (AD), and their aberrant immune responses contribute to the accumulation of pathological proteins and neuronal injury. Genetic studies implicate microglia in the development of AD, prompting interest in developing immunomodulatory therapies to prevent or ameliorate disease. However, microglia take on diverse functional states in disease, playing both protective and detrimental roles in AD, which largely overlap and may shift over the disease course, complicating the identification of effective therapeutic targets. Extensive evidence gathered using transgenic mouse models supports an active role of microglia in pathology progression, though results vary and can be contradictory between different types of models and the degree of pathology at the time of study. Here, we review microglial immune signaling and responses that contribute to the accumulation and spread of pathological proteins or directly affect neuronal health. We additionally explore the use of induced pluripotent stem cell (iPSC)-derived models to study living human microglia and how they have contributed to our knowledge of AD and may begin to fill in the gaps left by mouse models. Ultimately, mouse and iPSC-derived models have their own limitations, and a comprehensive understanding of microglial dysfunction in AD will only be established by an integrated view across models and an appreciation for their complementary viewpoints and limitations.
Collapse
Affiliation(s)
- Madison K Kuhn
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Elizabeth A Proctor
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Engineering Science & Mechanics, The Pennsylvania State University, University Park, Pennsylvania, USA
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
8
|
Wenzel TJ, Desjarlais JD, Mousseau DD. Human brain organoids containing microglia that have arisen innately adapt to a β-amyloid challenge better than those in which microglia are integrated by co-culture. Stem Cell Res Ther 2024; 15:258. [PMID: 39135132 PMCID: PMC11320858 DOI: 10.1186/s13287-024-03876-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Alzheimer disease (AD) is a heterogenous and multifactorial disease, and its pathology is partly driven by microglia and their activated phenotype. Brain organoids (BOs) are gaining prominence as a relevant model of the human brain for the study of AD; however, BOs are commonly devoid of microglia. To overcome this limitation, current protocols incorporate microglia through either (1) co-culture (BO co-culture), or (2) molecular manipulation at critical windows of BO development to have microglia arise innately (BO innate cultures). It is currently unclear whether the microglia incorporated into BOs by either of these two protocols differ in function. METHODS At in vitro day 90, BO innate cultures and BO-co-cultures were challenged with the AD-related β-amyloid peptide (Aβ) for up to 72 h. After Aβ challenge, BOs were collected for immunoblotting. Immunoblots compared immunodensity and protein banding of Aβ and ionized calcium-binding adapter molecule 1 (IBA1, a marker of microglial activation) in BOs. The translational potential of these observations was supported using 56 human cortical samples from neurocognitively normal donors and patients with early-onset AD and late-onset AD. Statistical analyses were conducted using the Kruskal-Wallis test, a two-way ANOVA, or a simple linear regression, and where applicable, followed by Dunn's or Sidak's test. RESULTS We show that BO co-cultures promote Aβ oligomerization as early as 24 h and this coincides with a significant increase in IBA1 levels. In contrast, the Aβs do not oligomerize in BO innate cultures and the IBA1 response was modest and only emerged after 48 h. In human cortical samples, we found IBA1 levels correlated with age at onset, age at death, and the putative diagnostic Aβ(1-42)/Aβ(1-40) ratio (particularly in their oligomeric forms) in a sex-dependent manner. CONCLUSIONS Our unique observations suggest that BOs with innate microglia model the response of a healthy brain to Aβ, and by extension the initial stages of Aβ challenge. It would be impossible to model these early stages of pathogenesis in BOs where microglia are already compromised, such as those with microglia incorporated by co-culture.
Collapse
Affiliation(s)
- Tyler J Wenzel
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada.
| | - Joseph D Desjarlais
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| |
Collapse
|
9
|
Cohen BM, Sonntag KC. Identifying the earliest-occurring clinically targetable precursors of late-onset Alzheimer's disease. EBioMedicine 2024; 106:105238. [PMID: 39002387 PMCID: PMC11284560 DOI: 10.1016/j.ebiom.2024.105238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024] Open
Abstract
Most cases of Alzheimer's disease (AD) are late-onset dementias (LOAD). However, research on AD is predominantly of early-onset disease (EOAD). The determinants of EOAD, gene variants of APP and presenilin proteins, are not the basic precursors of LOAD. Rather, multiple other genes and associated cellular processes underlie risk for LOAD. These determinants could be modified in individuals at risk for LOAD well before signs and symptoms appear. Studying brain cells produced from patient-derived induced-pluripotent-stem-cells (iPSC), in culture, will be instrumental in developing such interventions. This paper summarises evidence accrued from iPSC culture models identifying the earliest occurring clinically targetable determinants of LOAD. Results obtained and replicated, thus far, suggest that abnormalities of bioenergetics, lipid metabolism, digestive organelle function and inflammatory activity are primary processes underlying LOAD. The application of cell culture platforms will become increasingly important in research and also on LOAD detection, assessment, and treatment in the years ahead.
Collapse
Affiliation(s)
- Bruce M Cohen
- Harvard Medical School, Boston, MA, USA; Program for Neuropsychiatric Research, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA.
| | - Kai-Christian Sonntag
- Harvard Medical School, Boston, MA, USA; Laboratory for Translational Research on Neurodegeneration, Program for Neuropsychiatric Research, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA.
| |
Collapse
|
10
|
Nevelchuk S, Brawek B, Schwarz N, Valiente-Gabioud A, Wuttke TV, Kovalchuk Y, Koch H, Höllig A, Steiner F, Figarella K, Griesbeck O, Garaschuk O. Morphotype-specific calcium signaling in human microglia. J Neuroinflammation 2024; 21:175. [PMID: 39020359 PMCID: PMC11256502 DOI: 10.1186/s12974-024-03169-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Key functions of Ca2+ signaling in rodent microglia include monitoring the brain state as well as the surrounding neuronal activity and sensing the danger or damage in their vicinity. Microglial Ca2+ dyshomeostasis is a disease hallmark in many mouse models of neurological disorders but the Ca2+ signal properties of human microglia remain unknown. METHODS We developed a novel genetically-encoded ratiometric Ca2+ indicator, targeting microglial cells in the freshly resected human tissue, organotypically cultured tissue slices and analyzed in situ ongoing Ca2+ signaling of decades-old microglia dwelling in their native microenvironment. RESULTS The data revealed marked compartmentalization of Ca2+ signals, with signal properties differing across the compartments and resident morphotypes. The basal Ca2+ levels were low in ramified and high in ameboid microglia. The fraction of cells with ongoing Ca2+ signaling, the fraction and the amplitude of process Ca2+ signals and the duration of somatic Ca2+ signals decreased when moving from ramified via hypertrophic to ameboid microglia. In contrast, the size of active compartments, the fraction and amplitude of somatic Ca2+ signals and the duration of process Ca2+ signals increased along this pathway.
Collapse
Affiliation(s)
- Sofia Nevelchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Bianca Brawek
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ariel Valiente-Gabioud
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Yury Kovalchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Frederik Steiner
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Katherine Figarella
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Oliver Griesbeck
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany.
| |
Collapse
|
11
|
Patel R, Cardona CL, Angeles E, Singh G, Ashok A, Teich AF, Sproul AA. Reduced SH3RF3 may protect against Alzheimer's disease by lowering microglial pro-inflammatory responses via modulation of JNK and NFkB signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.23.600281. [PMID: 38979369 PMCID: PMC11230201 DOI: 10.1101/2024.06.23.600281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Understanding how high-risk individuals are protected from Alzheimer's disease (AD) may illuminate potential therapeutic targets. A previously identified non-coding SNP in SH3RF3/POSH2 significantly delayed disease onset in a Caribbean Hispanic cohort carrying the PSEN1 G206A mutation sufficient to cause early-onset AD and microglial expression of SH3RF3 has been reported to be a key driver of late-onset AD. SH3RF3 acts as a JNK pathway scaffold and can activate NFκB signaling. While effects of SH3RF3 knockdown in human neurons were subtle, including decreased phospho-tau S422, knockdown in human microglia significantly reduced inflammatory cytokines in response to either a viral mimic or oligomeric Aβ42. This was associated with reduced activation of JNK and NFκB pathways in response to these stimuli. Pharmacological inhibition of JNK or NFκB signaling phenocopied SH3RF3 knockdown. We also found PSEN1 G206A microglia have reduced inflammatory responses to oAβ42. Thus, further reduction of microglial inflammatory responses in PSEN1 mutant carriers by protective SNPs in SH3RF3 might reduce the link between amyloid and neuroinflammation to subsequently delay the onset of AD.
Collapse
|
12
|
Xia Z, Prescott EE, Urbanek A, Wareing HE, King MC, Olerinyova A, Dakin H, Leah T, Barnes KA, Matuszyk MM, Dimou E, Hidari E, Zhang YP, Lam JYL, Danial JSH, Strickland MR, Jiang H, Thornton P, Crowther DC, Ohtonen S, Gómez-Budia M, Bell SM, Ferraiuolo L, Mortiboys H, Higginbottom A, Wharton SB, Holtzman DM, Malm T, Ranasinghe RT, Klenerman D, De S. Co-aggregation with Apolipoprotein E modulates the function of Amyloid-β in Alzheimer's disease. Nat Commun 2024; 15:4695. [PMID: 38824138 PMCID: PMC11144216 DOI: 10.1038/s41467-024-49028-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024] Open
Abstract
Which isoforms of apolipoprotein E (apoE) we inherit determine our risk of developing late-onset Alzheimer's Disease (AD), but the mechanism underlying this link is poorly understood. In particular, the relevance of direct interactions between apoE and amyloid-β (Aβ) remains controversial. Here, single-molecule imaging shows that all isoforms of apoE associate with Aβ in the early stages of aggregation and then fall away as fibrillation happens. ApoE-Aβ co-aggregates account for ~50% of the mass of diffusible Aβ aggregates detected in the frontal cortices of homozygotes with the higher-risk APOE4 gene. We show how dynamic interactions between apoE and Aβ tune disease-related functions of Aβ aggregates throughout the course of aggregation. Our results connect inherited APOE genotype with the risk of developing AD by demonstrating how, in an isoform- and lipidation-specific way, apoE modulates the aggregation, clearance and toxicity of Aβ. Selectively removing non-lipidated apoE4-Aβ co-aggregates enhances clearance of toxic Aβ by glial cells, and reduces secretion of inflammatory markers and membrane damage, demonstrating a clear path to AD therapeutics.
Collapse
Affiliation(s)
- Zengjie Xia
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Emily E Prescott
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Agnieszka Urbanek
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Hollie E Wareing
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Marianne C King
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Anna Olerinyova
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Helen Dakin
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
- Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Tom Leah
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Katy A Barnes
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Martyna M Matuszyk
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Eleni Dimou
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Eric Hidari
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Yu P Zhang
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Jeff Y L Lam
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - John S H Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
- SUPA School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, KY16 9SS, UK
| | - Michael R Strickland
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hong Jiang
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Thornton
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Sohvi Ohtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mireia Gómez-Budia
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Simon M Bell
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Rohan T Ranasinghe
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK.
| | - Suman De
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK.
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
13
|
Dorion MF, Casas D, Shlaifer I, Yaqubi M, Fleming P, Karpilovsky N, Chen CXQ, Nicouleau M, Piscopo VEC, MacDougall EJ, Alluli A, Goldsmith TM, Schneider A, Dorion S, Aprahamian N, MacDonald A, Thomas RA, Dudley RWR, Hall JA, Fon EA, Antel JP, Stratton JA, Durcan TM, La Piana R, Healy LM. An adapted protocol to derive microglia from stem cells and its application in the study of CSF1R-related disorders. Mol Neurodegener 2024; 19:31. [PMID: 38576039 PMCID: PMC10996091 DOI: 10.1186/s13024-024-00723-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/17/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Induced pluripotent stem cell-derived microglia (iMGL) represent an excellent tool in studying microglial function in health and disease. Yet, since differentiation and survival of iMGL are highly reliant on colony-stimulating factor 1 receptor (CSF1R) signaling, it is difficult to use iMGL to study microglial dysfunction associated with pathogenic defects in CSF1R. METHODS Serial modifications to an existing iMGL protocol were made, including but not limited to changes in growth factor combination to drive microglial differentiation, until successful derivation of microglia-like cells from an adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) patient carrying a c.2350G > A (p.V784M) CSF1R variant. Using healthy control lines, the quality of the new iMGL protocol was validated through cell yield assessment, measurement of microglia marker expression, transcriptomic comparison to primary microglia, and evaluation of inflammatory and phagocytic activities. Similarly, molecular and functional characterization of the ALSP patient-derived iMGL was carried out in comparison to healthy control iMGL. RESULTS The newly devised protocol allowed the generation of iMGL with enhanced transcriptomic similarity to cultured primary human microglia and with higher scavenging and inflammatory competence at ~ threefold greater yield compared to the original protocol. Using this protocol, decreased CSF1R autophosphorylation and cell surface expression was observed in iMGL derived from the ALSP patient compared to those derived from healthy controls. Additionally, ALSP patient-derived iMGL presented a migratory defect accompanying a temporal reduction in purinergic receptor P2Y12 (P2RY12) expression, a heightened capacity to internalize myelin, as well as heightened inflammatory response to Pam3CSK4. Poor P2RY12 expression was confirmed to be a consequence of CSF1R haploinsufficiency, as this feature was also observed following CSF1R knockdown or inhibition in mature control iMGL, and in CSF1RWT/KO and CSF1RWT/E633K iMGL compared to their respective isogenic controls. CONCLUSIONS We optimized a pre-existing iMGL protocol, generating a powerful tool to study microglial involvement in human neurological diseases. Using the optimized protocol, we have generated for the first time iMGL from an ALSP patient carrying a pathogenic CSF1R variant, with preliminary characterization pointing toward functional alterations in migratory, phagocytic and inflammatory activities.
Collapse
Affiliation(s)
- Marie-France Dorion
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Diana Casas
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Irina Shlaifer
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Peter Fleming
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Nathan Karpilovsky
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Disorders Research Group, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Carol X-Q Chen
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Michael Nicouleau
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Valerio E C Piscopo
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Emma J MacDougall
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Disorders Research Group, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Aeshah Alluli
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Taylor M Goldsmith
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Alexandria Schneider
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Samuel Dorion
- Faculty of Arts and Sciences, Université de Montréal, Montreal, H3T 1NB, Canada
| | - Nathalia Aprahamian
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Adam MacDonald
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Rhalena A Thomas
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Disorders Research Group, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Roy W R Dudley
- Department of Pediatric Surgery, Division of Neurosurgery, Montreal Children's Hospital, McGill University Health Centers, Montreal, H4A 3J1, Canada
| | - Jeffrey A Hall
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- McGill Parkinson Program and Neurodegenerative Disorders Research Group, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Jo Anne Stratton
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada
| | - Roberta La Piana
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada.
| | - Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada.
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, H3A 2B4, Canada.
| |
Collapse
|
14
|
Romero-Molina C, Neuner SM, Ryszawiec M, Pébay A, Marcora E, Goate A. Autosomal Dominant Alzheimer's Disease Mutations in Human Microglia Are Not Sufficient to Trigger Amyloid Pathology in WT Mice but Might Affect Pathology in 5XFAD Mice. Int J Mol Sci 2024; 25:2565. [PMID: 38473822 PMCID: PMC10932392 DOI: 10.3390/ijms25052565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024] Open
Abstract
Several genetic variants that affect microglia function have been identified as risk factors for Alzheimer's Disease (AD), supporting the importance of this cell type in disease progression. However, the effect of autosomal dominant mutations in the amyloid precursor protein (APP) or the presenilin (PSEN1/2) genes has not been addressed in microglia in vivo. We xenotransplanted human microglia derived from non-carriers and carriers of autosomal dominant AD (ADAD)-causing mutations in the brain of hCSF1 WT or 5XFAD mice. We observed that ADAD mutations in microglia are not sufficient to trigger amyloid pathology in WT mice. In 5XFAD mice, we observed a non-statistically significant increase in amyloid plaque volume and number of dystrophic neurites, coupled with a reduction in plaque-associated microglia in the brain of mice xenotransplanted with ADAD human microglia compared to mice xenotransplanted with non-ADAD microglia. In addition, we observed a non-statistically significant impairment in working and contextual memory in 5XFAD mice xenotransplanted with ADAD microglia compared to those xenotransplanted with non-ADAD-carrier microglia. We conclude that, although not sufficient to initiate amyloid pathology in the healthy brain, mutations in APP and PSEN1 in human microglia might cause mild changes in pathological and cognitive outcomes in 5XFAD mice in a manner consistent with increased AD risk.
Collapse
Affiliation(s)
- Carmen Romero-Molina
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| | - Sarah M. Neuner
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| | - Marcelina Ryszawiec
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| | - Alice Pébay
- Department of Anatomy and Physiology, Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | | | - Edoardo Marcora
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| | - Alison Goate
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| |
Collapse
|
15
|
Beghini DG, Kasai-Brunswick TH, Henriques-Pons A. Induced Pluripotent Stem Cells in Drug Discovery and Neurodegenerative Disease Modelling. Int J Mol Sci 2024; 25:2392. [PMID: 38397069 PMCID: PMC10889263 DOI: 10.3390/ijms25042392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 02/25/2024] Open
Abstract
Induced pluripotent stem cells (iPSCs) are derived from reprogrammed adult somatic cells. These adult cells are manipulated in vitro to express genes and factors essential for acquiring and maintaining embryonic stem cell (ESC) properties. This technology is widely applied in many fields, and much attention has been given to developing iPSC-based disease models to validate drug discovery platforms and study the pathophysiological molecular processes underlying disease onset. Especially in neurological diseases, there is a great need for iPSC-based technological research, as these cells can be obtained from each patient and carry the individual's bulk of genetic mutations and unique properties. Moreover, iPSCs can differentiate into multiple cell types. These are essential characteristics, since the study of neurological diseases is affected by the limited access to injury sites, the need for in vitro models composed of various cell types, the complexity of reproducing the brain's anatomy, the challenges of postmortem cell culture, and ethical issues. Neurodegenerative diseases strongly impact global health due to their high incidence, symptom severity, and lack of effective therapies. Recently, analyses using disease specific, iPSC-based models confirmed the efficacy of these models for testing multiple drugs. This review summarizes the advances in iPSC technology used in disease modelling and drug testing, with a primary focus on neurodegenerative diseases, including Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Daniela Gois Beghini
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Tais Hanae Kasai-Brunswick
- Centro Nacional de Biologia Estrutural e Bioimagem, CENABIO, Universidade Federal do Rio de Janeiro, Seropédica 23890-000, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-REGENERA, Universidade Federal do Rio de Janeiro, Seropédica 23890-000, RJ, Brazil
| | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil;
| |
Collapse
|
16
|
Jäntti H, Jonk S, Gómez Budia M, Ohtonen S, Fagerlund I, Fazaludeen MF, Aakko-Saksa P, Pebay A, Lehtonen Š, Koistinaho J, Kanninen KM, Jalava PI, Malm T, Korhonen P. Particulate matter from car exhaust alters function of human iPSC-derived microglia. Part Fibre Toxicol 2024; 21:6. [PMID: 38360668 PMCID: PMC10870637 DOI: 10.1186/s12989-024-00564-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 01/25/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Air pollution is recognized as an emerging environmental risk factor for neurological diseases. Large-scale epidemiological studies associate traffic-related particulate matter (PM) with impaired cognitive functions and increased incidence of neurodegenerative diseases such as Alzheimer's disease. Inhaled components of PM may directly invade the brain via the olfactory route, or act through peripheral system responses resulting in inflammation and oxidative stress in the brain. Microglia are the immune cells of the brain implicated in the progression of neurodegenerative diseases. However, it remains unknown how PM affects live human microglia. RESULTS Here we show that two different PMs derived from exhausts of cars running on EN590 diesel or compressed natural gas (CNG) alter the function of human microglia-like cells in vitro. We exposed human induced pluripotent stem cell (iPSC)-derived microglia-like cells (iMGLs) to traffic related PMs and explored their functional responses. Lower concentrations of PMs ranging between 10 and 100 µg ml-1 increased microglial survival whereas higher concentrations became toxic over time. Both tested pollutants impaired microglial phagocytosis and increased secretion of a few proinflammatory cytokines with distinct patterns, compared to lipopolysaccharide induced responses. iMGLs showed pollutant dependent responses to production of reactive oxygen species (ROS) with CNG inducing and EN590 reducing ROS production. CONCLUSIONS Our study indicates that traffic-related air pollutants alter the function of human microglia and warrant further studies to determine whether these changes contribute to adverse effects in the brain and on cognition over time. This study demonstrates human iPSC-microglia as a valuable tool to study functional microglial responses to environmental agents.
Collapse
Affiliation(s)
- Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steffi Jonk
- Division of Eye and Vision, Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mireia Gómez Budia
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sohvi Ohtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ilkka Fagerlund
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | - Alice Pebay
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Šárka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pasi I Jalava
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
17
|
Ceylan D, Arat-Çelik HE, Aksahin IC. Integrating mitoepigenetics into research in mood disorders: a state-of-the-art review. Front Physiol 2024; 15:1338544. [PMID: 38410811 PMCID: PMC10895490 DOI: 10.3389/fphys.2024.1338544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/24/2024] [Indexed: 02/28/2024] Open
Abstract
Mood disorders, including major depressive disorder and bipolar disorder, are highly prevalent and stand among the leading causes of disability. Despite the largely elusive nature of the molecular mechanisms underpinning these disorders, two pivotal contributors-mitochondrial dysfunctions and epigenetic alterations-have emerged as significant players in their pathogenesis. This state-of-the-art review aims to present existing data on epigenetic alterations in the mitochondrial genome in mood disorders, laying the groundwork for future research into their pathogenesis. Associations between abnormalities in mitochondrial function and mood disorders have been observed, with evidence pointing to notable changes in mitochondrial DNA (mtDNA). These changes encompass variations in copy number and oxidative damage. However, information on additional epigenetic alterations in the mitochondrial genome remains limited. Recent studies have delved into alterations in mtDNA and regulations in the mitochondrial genome, giving rise to the burgeoning field of mitochondrial epigenetics. Mitochondrial epigenetics encompasses three main categories of modifications: mtDNA methylation/hydroxymethylation, modifications of mitochondrial nucleoids, and mitochondrial RNA alterations. The epigenetic modulation of mitochondrial nucleoids, lacking histones, may impact mtDNA function. Additionally, mitochondrial RNAs, including non-coding RNAs, present a complex landscape influencing interactions between the mitochondria and the nucleus. The exploration of mitochondrial epigenetics offers valuable perspectives on how these alterations impact neurodegenerative diseases, presenting an intriguing avenue for research on mood disorders. Investigations into post-translational modifications and the role of mitochondrial non-coding RNAs hold promise to unravel the dynamics of mitoepigenetics in mood disorders, providing crucial insights for future therapeutic interventions.
Collapse
Affiliation(s)
- Deniz Ceylan
- Department of Psychiatry, School of Medicine, Koç University, Istanbul, Türkiye
- Koç University Research Center for Translational Medicine (KUTTAM), Affective Laboratory, Istanbul, Türkiye
| | | | - Izel Cemre Aksahin
- Koç University Research Center for Translational Medicine (KUTTAM), Affective Laboratory, Istanbul, Türkiye
- Graduate School of Health Sciences, Koç University, Istanbul, Türkiye
| |
Collapse
|
18
|
Carling GK, Fan L, Foxe NR, Norman K, Ye P, Wong MY, Zhu D, Yu F, Xu J, Yarahmady A, Chen H, Huang Y, Amin S, Zacharioudakis E, Chen X, Holtzman DM, Mok SA, Gavathiotis E, Sinha SC, Cheng F, Luo W, Gong S, Gan L. Alzheimer's disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577107. [PMID: 38328219 PMCID: PMC10849737 DOI: 10.1101/2024.01.24.577107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The strongest risk factors for Alzheimer's disease (AD) include the χ4 allele of apolipoprotein E (APOE), the R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2), and female sex. Here, we combine APOE4 and TREM2R47H ( R47H ) in female P301S tauopathy mice to identify the pathways activated when AD risk is the strongest, thereby highlighting disease-causing mechanisms. We find that the R47H variant induces neurodegeneration in female APOE4 mice without impacting hippocampal tau load. The combination of APOE4 and R47H amplified tauopathy-induced cell-autonomous microglial cGAS-STING signaling and type-I interferon response, and interferon signaling converged across glial cell types in the hippocampus. APOE4-R47H microglia displayed cGAS- and BAX-dependent upregulation of senescence, showing association between neurotoxic signatures and implicating mitochondrial permeabilization in pathogenesis. By uncovering pathways enhanced by the strongest AD risk factors, our study points to cGAS-STING signaling and associated microglial senescence as potential drivers of AD risk.
Collapse
|
19
|
Sullivan MA, Lane SD, McKenzie ADJ, Ball SR, Sunde M, Neely GG, Moreno CL, Maximova A, Werry EL, Kassiou M. iPSC-derived PSEN2 (N141I) astrocytes and microglia exhibit a primed inflammatory phenotype. J Neuroinflammation 2024; 21:7. [PMID: 38178159 PMCID: PMC10765839 DOI: 10.1186/s12974-023-02951-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/07/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Widescale evidence points to the involvement of glia and immune pathways in the progression of Alzheimer's disease (AD). AD-associated iPSC-derived glial cells show a diverse range of AD-related phenotypic states encompassing cytokine/chemokine release, phagocytosis and morphological profiles, but to date studies are limited to cells derived from PSEN1, APOE and APP mutations or sporadic patients. The aim of the current study was to successfully differentiate iPSC-derived microglia and astrocytes from patients harbouring an AD-causative PSEN2 (N141I) mutation and characterise the inflammatory and morphological profile of these cells. METHODS iPSCs from three healthy control individuals and three familial AD patients harbouring a heterozygous PSEN2 (N141I) mutation were used to derive astrocytes and microglia-like cells and cell identity and morphology were characterised through immunofluorescent microscopy. Cellular characterisation involved the stimulation of these cells by LPS and Aβ42 and analysis of cytokine/chemokine release was conducted through ELISAs and multi-cytokine arrays. The phagocytic capacity of these cells was then indexed by the uptake of fluorescently-labelled fibrillar Aβ42. RESULTS AD-derived astrocytes and microglia-like cells exhibited an atrophied and less complex morphological appearance than healthy controls. AD-derived astrocytes showed increased basal expression of GFAP, S100β and increased secretion and phagocytosis of Aβ42 while AD-derived microglia-like cells showed decreased IL-8 secretion compared to healthy controls. Upon immunological challenge AD-derived astrocytes and microglia-like cells showed exaggerated secretion of the pro-inflammatory IL-6, CXCL1, ICAM-1 and IL-8 from astrocytes and IL-18 and MIF from microglia. CONCLUSION Our study showed, for the first time, the differentiation and characterisation of iPSC-derived astrocytes and microglia-like cells harbouring a PSEN2 (N141I) mutation. PSEN2 (N141I)-mutant astrocytes and microglia-like cells presented with a 'primed' phenotype characterised by reduced morphological complexity, exaggerated pro-inflammatory cytokine secretion and altered Aβ42 production and phagocytosis.
Collapse
Affiliation(s)
- Michael A Sullivan
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Samuel D Lane
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - André D J McKenzie
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Sarah R Ball
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Margaret Sunde
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - G Gregory Neely
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camperdown, Australia
| | - Cesar L Moreno
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camperdown, Australia
| | - Alexandra Maximova
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Eryn L Werry
- School of Medical Sciences, The Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
- School of Chemistry, The Faculty of Science, The University of Sydney, Camperdown, Australia.
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
| | - Michael Kassiou
- School of Chemistry, The Faculty of Science, The University of Sydney, Camperdown, Australia.
| |
Collapse
|
20
|
Jäntti H, Kistemaker L, Buonfiglioli A, De Witte LD, Malm T, Hol EM. Emerging Models to Study Human Microglia In vitro. ADVANCES IN NEUROBIOLOGY 2024; 37:545-568. [PMID: 39207712 DOI: 10.1007/978-3-031-55529-9_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
New in vitro models provide an exciting opportunity to study live human microglia. Previously, a major limitation in understanding human microglia in health and disease has been their limited availability. Here, we provide an overview of methods to obtain human stem cell or blood monocyte-derived microglia-like cells that provide a nearly unlimited source of live human microglia for research. We address how understanding microglial ontogeny can help modeling microglial identity and function in a dish with increased accuracy. Moreover, we categorize stem cell-derived differentiation methods into embryoid body based, growth factor driven, and coculture-driven approaches, and review novel viral approaches to reprogram stem cells directly into microglia-like cells. Furthermore, we review typical readouts used in the field to verify microglial identity and characterize functional microglial phenotypes. We provide an overview of methods used to study microglia in environments more closely resembling the (developing) human CNS, such as cocultures and brain organoid systems with incorporated or innately developing microglia. We highlight how microglia-like cells can be utilized to reveal molecular and functional mechanisms in human disease context, focusing on Alzheimer's disease and other neurodegenerative diseases as well as neurodevelopmental diseases. Finally, we provide a critical overview of challenges and future opportunities to more accurately model human microglia in a dish and conclude that novel in vitro microglia-like cells provide an exciting potential to bring preclinical research of microglia to a new era.
Collapse
Affiliation(s)
- Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Lois Kistemaker
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Alice Buonfiglioli
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lot D De Witte
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
21
|
Wu X, Ni Y, Li W, Yang B, Yang X, Zhu Z, Zhang J, Wu X, Shen Q, Liao Z, Yuan L, Chen Y, Du Q, Wang C, Liu P, Miao Y, Li N, Zhang S, Liao M, Hua J. Rapid conversion of porcine pluripotent stem cells into macrophages with chemically defined conditions. J Biol Chem 2024; 300:105556. [PMID: 38097188 PMCID: PMC10825052 DOI: 10.1016/j.jbc.2023.105556] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 01/11/2024] Open
Abstract
A renewable source of porcine macrophages derived from pluripotent stem cells (PSCs) would be a valuable alternative to primary porcine alveolar macrophages (PAMs) in the research of host-pathogen interaction mechanisms. We developed an efficient and rapid protocol, within 11 days, to derive macrophages from porcine PSCs (pPSCs). The pPSC-derived macrophages (pPSCdMs) exhibited molecular and functional characteristics of primary macrophages. The pPSCdMs showed macrophage-specific surface protein expression and macrophage-specific transcription factors, similar to PAMs. The pPSCdMs also exhibited the functional characteristics of macrophages, such as endocytosis, phagocytosis, porcine respiratory and reproductive syndrome virus infection and the response to lipopolysaccharide stimulation. Furthermore, we performed transcriptome sequencing of the whole differentiation process to track the fate transitions of porcine PSCs involved in the signaling pathway. The activation of transforming growth factor beta signaling was required for the formation of mesoderm and the inhibition of the transforming growth factor beta signaling pathway at the hematopoietic endothelium stage could enhance the fate transformation of hematopoiesis. In summary, we developed an efficient and rapid protocol to generate pPSCdMs that showed aspects of functional maturity comparable with PAMs. pPSCdMs could provide a broad prospect for the platforms of host-pathogen interaction mechanisms.
Collapse
Affiliation(s)
- Xiaolong Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yu Ni
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China
| | - Wenhao Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Bin Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xinchun Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhenshuo Zhu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Juqing Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaojie Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Qiaoyan Shen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Zheng Liao
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Liming Yuan
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yunlong Chen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Qian Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chengbao Wang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Pentao Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, Stem Cell and Regenerative Medicine Consortium, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yiliang Miao
- Institute of Stem Cell and Regenerative Biology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Shiqiang Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| | - Mingzhi Liao
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, China.
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
22
|
Ohtonen S, Giudice L, Jäntti H, Fazaludeen MF, Shakirzyanova A, Gómez-Budia M, Välimäki NN, Niskanen J, Korvenlaita N, Fagerlund I, Koistinaho J, Amiry-Moghaddam M, Savchenko E, Roybon L, Lehtonen Š, Korhonen P, Malm T. Human iPSC-derived microglia carrying the LRRK2-G2019S mutation show a Parkinson's disease related transcriptional profile and function. Sci Rep 2023; 13:22118. [PMID: 38092815 PMCID: PMC10719377 DOI: 10.1038/s41598-023-49294-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
LRRK2-G2019S is one of the most common Parkinson's disease (PD)-associated mutations and has been shown to alter microglial functionality. However, the impact of LRRK2-G2019S on transcriptional profile of human induced pluripotent stem cell-derived microglia-like cells (iMGLs) and how it corresponds to microglia in idiopathic PD brain is not known. Here we demonstrate that LRRK2-G2019S carrying iMGL recapitulate aspects of the transcriptional signature of human idiopathic PD midbrain microglia. LRRK2-G2019S induced subtle and donor-dependent alterations in iMGL mitochondrial respiration, phagocytosis and cytokine secretion. Investigation of microglial transcriptional state in the midbrains of PD patients revealed a subset of microglia with a transcriptional overlap between the in vitro PD-iMGL and human midbrain PD microglia. We conclude that LRRK2-G2019S iMGL serve as a model to study PD-related effects in human microglia.
Collapse
Affiliation(s)
- Sohvi Ohtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Luca Giudice
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Anastasia Shakirzyanova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mireia Gómez-Budia
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nelli-Noora Välimäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jonna Niskanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nea Korvenlaita
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ilkka Fagerlund
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ekaterina Savchenko
- Stem Cell Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Laurent Roybon
- Stem Cell Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Neurodegenerative Science, The MiND Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Šárka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
23
|
Tripathi SM, Vishwakarma PK, Ojha S, Mishra S. Relationship between COVID-19 and Neurological Disorder. CORONAVIRUSES 2023; 4. [DOI: 10.2174/0126667975253863230920070917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/20/2023] [Accepted: 08/18/2023] [Indexed: 01/03/2025]
Abstract
Abstract:
The COVID-19 pandemic has resulted in a socially isolating way of life, and dementia
patients are among those who are most affected. Lockdown procedures and the inability to monitor
illnesses have led to a rapid decline in cognitive function in these individuals, with neuropsychiatric
symptoms, such as agitation, delirium, and impaired motor performance being prevalent. However, the
use of antipsychotics in treating these symptoms can increase the risk of death during COVID-19. Effective
pain therapy can be used as an alternative to reduce or avoid the use of antipsychotics, given the
consistent relationship between agitation and pain in dementia patients. The importance of properly
assessing and managing pain in dementia patients is highlighted. Additionally, the article discusses
how COVID-19 can affect brain health through inflammation, blood clotting, and blood vessel damage,
leading to potential long-term effects on cognitive function. Healthcare professionals must be
aware of the increased risk of neuropsychiatric symptoms in dementia patients during the pandemic
and prioritize pain management as a viable alternative to antipsychotics. Proper care and attention are
necessary to prevent cognitive decline and potential long-term effects on brain health in these vulnerable
individuals.
Collapse
Affiliation(s)
- Shivendra Mani Tripathi
- Department of Pharmaceutical Science & Technology, Madan Mohan Malaviya University of Technology, Gorakhpur,
Uttar Pradesh, India
| | - Pratik Kumar Vishwakarma
- Department of Pharmaceutical Science & Technology, Madan Mohan Malaviya University of Technology, Gorakhpur,
Uttar Pradesh, India
| | - Smriti Ojha
- Department of Pharmaceutical Science & Technology, Madan Mohan Malaviya University of Technology, Gorakhpur,
Uttar Pradesh, India
| | - Sudhanshu Mishra
- Department of Pharmaceutical Science & Technology, Madan Mohan Malaviya University of Technology, Gorakhpur,
Uttar Pradesh, India
| |
Collapse
|
24
|
Cacace R, Zhou L, Hendrickx Van de Craen E, Buist A, Hoogmartens J, Sieben A, Cras P, Vandenberghe R, De Deyn PP, Oehlrich D, De Bondt A, Engelborghs S, Moechars D, Van Broeckhoven C. Mutated Toll-like receptor 9 increases Alzheimer's disease risk by compromising innate immunity protection. Mol Psychiatry 2023; 28:5380-5389. [PMID: 37433968 PMCID: PMC11041692 DOI: 10.1038/s41380-023-02166-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 07/13/2023]
Abstract
The development of Alzheimer's disease (AD) involves central and peripheral immune deregulation. Gene identification and studies of AD genetic variants of peripheral immune components may aid understanding of peripheral-central immune crosstalk and facilitate new opportunities for therapeutic intervention. In this study, we have identified in a Flanders-Belgian family a novel variant p.E317D in the Toll-like receptor 9 gene (TLR9), co-segregating with EOAD in an autosomal dominant manner. In human, TLR9 is an essential innate and adaptive immune component predominantly expressed in peripheral immune cells. The p.E317D variant caused 50% reduction in TLR9 activation in the NF-κB luciferase assay suggesting that p.E317D is a loss-of-function mutation. Cytokine profiling of human PBMCs upon TLR9 activation revealed a predominantly anti-inflammatory response in contrast to the inflammatory responses from TLR7/8 activation. The cytokines released upon TLR9 activation suppressed inflammation and promoted phagocytosis of Aβ42 oligomers in human iPSC-derived microglia. Transcriptome analysis identified upregulation of AXL, RUBICON and associated signaling pathways, which may underline the effects of TLR9 signaling-induced cytokines in regulating the inflammatory status and phagocytic property of microglia. Our data suggest a protective role of TLR9 signaling in AD pathogenesis, and we propose that TLR9 loss-of-function may disrupt a peripheral-central immune crosstalk that promotes dampening of inflammation and clearance of toxic protein species, leading to the build-up of neuroinflammation and pathogenic protein aggregates in AD development.
Collapse
Affiliation(s)
- Rita Cacace
- Neurodegenerative Brain Diseases, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Lujia Zhou
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Elisabeth Hendrickx Van de Craen
- Neurodegenerative Brain Diseases, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology, University Hospital Antwerp, Edegem, Belgium
| | - Arjan Buist
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Julie Hoogmartens
- Neurodegenerative Brain Diseases, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anne Sieben
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | - Patrick Cras
- Department of Neurology, University Hospital Antwerp, Edegem, Belgium
- Institute Born-Bunge, Antwerp, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals Leuven, and Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Peter P De Deyn
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp, Antwerp, Belgium
| | - Daniel Oehlrich
- Discovery Sciences, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - An De Bondt
- Discovery Sciences, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology, Universitair Ziekenhuis Brussel, and Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Diederik Moechars
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
25
|
Marei HE, Khan MUA, Hasan A. Potential use of iPSCs for disease modeling, drug screening, and cell-based therapy for Alzheimer's disease. Cell Mol Biol Lett 2023; 28:98. [PMID: 38031028 PMCID: PMC10687886 DOI: 10.1186/s11658-023-00504-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic illness marked by increasing cognitive decline and nervous system deterioration. At this time, there is no known medication that will stop the course of Alzheimer's disease; instead, most symptoms are treated. Clinical trial failure rates for new drugs remain high, highlighting the urgent need for improved AD modeling for improving understanding of the underlying pathophysiology of disease and improving drug development. The development of induced pluripotent stem cells (iPSCs) has made it possible to model neurological diseases like AD, giving access to an infinite number of patient-derived cells capable of differentiating neuronal fates. This advance will accelerate Alzheimer's disease research and provide an opportunity to create more accurate patient-specific models of Alzheimer's disease to support pathophysiological research, drug development, and the potential application of stem cell-based therapeutics. This review article provides a complete summary of research done to date on the potential use of iPSCs from AD patients for disease modeling, drug discovery, and cell-based therapeutics. Current technological developments in AD research including 3D modeling, genome editing, gene therapy for AD, and research on familial (FAD) and sporadic (SAD) forms of the disease are discussed. Finally, we outline the issues that need to be elucidated and future directions for iPSC modeling in AD.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| | - Muhammad Umar Aslam Khan
- Biomedical Research Center, Qatar University, 2713, Doha, Qatar
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
26
|
Michalski C, Wen Z. Leveraging iPSC technology to assess neuro-immune interactions in neurological and psychiatric disorders. Front Psychiatry 2023; 14:1291115. [PMID: 38025464 PMCID: PMC10672983 DOI: 10.3389/fpsyt.2023.1291115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Communication between the immune and the nervous system is essential for human brain development and homeostasis. Disruption of this intricately regulated crosstalk can lead to neurodevelopmental, psychiatric, or neurodegenerative disorders. While animal models have been essential in characterizing the role of neuroimmunity in development and disease, they come with inherent limitations due to species specific differences, particularly with regard to microglia, the major subset of brain resident immune cells. The advent of induced pluripotent stem cell (iPSC) technology now allows the development of clinically relevant models of the central nervous system that adequately reflect human genetic architecture. This article will review recent publications that have leveraged iPSC technology to assess neuro-immune interactions. First, we will discuss the role of environmental stressors such as neurotropic viruses or pro-inflammatory cytokines on neuronal and glial function. Next, we will review how iPSC models can be used to study genetic risk factors in neurological and psychiatric disorders. Lastly, we will evaluate current challenges and future potential for iPSC models in the field of neuroimmunity.
Collapse
Affiliation(s)
- Christina Michalski
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
27
|
Guo X, Jiang M, Dai X, Shen J, Wang X. Presenilin-1, mutated in familial Alzheimer's disease, maintains genome stability via a γ-secretase dependent way. DNA Repair (Amst) 2023; 131:103580. [PMID: 37804602 DOI: 10.1016/j.dnarep.2023.103580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
Mutations in Presenilin-1 (PS1) account for over 80 % mutations linked to familial Alzheimer's disease (AD). However, the mechanisms of action of PS1 mutations in causing familial AD are not fully understood, limiting opportunities to develop targeted disease-modifying therapies for individuals carrying PS1 mutation. To gain more comprehensive insights into the impact of PS1 mutations on genome stability, we knocked down PS1 in SH-SY5Y, HMC3 and A549 cells. This revealed that PS1 knockdown (KD) dramatically induces genome instability (GIN) in all cell types, as indicated by the increased incidence of micronuclei, nucleoplasmic bridges and/or nuclear buds. Although amyloid β (Aβ) was able to induce GIN, PS1-KD was associated with decreased expression of Aβ in SH-SY5Y cells, suggesting Aβ is not the primary cause of GIN in PS1-KD cells. In contrast, inhibiting the PS1 γ-secretase activity by DAPT recapitulated GIN phenotype as seen in PS1-KD cells, indicating that the induction of GIN following PS1 KD can be attributed to the loss of γ-secretase activity. PS1 KD or γ-secretase inhibition markedly sensitizes SH-SY5Y to the genotoxicity of mitomycin C. Interestingly, overexpression of the wildtype PS1 dramatically increased GIN in SH-SY5Y. Collectively, our study demonstrates the potential of PS1 and its γ-secretase activity in maintaining genome stability, highlighting a novel potential link between PS1 loss-of-function or gain-of-function mutations and familial AD through GIN. Several mechanisms by which GIN induced by PS1 dys-expression may contribute to AD are discussed.
Collapse
Affiliation(s)
- Xihan Guo
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan 650500, China; The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan 650500, China.
| | - Minyan Jiang
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan 650500, China
| | - Xueqin Dai
- Academy of Biomedical Engineering, Kunming Medical University, Kunming 650500, China
| | - Jie Shen
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan 650500, China
| | - Xu Wang
- School of Life Sciences, Yunnan Normal University, Kunming, Yunnan 650500, China; The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan 650500, China; Yeda Institute of Gene and Cell Therapy, Taizhou, Zhejiang 318000, China
| |
Collapse
|
28
|
Theme 03 - In Vitro Experimental Models. Amyotroph Lateral Scler Frontotemporal Degener 2023; 24:115-127. [PMID: 37966318 DOI: 10.1080/21678421.2023.2260193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
|
29
|
Stöberl N, Maguire E, Salis E, Shaw B, Hall-Roberts H. Human iPSC-derived glia models for the study of neuroinflammation. J Neuroinflammation 2023; 20:231. [PMID: 37817184 PMCID: PMC10566197 DOI: 10.1186/s12974-023-02919-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/02/2023] [Indexed: 10/12/2023] Open
Abstract
Neuroinflammation is a complex biological process that plays a significant role in various brain disorders. Microglia and astrocytes are the key cell types involved in inflammatory responses in the central nervous system. Neuroinflammation results in increased levels of secreted inflammatory factors, such as cytokines, chemokines, and reactive oxygen species. To model neuroinflammation in vitro, various human induced pluripotent stem cell (iPSC)-based models have been utilized, including monocultures, transfer of conditioned media between cell types, co-culturing multiple cell types, neural organoids, and xenotransplantation of cells into the mouse brain. To induce neuroinflammatory responses in vitro, several stimuli have been established that can induce responses in either microglia, astrocytes, or both. Here, we describe and critically evaluate the different types of iPSC models that can be used to study neuroinflammation and highlight how neuroinflammation has been induced and measured in these cultures.
Collapse
Affiliation(s)
- Nina Stöberl
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Emily Maguire
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Elisa Salis
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Bethany Shaw
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Hazel Hall-Roberts
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| |
Collapse
|
30
|
Pomeshchik Y, Velasquez E, Gil J, Klementieva O, Gidlöf R, Sydoff M, Bagnoli S, Nacmias B, Sorbi S, Westergren-Thorsson G, Gouras GK, Rezeli M, Roybon L. Proteomic analysis across patient iPSC-based models and human post-mortem hippocampal tissue reveals early cellular dysfunction and progression of Alzheimer's disease pathogenesis. Acta Neuropathol Commun 2023; 11:150. [PMID: 37715247 PMCID: PMC10504768 DOI: 10.1186/s40478-023-01649-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/30/2023] [Indexed: 09/17/2023] Open
Abstract
The hippocampus is a primary region affected in Alzheimer's disease (AD). Because AD postmortem brain tissue is not available prior to symptomatic stage, we lack understanding of early cellular pathogenic mechanisms. To address this issue, we examined the cellular origin and progression of AD pathogenesis by comparing patient-based model systems including iPSC-derived brain cells transplanted into the mouse brain hippocampus. Proteomic analysis of the graft enabled the identification of pathways and network dysfunction in AD patient brain cells, associated with increased levels of Aβ-42 and β-sheet structures. Interestingly, the host cells surrounding the AD graft also presented alterations in cellular biological pathways. Furthermore, proteomic analysis across human iPSC-based models and human post-mortem hippocampal tissue projected coherent longitudinal cellular changes indicative of early to end stage AD cellular pathogenesis. Our data showcase patient-based models to study the cell autonomous origin and progression of AD pathogenesis.
Collapse
Affiliation(s)
- Yuriy Pomeshchik
- iPSC Laboratory for CNS Disease Modelling, Department of Experimental Medical Science, BMC D10, Lund University, 22184, Lund, Sweden.
- Strategic Research Area MultiPark, Lund University, 22184, Lund, Sweden.
- Lund Stem Cell Center, Lund University, 22184, Lund, Sweden.
| | - Erika Velasquez
- iPSC Laboratory for CNS Disease Modelling, Department of Experimental Medical Science, BMC D10, Lund University, 22184, Lund, Sweden
- Strategic Research Area MultiPark, Lund University, 22184, Lund, Sweden
- Lund Stem Cell Center, Lund University, 22184, Lund, Sweden
| | - Jeovanis Gil
- Clinical Protein Science & Imaging, Department of Biomedical Engineering, BMC D13, Lund University, 22184, Lund, Sweden
| | - Oxana Klementieva
- Strategic Research Area MultiPark, Lund University, 22184, Lund, Sweden
- Medical Micro-Spectroscopy, Department of Experimental Medical Science, BMC B10, Lund University, 22184, Lund, Sweden
| | - Ritha Gidlöf
- Lund University BioImaging Centre, Faculty of Medicine, Lund University, 22142, Lund, Sweden
| | - Marie Sydoff
- Lund University BioImaging Centre, Faculty of Medicine, Lund University, 22142, Lund, Sweden
| | - Silvia Bagnoli
- Laboratorio Di Neurogenetica, Dipartimento Di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Laboratorio Di Neurogenetica, Dipartimento Di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Sandro Sorbi
- Laboratorio Di Neurogenetica, Dipartimento Di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, 50134, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Gunilla Westergren-Thorsson
- Department of Experimental Medical Science, BMC C12, Faculty of Medicine, Lund University, 22142, Lund, Sweden
| | - Gunnar K Gouras
- Strategic Research Area MultiPark, Lund University, 22184, Lund, Sweden
- Experimental Dementia Research Unit, Department of Experimental Medical Science, BMC B11, Lund University, 22184, Lund, Sweden
| | - Melinda Rezeli
- Clinical Protein Science & Imaging, Department of Biomedical Engineering, BMC D13, Lund University, 22184, Lund, Sweden
- Swedish National Infrastructure for Biological Mass Spectrometry (BioMS), Lund University, 22184, Lund, Sweden
| | - Laurent Roybon
- iPSC Laboratory for CNS Disease Modelling, Department of Experimental Medical Science, BMC D10, Lund University, 22184, Lund, Sweden.
- Strategic Research Area MultiPark, Lund University, 22184, Lund, Sweden.
- Lund Stem Cell Center, Lund University, 22184, Lund, Sweden.
- Department of Neurodegenerative Science, The MiND Program, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
31
|
Costa-Laparra I, Juárez-Escoto E, Vicario C, Moratalla R, García-Sanz P. APOE ε4 allele, along with G206D- PSEN1 mutation, alters mitochondrial networks and their degradation in Alzheimer's disease. Front Aging Neurosci 2023; 15:1087072. [PMID: 37455931 PMCID: PMC10340123 DOI: 10.3389/fnagi.2023.1087072] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Alzheimer's disease remains the most common neurodegenerative disorder, depicted mainly by memory loss and the presence in the brain of senile plaques and neurofibrillary tangles. This disease is related to several cellular alterations like the loss of synapses, neuronal death, disruption of lipid homeostasis, mitochondrial fragmentation, or raised oxidative stress. Notably, changes in the autophagic pathway have turned out to be a key factor in the early development of the disease. The aim of this research is to determine the impact of the APOE allele ε4 and G206D-PSEN1 on the underlying mechanisms of Alzheimer's disease. Methods Fibroblasts from Alzheimer's patients with APOE 3/4 + G206D-PSEN1 mutation and homozygous APOE ε4 were used to study the effects of APOE polymorphism and PSEN1 mutation on the autophagy pathway, mitochondrial network fragmentation, superoxide anion levels, lysosome clustering, and p62/SQSTM1 levels. Results We observed that the APOE allele ε4 in homozygosis induces mitochondrial network fragmentation that correlates with an increased colocalization with p62/SQSTM1, probably due to an inefficient autophagy. Moreover, G206D-PSEN1 mutation causes an impairment of the integrity of mitochondrial networks, triggering high superoxide anion levels and thus making APOE 3/4 + PSEN1 fibroblasts more vulnerable to cell death induced by oxidative stress. Of note, PSEN1 mutation induces accumulation and clustering of lysosomes that, along with an increase of global p62/SQSTM1, could compromise lysosomal function and, ultimately, its degradation. Conclusion The findings suggest that all these modifications could eventually contribute to the neuronal degeneration that underlies the pathogenesis of Alzheimer's disease. Further research in this area may help to develop targeted therapies for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Irene Costa-Laparra
- Neurobiology of the Basal Ganglia Laboratory, Department of Functional Systems and Neurobiology, Instituto Cajal, Spanish National Research Council (CSIC), Madrid, Spain
| | - Elena Juárez-Escoto
- Neurobiology of the Basal Ganglia Laboratory, Department of Functional Systems and Neurobiology, Instituto Cajal, Spanish National Research Council (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Vicario
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Stem Cells, Neurogenesis and Neurodegeneration Laboratory, Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Spanish National Research Council (CSIC), Madrid, Spain
| | - Rosario Moratalla
- Neurobiology of the Basal Ganglia Laboratory, Department of Functional Systems and Neurobiology, Instituto Cajal, Spanish National Research Council (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia García-Sanz
- Neurobiology of the Basal Ganglia Laboratory, Department of Functional Systems and Neurobiology, Instituto Cajal, Spanish National Research Council (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
32
|
Muñoz Herrera OM, Hong BV, Ruiz Mendiola U, Maezawa I, Jin LW, Lebrilla CB, Harvey DJ, Zivkovic AM. Cholesterol, Amyloid Beta, Fructose, and LPS Influence ROS and ATP Concentrations and the Phagocytic Capacity of HMC3 Human Microglia Cell Line. Int J Mol Sci 2023; 24:10396. [PMID: 37373543 PMCID: PMC10299308 DOI: 10.3390/ijms241210396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/16/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Research has found that genes specific to microglia are among the strongest risk factors for Alzheimer's disease (AD) and that microglia are critically involved in the etiology of AD. Thus, microglia are an important therapeutic target for novel approaches to the treatment of AD. High-throughput in vitro models to screen molecules for their effectiveness in reversing the pathogenic, pro-inflammatory microglia phenotype are needed. In this study, we used a multi-stimulant approach to test the usefulness of the human microglia cell 3 (HMC3) cell line, immortalized from a human fetal brain-derived primary microglia culture, in duplicating critical aspects of the dysfunctional microglia phenotype. HMC3 microglia were treated with cholesterol (Chol), amyloid beta oligomers (AβO), lipopolysaccharide (LPS), and fructose individually and in combination. HMC3 microglia demonstrated changes in morphology consistent with activation when treated with the combination of Chol + AβO + fructose + LPS. Multiple treatments increased the cellular content of Chol and cholesteryl esters (CE), but only the combination treatment of Chol + AβO + fructose + LPS increased mitochondrial Chol content. Microglia treated with combinations containing Chol + AβO had lower apolipoprotein E (ApoE) secretion, with the combination of Chol + AβO + fructose + LPS having the strongest effect. Combination treatment with Chol + AβO + fructose + LPS also induced APOE and TNF-α expression, reduced ATP production, increased reactive oxygen species (ROS) concentration, and reduced phagocytosis events. These findings suggest that HMC3 microglia treated with the combination of Chol + AβO + fructose + LPS may be a useful high-throughput screening model amenable to testing on 96-well plates to test potential therapeutics to improve microglial function in the context of AD.
Collapse
Affiliation(s)
- Oscar M. Muñoz Herrera
- Department of Nutrition, University of California, Davis, CA 95616, USA; (O.M.M.H.); (B.V.H.)
| | - Brian V. Hong
- Department of Nutrition, University of California, Davis, CA 95616, USA; (O.M.M.H.); (B.V.H.)
| | - Ulises Ruiz Mendiola
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA; (U.R.M.); (I.M.); (L.-W.J.)
| | - Izumi Maezawa
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA; (U.R.M.); (I.M.); (L.-W.J.)
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA; (U.R.M.); (I.M.); (L.-W.J.)
| | | | - Danielle J. Harvey
- Department of Public Health Sciences, University of California, Davis, CA 95616, USA;
| | - Angela M. Zivkovic
- Department of Nutrition, University of California, Davis, CA 95616, USA; (O.M.M.H.); (B.V.H.)
| |
Collapse
|
33
|
Whitfield JF, Rennie K, Chakravarthy B. Alzheimer's Disease and Its Possible Evolutionary Origin: Hypothesis. Cells 2023; 12:1618. [PMID: 37371088 PMCID: PMC10297544 DOI: 10.3390/cells12121618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The enormous, 2-3-million-year evolutionary expansion of hominin neocortices to the current enormity enabled humans to take over the planet. However, there appears to have been a glitch, and it occurred without a compensatory expansion of the entorhinal cortical (EC) gateway to the hippocampal memory-encoding system needed to manage the processing of the increasing volume of neocortical data converging on it. The resulting age-dependent connectopathic glitch was unnoticed by the early short-lived populations. It has now surfaced as Alzheimer's disease (AD) in today's long-lived populations. With advancing age, processing of the converging neocortical data by the neurons of the relatively small lateral entorhinal cortex (LEC) inflicts persistent strain and high energy costs on these cells. This may result in their hyper-release of harmless Aβ1-42 monomers into the interstitial fluid, where they seed the formation of toxic amyloid-β oligomers (AβOs) that initiate AD. At the core of connectopathic AD are the postsynaptic cellular prion protein (PrPC). Electrostatic binding of the negatively charged AβOs to the positively charged N-terminus of PrPC induces hyperphosphorylation of tau that destroys synapses. The spread of these accumulating AβOs from ground zero is supported by Aβ's own production mediated by target cells' Ca2+-sensing receptors (CaSRs). These data suggest that an early administration of a strongly positively charged, AβOs-interacting peptide or protein, plus an inhibitor of CaSR, might be an effective AD-arresting therapeutic combination.
Collapse
Affiliation(s)
- James F. Whitfield
- Human Health Therapeutics, National Research Council, Ottawa, ON K1A 0R6, Canada
| | | | | |
Collapse
|
34
|
Liu X, Jiang N, Zhou W. Various Energetic Metabolism of Microglia in Response to Different Stimulations. Molecules 2023; 28:molecules28114501. [PMID: 37298976 DOI: 10.3390/molecules28114501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/19/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
The activation of the microglia plays an important role in the neuroinflammation induced by different stimulations associated with Alzheimer's disease (AD). Different stimulations, such as pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs) and cytokines, trigger a consequence of activation in the microglia with diverse changes of the microglial cell type response in AD. The activation of the microglia is often accompanied by metabolic changes in response to PAMPs, DAMPs and cytokines in AD. Actually, we do not know the distinct differences on the energetic metabolism of microglia when subject to these stimuli. This research assessed the changes of the cell type response and energetic metabolism in mouse-derived immortalized cells (BV-2 cells) induced by a PAMP (LPS), DAMPs (Aβ and ATP) and a cytokine (IL-4) in mouse-derived immortalized cells (BV-2 cells) and whether the microglial cell type response was improved by targeting the metabolism. We uncovered that LPS, a proinflammatory stimulation of PAMPs, modified the morphology from irregular to fusiform, with stronger cell viability, fusion rates and phagocytosis in the microglia accompanied by a metabolic shift to the promotion of glycolysis and the inhibition of oxidative phosphorylation (OXPHOS). Aβ and ATP, which are two known kinds of DAMPs that trigger microglial sterile activation, induced the morphology from irregular to amoebic, and significantly decreased others in the microglia, accompanied by boosting or reducing both glycolysis and OXPHOS. Monotonous pathological changes and energetic metabolism of microglia were observed under IL-4 exposure. Further, the inhibition of glycolysis transformed the LPS-induced proinflammatory morphology and decreased the enhancement of LPS-induced cell viability, the fusion rate and phagocytosis. However, the promotion of glycolysis exerted a minimal effect on the changes of morphology, the fusion rate, cell viability and phagocytosis induced by ATP. Our study reveals that microglia induced diverse pathological changes accompanied by various changes in the energetic metabolism in response to PAMPs, DAMPs and cytokines, and it may be a potential application of targeting the cellular metabolism to interfere with the microglia-mediated pathological changes in AD.
Collapse
Affiliation(s)
- Xiaohui Liu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasure, Beijing 100850, China
| | - Ning Jiang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasure, Beijing 100850, China
| | - Wenxia Zhou
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasure, Beijing 100850, China
| |
Collapse
|
35
|
Qu W, Canoll P, Hargus G. Molecular Insights into Cell Type-specific Roles in Alzheimer's Disease: Human Induced Pluripotent Stem Cell-based Disease Modelling. Neuroscience 2023; 518:10-26. [PMID: 35569647 PMCID: PMC9974106 DOI: 10.1016/j.neuroscience.2022.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia resulting in widespread degeneration of the central nervous system with severe cognitive impairment. Despite the devastating toll of AD, the incomplete understanding of the complex molecular mechanisms hinders the expeditious development of effective cures. Emerging evidence from animal studies has shown that different brain cell types play distinct roles in the pathogenesis of AD. Glutamatergic neurons are preferentially affected in AD and pronounced gliosis contributes to the progression of AD in both a cell-autonomous and a non-cell-autonomous manner. Much has been discovered through genetically modified animal models, yet frequently failed translational attempts to clinical applications call for better disease models. Emerging evidence supports the significance of human-induced pluripotent stem cell (iPSC) derived brain cells in modeling disease development and progression, opening new avenues for the discovery of molecular mechanisms. This review summarizes the function of different cell types in the pathogenesis of AD, such as neurons, microglia, and astrocytes, and recognizes the potential of utilizing the rapidly growing iPSC technology in modeling AD.
Collapse
Affiliation(s)
- Wenhui Qu
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.
| |
Collapse
|
36
|
Lee S, Devanney NA, Golden LR, Smith CT, Schwartz JL, Walsh AE, Clarke HA, Goulding DS, Allenger EJ, Morillo-Segovia G, Friday CM, Gorman AA, Hawkinson TR, MacLean SM, Williams HC, Sun RC, Morganti JM, Johnson LA. APOE modulates microglial immunometabolism in response to age, amyloid pathology, and inflammatory challenge. Cell Rep 2023; 42:112196. [PMID: 36871219 PMCID: PMC10117631 DOI: 10.1016/j.celrep.2023.112196] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/29/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
The E4 allele of Apolipoprotein E (APOE) is associated with both metabolic dysfunction and a heightened pro-inflammatory response: two findings that may be intrinsically linked through the concept of immunometabolism. Here, we combined bulk, single-cell, and spatial transcriptomics with cell-specific and spatially resolved metabolic analyses in mice expressing human APOE to systematically address the role of APOE across age, neuroinflammation, and AD pathology. RNA sequencing (RNA-seq) highlighted immunometabolic changes across the APOE4 glial transcriptome, specifically in subsets of metabolically distinct microglia enriched in the E4 brain during aging or following an inflammatory challenge. E4 microglia display increased Hif1α expression and a disrupted tricarboxylic acid (TCA) cycle and are inherently pro-glycolytic, while spatial transcriptomics and mass spectrometry imaging highlight an E4-specific response to amyloid that is characterized by widespread alterations in lipid metabolism. Taken together, our findings emphasize a central role for APOE in regulating microglial immunometabolism and provide valuable, interactive resources for discovery and validation research.
Collapse
Affiliation(s)
- Sangderk Lee
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Nicholas A Devanney
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Lesley R Golden
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Cathryn T Smith
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - James L Schwartz
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Adeline E Walsh
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Harrison A Clarke
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Danielle S Goulding
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | | | | | - Cassi M Friday
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Amy A Gorman
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Tara R Hawkinson
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Steven M MacLean
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Holden C Williams
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Ramon C Sun
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Josh M Morganti
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA.
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
37
|
Yassine HN, Self W, Kerman BE, Santoni G, Navalpur Shanmugam N, Abdullah L, Golden LR, Fonteh AN, Harrington MG, Gräff J, Gibson GE, Kalaria R, Luchsinger JA, Feldman HH, Swerdlow RH, Johnson LA, Albensi BC, Zlokovic BV, Tanzi R, Cunnane S, Samieri C, Scarmeas N, Bowman GL. Nutritional metabolism and cerebral bioenergetics in Alzheimer's disease and related dementias. Alzheimers Dement 2023; 19:1041-1066. [PMID: 36479795 PMCID: PMC10576546 DOI: 10.1002/alz.12845] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/12/2022] [Accepted: 10/05/2022] [Indexed: 12/13/2022]
Abstract
Disturbances in the brain's capacity to meet its energy demand increase the risk of synaptic loss, neurodegeneration, and cognitive decline. Nutritional and metabolic interventions that target metabolic pathways combined with diagnostics to identify deficits in cerebral bioenergetics may therefore offer novel therapeutic potential for Alzheimer's disease (AD) prevention and management. Many diet-derived natural bioactive components can govern cellular energy metabolism but their effects on brain aging are not clear. This review examines how nutritional metabolism can regulate brain bioenergetics and mitigate AD risk. We focus on leading mechanisms of cerebral bioenergetic breakdown in the aging brain at the cellular level, as well as the putative causes and consequences of disturbed bioenergetics, particularly at the blood-brain barrier with implications for nutrient brain delivery and nutritional interventions. Novel therapeutic nutrition approaches including diet patterns are provided, integrating studies of the gut microbiome, neuroimaging, and other biomarkers to guide future personalized nutritional interventions.
Collapse
Affiliation(s)
- Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern, California, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wade Self
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bilal E Kerman
- Department of Medicine, Keck School of Medicine, University of Southern, California, Los Angeles, California, USA
| | - Giulia Santoni
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland
| | - NandaKumar Navalpur Shanmugam
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Lesley R Golden
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alfred N Fonteh
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Huntington Medical Research Institutes, Pasadena, California, USA
| | - Michael G Harrington
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland
| | - Gary E Gibson
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, New York, USA
| | - Raj Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jose A Luchsinger
- Department of Medicine and Epidemiology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Howard H Feldman
- Department of Neurosciences, University of California, San Diego, California, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Benedict C Albensi
- Nova Southeastern Univ. College of Pharmacy, Davie, Florida, USA
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rudolph Tanzi
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Cécilia Samieri
- Univ. Bordeaux, INSERM, BPH, U1219, F-33000, Bordeaux, France
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, Columbia University, New York City, New York, USA
| | - Gene L Bowman
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Helfgott Research Institute, National University of Natural Medicine, Portland, Oregon, USA
| |
Collapse
|
38
|
Lysikova EA, Kuzubova EV, Radchenko AI, Patrakhanov EA, Chaprov KD, Korokin MV, Deykin AV, Gudyrev OS, Pokrovskii MV. APPswe/PS1dE9/Blg Transgenic Mouse Line for Modeling Cerebral Amyloid Angiopathy Associated with Alzheimer’s Disease. Mol Biol 2023. [DOI: 10.1134/s0026893323010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
39
|
Mathews M, Wißfeld J, Flitsch LJ, Shahraz A, Semkova V, Breitkreuz Y, Neumann H, Brüstle O. Reenacting Neuroectodermal Exposure of Hematopoietic Progenitors Enables Scalable Production of Cryopreservable iPSC-Derived Human Microglia. Stem Cell Rev Rep 2023; 19:455-474. [PMID: 35971018 PMCID: PMC9902330 DOI: 10.1007/s12015-022-10433-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 02/07/2023]
Abstract
Human microglia, as innate immune cells of the central nervous system (CNS), play a central role in the pathogenesis of a large number of neurological and psychiatric disorders. However, experimental access to primary human microglia for biomedical applications such as disease modeling is extremely limited. While induced pluripotent stem cells (iPSCs) could provide an alternative source of microglia, the reenactment of their complex ontogenesis with a yolk sac origin and subsequent priming upon CNS invasion has remained a challenge. Here, we report a developmentally informed in vitro differentiation method for large-scale production and cryopreservation of iPSC-derived microglia (iPSdMiG). Specifically, iPSCs were propagated in conditions yielding both yolk sac hematopoietic derivatives and early neuroepithelial cells. To enable large-scale production, we implemented 3D bioreactor-based dynamic culture conditions and the use of novel mesh macrocarriers. Under these conditions, microglia could be harvested across a time period of at least 6 weeks, with 1 × 106 iPSCs giving rise to up to 45 × 106 iPSdMiG. The transcriptomic profile of iPSdMiG showed high similarity to adult human microglia, and harvested cells were immunopositive for typical microglial markers. In addition, iPSdMiG were able to secrete pro-inflammatory cytokines, engaged in phagocytotic activity, produced reactive oxygen species and lent themselves to co-culture studies in neural 2D and 3D systems. Importantly, iPSdMiG were efficiently cryopreserved, enabling the establishment of donor-specific microglia cell banks for disease modeling, drug discovery and eventually cell therapy. Main points. Scalable generation of iPSC-derived multi-lineage embryoid bodies on macrocarriers, reproducibly releasing microglia exhibiting characteristic markers and function. Cells are transcriptomically similar to primary human microglia and cryopreservable.
Collapse
Affiliation(s)
- Mona Mathews
- LIFE & BRAIN GmbH, Venusberg-Campus 1, 53127, Bonn, Germany.,Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jannis Wißfeld
- Institute of Reconstructive Neurobiology, Neural Regeneration Group, University of Bonn Medical Faculty and University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Lea Jessica Flitsch
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Anahita Shahraz
- Institute of Reconstructive Neurobiology, Neural Regeneration Group, University of Bonn Medical Faculty and University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Vesselina Semkova
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Yannik Breitkreuz
- LIFE & BRAIN GmbH, Venusberg-Campus 1, 53127, Bonn, Germany.,Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, Neural Regeneration Group, University of Bonn Medical Faculty and University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Oliver Brüstle
- LIFE & BRAIN GmbH, Venusberg-Campus 1, 53127, Bonn, Germany. .,Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
40
|
Zhang X, Wu L, Swerdlow RH, Zhao L. Opposing Effects of ApoE2 and ApoE4 on Glycolytic Metabolism in Neuronal Aging Supports a Warburg Neuroprotective Cascade against Alzheimer's Disease. Cells 2023; 12:410. [PMID: 36766752 PMCID: PMC9914046 DOI: 10.3390/cells12030410] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Apolipoprotein E4 (ApoE4) is the most recognized genetic risk factor for late-onset Alzheimer's disease (LOAD), whereas ApoE2 reduces the risk for LOAD. The underlying mechanisms are unclear but may include effects on brain energy metabolism. Here, we used neuro-2a (N2a) cells that stably express human ApoE isoforms (N2a-hApoE), differentiated N2a-hApoE neuronal cells, and humanized ApoE knock-in mouse models to investigate relationships among ApoE isoforms, glycolytic metabolism, and neuronal health and aging. ApoE2-expressing cells retained robust hexokinase (HK) expression and glycolytic activity, whereas these endpoints progressively declined with aging in ApoE4-expressing cells. These divergent ApoE2 and ApoE4 effects on glycolysis directly correlated with markers of cellular wellness. Moreover, ApoE4-expressing cells upregulated phosphofructokinase and pyruvate kinase with the apparent intent of compensating for the HK-dependent glycolysis reduction. The introduction of ApoE2 increased HK levels and glycolysis flux in ApoE4 cells. PI3K/Akt signaling was distinctively regulated by ApoE isoforms but was only partially responsible for the ApoE-mediated effects on HK. Collectively, our findings indicate that human ApoE isoforms differentially modulate neuronal glycolysis through HK regulation, with ApoE2 upregulating and ApoE4 downregulating, which markedly impacts neuronal health during aging. These findings lend compelling support to the emerging inverse-Warburg theory of AD and highlight a therapeutic opportunity for bolstering brain glycolytic resilience to prevent and treat AD.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Long Wu
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Russell H. Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
41
|
Warden AS, Han C, Hansen E, Trescott S, Nguyen C, Kim R, Schafer D, Johnson A, Wright M, Ramirez G, Lopez-Sanchez M, Coufal NG. Tools for studying human microglia: In vitro and in vivo strategies. Brain Behav Immun 2023; 107:369-382. [PMID: 36336207 PMCID: PMC9810377 DOI: 10.1016/j.bbi.2022.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/11/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Microglia may only represent 10% of central nervous system (CNS) cells but they perform critical roles in development, homeostasis and neurological disease. Microglia are also environmentally regulated, quickly losing their transcriptomic and epigenetic signature after leaving the CNS. This facet of microglia biology is both fascinating and technically challenging influencing the study of the genetics and function of human microglia in a manner that recapitulates the CNS environment. In this review we provide a comprehensive overview of existing in vitro and in vivo methodology to study human microglia, such as immortalized cells lines, stem cell-derived microglia, cerebral organoids and xenotransplantation. Since there is currently no single method that completely recapitulates all hallmarks of human ex vivo adult homeostatic microglia, we also discuss the advantages and limitations of each existing model as a practical guide for researchers.
Collapse
Affiliation(s)
- Anna S Warden
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Claudia Han
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Emily Hansen
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Samantha Trescott
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Celina Nguyen
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Roy Kim
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Danielle Schafer
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Avalon Johnson
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Madison Wright
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gabriela Ramirez
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark Lopez-Sanchez
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicole G Coufal
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
42
|
iPSC-Derived Macrophages: The Differentiation Protocol Affects Cell Immune Characteristics and Differentiation Trajectories. Int J Mol Sci 2022; 23:ijms232416087. [PMID: 36555728 PMCID: PMC9781144 DOI: 10.3390/ijms232416087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
The generation of human macrophages from induced pluripotent stem cells (iMacs) is a rapidly developing approach used to create disease models, screen drugs, study macrophage-pathogen interactions and develop macrophage-based cell therapy. To generate iMacs, different types of protocols have been suggested, all thought to result in the generation of similar iMac populations. However, direct comparison of iMacs generated using different protocols has not been performed. We have compared the productivity, the differentiation trajectories and the characteristics of iMacs generated using two widely used protocols: one based on the formation of embryoid bodies and the induction of myeloid differentiation by only two cytokines, interleukin-3 and macrophage colony-stimulating factor, and the other utilizing multiple exogenous factors for iMac generation. We report inter-protocol differences in the following: (i) protocol productivity; (ii) dynamic changes in the expression of genes related to inflammation and lipid homeostasis following iMac differentiation and (iii) the transcriptomic profiles of terminally differentiated iMacs, including the expression of genes involved in inflammatory response, antigen presentation and lipid homeostasis. The results document the dependence of fine iMac characteristics on the type of differentiation protocol, which is important for further development of the field, including the development of iMac-based cell therapy.
Collapse
|
43
|
Pantazis CB, Yang A, Lara E, McDonough JA, Blauwendraat C, Peng L, Oguro H, Kanaujiya J, Zou J, Sebesta D, Pratt G, Cross E, Blockwick J, Buxton P, Kinner-Bibeau L, Medura C, Tompkins C, Hughes S, Santiana M, Faghri F, Nalls MA, Vitale D, Ballard S, Qi YA, Ramos DM, Anderson KM, Stadler J, Narayan P, Papademetriou J, Reilly L, Nelson MP, Aggarwal S, Rosen LU, Kirwan P, Pisupati V, Coon SL, Scholz SW, Priebe T, Öttl M, Dong J, Meijer M, Janssen LJM, Lourenco VS, van der Kant R, Crusius D, Paquet D, Raulin AC, Bu G, Held A, Wainger BJ, Gabriele RMC, Casey JM, Wray S, Abu-Bonsrah D, Parish CL, Beccari MS, Cleveland DW, Li E, Rose IVL, Kampmann M, Calatayud Aristoy C, Verstreken P, Heinrich L, Chen MY, Schüle B, Dou D, Holzbaur ELF, Zanellati MC, Basundra R, Deshmukh M, Cohen S, Khanna R, Raman M, Nevin ZS, Matia M, Van Lent J, Timmerman V, Conklin BR, Johnson Chase K, Zhang K, Funes S, Bosco DA, Erlebach L, Welzer M, Kronenberg-Versteeg D, Lyu G, Arenas E, Coccia E, Sarrafha L, Ahfeldt T, Marioni JC, Skarnes WC, Cookson MR, Ward ME, Merkle FT. A reference human induced pluripotent stem cell line for large-scale collaborative studies. Cell Stem Cell 2022; 29:1685-1702.e22. [PMID: 36459969 PMCID: PMC9782786 DOI: 10.1016/j.stem.2022.11.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 10/07/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022]
Abstract
Human induced pluripotent stem cell (iPSC) lines are a powerful tool for studying development and disease, but the considerable phenotypic variation between lines makes it challenging to replicate key findings and integrate data across research groups. To address this issue, we sub-cloned candidate human iPSC lines and deeply characterized their genetic properties using whole genome sequencing, their genomic stability upon CRISPR-Cas9-based gene editing, and their phenotypic properties including differentiation to commonly used cell types. These studies identified KOLF2.1J as an all-around well-performing iPSC line. We then shared KOLF2.1J with groups around the world who tested its performance in head-to-head comparisons with their own preferred iPSC lines across a diverse range of differentiation protocols and functional assays. On the strength of these findings, we have made KOLF2.1J and its gene-edited derivative clones readily accessible to promote the standardization required for large-scale collaborative science in the stem cell field.
Collapse
Affiliation(s)
- Caroline B Pantazis
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Andrian Yang
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Wellcome Trust - Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Erika Lara
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Lirong Peng
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International LLC, Washington, DC, USA; Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Hideyuki Oguro
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Jitendra Kanaujiya
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Jizhong Zou
- iPS Cell Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | - Marianita Santiana
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Faraz Faghri
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International LLC, Washington, DC, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International LLC, Washington, DC, USA
| | - Daniel Vitale
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International LLC, Washington, DC, USA
| | - Shannon Ballard
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International LLC, Washington, DC, USA
| | - Yue A Qi
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Daniel M Ramos
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kailyn M Anderson
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Julia Stadler
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Priyanka Narayan
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Genetics and Biochemistry Branch, NIDDK, NINDS, National Institutes of Health, Bethesda, MD 20814, USA
| | - Jason Papademetriou
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Luke Reilly
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Matthew P Nelson
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sanya Aggarwal
- Wellcome Trust - Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Leah U Rosen
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Peter Kirwan
- Wellcome Trust - Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Venkat Pisupati
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK; John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK
| | - Steven L Coon
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA; Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Theresa Priebe
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands
| | - Miriam Öttl
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands
| | - Jian Dong
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands
| | - Marieke Meijer
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands
| | - Lara J M Janssen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands
| | - Vanessa S Lourenco
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands
| | - Rik van der Kant
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands; Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - Dennis Crusius
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | | | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Aaron Held
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian J Wainger
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA; Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Rebecca M C Gabriele
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Jackie M Casey
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Dad Abu-Bonsrah
- The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia; Department of Pediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Melinda S Beccari
- Department of Cellular and Molecular Medicine and Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, CA, USA
| | - Don W Cleveland
- Department of Cellular and Molecular Medicine and Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, CA, USA
| | - Emmy Li
- Institute for Neurodegenerative Diseases and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Indigo V L Rose
- Institute for Neurodegenerative Diseases and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases and Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Carles Calatayud Aristoy
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Laurin Heinrich
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Max Y Chen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dan Dou
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria Clara Zanellati
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Richa Basundra
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah Cohen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Richa Khanna
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Malavika Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | | | | | - Jonas Van Lent
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp 2610, Belgium
| | | | | | - Ke Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Salome Funes
- Department of Neurology, UMass Chan Medical School, Worcester, MA, USA
| | - Daryl A Bosco
- Department of Neurology, UMass Chan Medical School, Worcester, MA, USA
| | - Lena Erlebach
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Marc Welzer
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Deborah Kronenberg-Versteeg
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Guochang Lyu
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Elena Coccia
- Nash Family Department of Neuroscience; Departments of Neurology and Cell, Developmental and Regenerative Biology; Ronald M. Loeb Center for Alzheimer's Disease; Friedman Brain Institute; Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA
| | - Lily Sarrafha
- Nash Family Department of Neuroscience; Departments of Neurology and Cell, Developmental and Regenerative Biology; Ronald M. Loeb Center for Alzheimer's Disease; Friedman Brain Institute; Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA
| | - Tim Ahfeldt
- Nash Family Department of Neuroscience; Departments of Neurology and Cell, Developmental and Regenerative Biology; Ronald M. Loeb Center for Alzheimer's Disease; Friedman Brain Institute; Black Family Stem Cell Institute at Mount Sinai, New York, NY, USA
| | - John C Marioni
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Mark R Cookson
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
| | - Michael E Ward
- Center for Alzheimer's and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Florian T Merkle
- Wellcome Trust - Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK.
| |
Collapse
|
44
|
Hübschmann V, Korkut-Demirbaş M, Siegert S. Assessing human iPSC-derived microglia identity and function by immunostaining, phagocytosis, calcium activity, and inflammation assay. STAR Protoc 2022; 3:101866. [PMID: 36595902 PMCID: PMC9678782 DOI: 10.1016/j.xpro.2022.101866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/28/2022] [Accepted: 10/28/2022] [Indexed: 11/19/2022] Open
Abstract
To understand how potential gene manipulations affect in vitro microglia, we provide a set of short protocols to evaluate microglia identity and function. We detail steps for immunostaining to determine microglia identity. We describe three functional assays for microglia: phagocytosis, calcium response following ATP stimulation, and cytokine expression upon inflammatory stimuli. We apply these protocols to human induced-pluripotent-stem-cell (hiPSC)-derived microglia, but they can be also applied to other in vitro microglial models including primary mouse microglia. For complete details on the use and execution of this protocol, please refer to Bartalska et al. (2022).1.
Collapse
Affiliation(s)
- Verena Hübschmann
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Medina Korkut-Demirbaş
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria.
| | - Sandra Siegert
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
45
|
Washer SJ, Perez-Alcantara M, Chen Y, Steer J, James WS, Trynka G, Bassett AR, Cowley SA. Single-cell transcriptomics defines an improved, validated monoculture protocol for differentiation of human iPSC to microglia. Sci Rep 2022; 12:19454. [PMID: 36376339 PMCID: PMC9663826 DOI: 10.1038/s41598-022-23477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/01/2022] [Indexed: 11/15/2022] Open
Abstract
There is increasing genetic evidence for the role of microglia in neurodegenerative diseases, including Alzheimer's, Parkinson's, and motor neuron disease. Therefore, there is a need to generate authentic in vitro models to study human microglial physiology. Various methods have been developed using human induced Pluripotent Stem Cells (iPSC) to generate microglia, however, systematic approaches to identify which media components are actually essential for functional microglia are mostly lacking. Here, we systematically assess medium components, coatings, and growth factors required for iPSC differentiation to microglia. Using single-cell RNA sequencing, qPCR, and functional assays, with validation across two labs, we have identified several medium components from previous protocols that are redundant and do not contribute to microglial identity. We provide an optimised, defined medium which produces both transcriptionally and functionally relevant microglia for modelling microglial physiology in neuroinflammation and for drug discovery.
Collapse
Affiliation(s)
- Sam J Washer
- James and Lillian Martin Centre for Stem Cell Research, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK. .,Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK. .,Open Targets, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.
| | - Marta Perez-Alcantara
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Yixi Chen
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Juliette Steer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - William S James
- James and Lillian Martin Centre for Stem Cell Research, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Gosia Trynka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Andrew R Bassett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Sally A Cowley
- James and Lillian Martin Centre for Stem Cell Research, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| |
Collapse
|
46
|
Cakir B, Kiral FR, Park IH. Advanced in vitro models: Microglia in action. Neuron 2022; 110:3444-3457. [PMID: 36327894 DOI: 10.1016/j.neuron.2022.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
In the central nervous system (CNS), microglia carry out multiple tasks related to brain development, maintenance of brain homeostasis, and function of the CNS. Recent advanced in vitro model systems allow us to perform more detailed and specific analyses of microglial functions in the CNS. The development of human pluripotent stem cells (hPSCs)-based 2D and 3D cell culture methods, particularly advancements in brain organoid models, offers a better platform to dissect microglial function in various contexts. Despite the improvement of these methods, there are still definite restrictions. Understanding their drawbacks and benefits ensures their proper use. In this primer, we review current developments regarding in vitro microglial production and characterization and their use to address fundamental questions about microglial function in healthy and diseased states, and we discuss potential future improvements with a particular emphasis on brain organoid models.
Collapse
Affiliation(s)
- Bilal Cakir
- Department of Genetics, Yale Stem Cell Center, Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Ferdi Ridvan Kiral
- Department of Genetics, Yale Stem Cell Center, Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
47
|
Pinals RL, Tsai LH. Building in vitro models of the brain to understand the role of APOE in Alzheimer's disease. Life Sci Alliance 2022; 5:5/11/e202201542. [PMID: 36167428 PMCID: PMC9515460 DOI: 10.26508/lsa.202201542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating, complex, and incurable disease that represents an increasingly problematic global health issue. The etiology of sporadic AD that accounts for a vast majority of cases remains poorly understood, with no effective therapeutic interventions. Genetic studies have identified AD risk genes including the most prominent, APOE, of which the ɛ4 allele increases risk in a dose-dependent manner. A breakthrough discovery enabled the creation of human induced pluripotent stem cells (hiPSCs) that can be differentiated into various brain cell types, facilitating AD research in genetically human models. Herein, we provide a brief background on AD in the context of APOE susceptibility and feature work employing hiPSC-derived brain cell and tissue models to interrogate the contribution of APOE in driving AD pathology. Such models have delivered crucial insights into cellular mechanisms and cell type-specific roles underlying the perturbed biological functions that trigger pathogenic cascades and propagate neurodegeneration. Collectively, hiPSC-based models are envisioned to be an impactful platform for uncovering fundamental AD understanding, with high translational value toward AD drug discovery and testing.
Collapse
Affiliation(s)
- Rebecca L Pinals
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA .,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.,Broad Institute of Harvard and MIT, Cambridge, MA, USA
| |
Collapse
|
48
|
Nazarian A, Loiko E, Yassine HN, Finch CE, Kulminski AM. APOE alleles modulate associations of plasma metabolites with variants from multiple genes on chromosome 19q13.3. Front Aging Neurosci 2022; 14:1023493. [PMID: 36389057 PMCID: PMC9650319 DOI: 10.3389/fnagi.2022.1023493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
The APOE ε2, ε3, and ε4 alleles differentially impact various complex diseases and traits. We examined whether these alleles modulated associations of 94 single-nucleotide polymorphisms (SNPs) harbored by 26 genes in 19q13.3 region with 217 plasma metabolites using Framingham Heart Study data. The analyses were performed in the E2 (ε2ε2 or ε2ε3 genotype), E3 (ε3ε3 genotype), and E4 (ε3ε4 or ε4ε4 genotype) groups separately. We identified 31, 17, and 22 polymorphism-metabolite associations in the E2, E3, and E4 groups, respectively, at a false discovery rate P FDR < 0.05. These entailed 51 and 19 associations with 20 lipid and 12 polar analytes. Contrasting the effect sizes between the analyzed groups showed 20 associations with group-specific effects at Bonferroni-adjusted P < 7.14E-04. Three associations with glutamic acid or dimethylglycine had significantly larger effects in the E2 than E3 group and 12 associations with triacylglycerol 56:5, lysophosphatidylethanolamines 16:0, 18:0, 20:4, or phosphatidylcholine 38:6 had significantly larger effects in the E2 than E4 group. Two associations with isocitrate or propionate and three associations with phosphatidylcholines 32:0, 32:1, or 34:0 had significantly larger effects in the E4 than E3 group. Nine of 70 SNP-metabolite associations identified in either E2, E3, or E4 groups attained P FDR < 0.05 in the pooled sample of these groups. However, none of them were among the 20 group-specific associations. Consistent with the evolutionary history of the APOE alleles, plasma metabolites showed higher APOE-cluster-related variations in the E4 than E2 and E3 groups. Pathway enrichment mainly highlighted lipids and amino acids metabolism and citrate cycle, which can be differentially impacted by the APOE alleles. These novel findings expand insights into the genetic heterogeneity of plasma metabolites and highlight the importance of the APOE-allele-stratified genetic analyses of the APOE-related diseases and traits.
Collapse
Affiliation(s)
- Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Elena Loiko
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Hussein N. Yassine
- Departments of Medicine and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Caleb E. Finch
- Andrus Gerontology Center, University of Southern California, Los Angeles, CA, United States
| | - Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| |
Collapse
|
49
|
Garcia G, Pinto S, Ferreira S, Lopes D, Serrador MJ, Fernandes A, Vaz AR, de Mendonça A, Edenhofer F, Malm T, Koistinaho J, Brites D. Emerging Role of miR-21-5p in Neuron-Glia Dysregulation and Exosome Transfer Using Multiple Models of Alzheimer's Disease. Cells 2022; 11:3377. [PMID: 36359774 PMCID: PMC9655962 DOI: 10.3390/cells11213377] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 08/25/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with neuron-glia dysfunction and dysregulated miRNAs. We previously reported upregulated miR-124/miR-21 in AD neurons and their exosomes. However, their glial distribution, phenotypic alterations and exosomal spread are scarcely documented. Here, we show glial cell activation and miR-21 overexpression in mouse organotypic hippocampal slices transplanted with SH-SY5Y cells expressing the human APP695 Swedish mutation. The upregulation of miR-21 only in the CSF from a small series of mild cognitive impairment (MCI) AD patients, but not in non-AD MCI individuals, supports its discriminatory potential. Microglia, neurons, and astrocytes differentiated from the same induced pluripotent stem cells from PSEN1ΔE9 AD patients all showed miR-21 elevation. In AD neurons, miR-124/miR-21 overexpression was recapitulated in their exosomes. In AD microglia, the upregulation of iNOS and miR-21/miR-146a supports their activation. AD astrocytes manifested a restrained inflammatory profile, with high miR-21 but low miR-155 and depleted exosomal miRNAs. Their immunostimulation with C1q + IL-1α + TNF-α induced morphological alterations and increased S100B, inflammatory transcripts, sAPPβ, cytokine release and exosomal miR-21. PPARα, a target of miR-21, was found to be repressed in all models, except in neurons, likely due to concomitant miR-125b elevation. The data from these AD models highlight miR-21 as a promising biomarker and a disease-modifying target to be further explored.
Collapse
Affiliation(s)
- Gonçalo Garcia
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Sara Pinto
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Sofia Ferreira
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Daniela Lopes
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Maria João Serrador
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Adelaide Fernandes
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Central Nervous System, Blood and Peripheral Inflammation Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Ana Rita Vaz
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | | | - Frank Edenhofer
- Department of Genomics, Stem Cell Biology and Regenerative Medicine, Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
| | - Dora Brites
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| |
Collapse
|
50
|
Harerimana NV, Paliwali D, Romero-Molina C, Bennett DA, Pa J, Goate A, Swerdlow RH, Andrews SJ. The role of mitochondrial genome abundance in Alzheimer's disease. Alzheimers Dement 2022; 19:2069-2083. [PMID: 36224040 DOI: 10.1002/alz.12812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/1912] [Revised: 12/12/1912] [Accepted: 12/12/1912] [Indexed: 11/05/2022]
Abstract
Mitochondrial dysfunction is an early and prominent feature of Alzheimer's disease (AD), with impaired energy metabolism preceding the onset of clinical symptoms. Here we propose an update to the mitochondrial dysfunction hypothesis of AD based on recent results examining the role of mitochondrial genome abundance in AD. In a large post mortem study, we show that lower brain mitochondrial genome abundance is associated with a greater odds of AD neuropathological change and worse cognitive performance. We hypothesize that lower mitochondrial genome abundance impairs mitochondrial function by reducing mitochondrial bioenergetics, thereby impacting neuronal and glial cell function. However, it remains to be determined if mitochondrial dysfunction causes, mediates, or is a by-product of AD pathogenesis. Additional support for this hypothesis will be generated by linking peripheral blood mitochondrial genome abundance to AD and establishing clinical trials of compounds that upregulate total mitochondrial genome abundance or boost mitochondrial mass.
Collapse
Affiliation(s)
- Nadia V Harerimana
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Devashi Paliwali
- Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Carmen Romero-Molina
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Judy Pa
- Department of Neurosciences, Alzheimer's Disease Cooperative Study (ADCS), University of California, San Diego, California, USA
| | - Alison Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, Kansas City, Kansas, USA
| | - Shea J Andrews
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|