Category Archives: Induced Pluripotent Stem Cells


Neurite outgrowth deficits caused by rare PLXNB1 mutation in … – Nature.com

Bebbington P, Ramana R. The epidemiology of bipolar affective disorder. Soc Psychiatry Psychiatr Epidemiol. 1995;30:27992.

Article CAS PubMed Google Scholar

Pini S, de Queiroz V, Pagnin D, Pezawas L, Angst J, Cassano GB, et al. Prevalence and burden of bipolar disorders in European countries. Eur Neuropsychopharmacol. 2005;15:42534.

Article CAS PubMed Google Scholar

Burton CZ, Ryan KA, Kamali M, Marshall DF, Harrington G, McInnis MG, et al. Psychosis in bipolar disorder: does it represent a more severe illness? Bipolar Disord. 2018;20:1826.

Article PubMed Google Scholar

Brus MJ, Solanto MV, Goldberg JF. Adult ADHD vs. bipolar disorder in the DSM-5 era: a challenging differentiation for clinicians. J Psychiatr Pract. 2014;20:42837.

Article PubMed Google Scholar

Schulze TG, Akula N, Breuer R, Steele J, Nalls MA, Singleton AB, et al. Molecular genetic overlap in bipolar disorder, schizophrenia, and major depressive disorder. World J Biol Psychiatry. 2014;15:2008.

Article PubMed Google Scholar

Smeland OB, Bahrami S, Frei O, Shadrin A, OConnell K, Savage J, et al. Genome-wide analysis reveals extensive genetic overlap between schizophrenia, bipolar disorder, and intelligence. Mol Psychiatry. 2020;25:84453.

Article CAS PubMed Google Scholar

Bipolar Disorder and Schizophrenia Working Group of the Psychiatric Genomics Consortium. Genomic dissection of bipolar disorder and schizophrenia, including 28 subphenotypes. Cell. 2018;173:170515.e16.

Article PubMed Central Google Scholar

Weller EB, Weller RA, Fristad MA. Bipolar disorder in children: misdiagnosis, underdiagnosis, and future directions. J Am Acad Child Adolesc Psychiatry. 1995;34:70914.

Article CAS PubMed Google Scholar

Renk K, White R, Lauer BA, McSwiggan M, Puff J, Lowell A. Bipolar disorder in children. Psychiatry J 2014;2014:119.

Article Google Scholar

Beyer DKE, Freund N. Animal models for bipolar disorder: from bedside to the cage. Int J Bipolar Disord. 2017;5:35.

Article PubMed PubMed Central Google Scholar

Strakowski SM, DelBello MP, Adler CM. The functional neuroanatomy of bipolar disorder: a review of neuroimaging findings. Mol Psychiatry. 2005;10:10516.

Article CAS PubMed Google Scholar

Pavuluri MN, OConnor MM, Harral EM, Sweeney JA. An fMRI study of the interface between affective and cognitive neural circuitry in pediatric bipolar disorder. Psychiatry Res. 2008;162:24455.

Article PubMed PubMed Central Google Scholar

Chen CH, Suckling J, Lennox BR, Ooi C, Bullmore ET. A quantitative meta-analysis of fMRI studies in bipolar disorder. Bipolar Disord. 2011;13:115.

Article CAS PubMed Google Scholar

OShea KS, McInnis MG. Neurodevelopmental origins of bipolar disorder: iPSC models. Mol Cell Neurosci. 2016;73:6383.

Article PubMed Google Scholar

Madison JM, Zhou F, Nigam A, Hussain A, Barker DD, Nehme R, et al. Characterization of bipolar disorder patient-specific induced pluripotent stem cells from a family reveals neurodevelopmental and mRNA expression abnormalities. Mol Psychiatry. 2015;20:70317.

Article CAS PubMed PubMed Central Google Scholar

Bavamian S, Mellios N, Lalonde J, Fass DM, Wang J, Sheridan SD, et al. Dysregulation of miR-34a Links neuronal development to genetic risk factors for bipolar disorder. Mol Psychiatry. 2015;20:57384.

Article CAS PubMed PubMed Central Google Scholar

Wang JL, Shamah SM, Sun AX, Waldman ID, Haggarty SJ, Perlis RH. Label-free, live optical imaging of reprogrammed bipolar disorder patient-derived cells reveals a functional correlate of lithium responsiveness. Transl Psychiatry. 2014;4:e4288.

Article CAS PubMed PubMed Central Google Scholar

Mertens J, Wang QW, Kim Y, Yu DX, Pham S, Yang B, et al. Differential responses to lithium in hyperexcitable neurons from patients with bipolar disorder. Nature. 2015;527:959.

Article CAS PubMed PubMed Central Google Scholar

Stern S, Santos R, Marchetto MC, Mendes APD, Rouleau GA, Biesmans S, et al. Neurons derived from patients with bipolar disorder divide into intrinsically different sub-populations of neurons, predicting the patients responsiveness to lithium. Mol Psychiatry. 2018;23:145365.

Article CAS PubMed Google Scholar

Craddock N, Sklar P. Genetics of bipolar disorder. Lancet 2013;381:165462.

Article CAS PubMed Google Scholar

Sullivan PF, Geschwind DH. Defining the genetic, genomic, cellular, and diagnostic architectures of psychiatric disorders. Cell. 2019;177:162.

Article CAS PubMed PubMed Central Google Scholar

Song J, Bergen SE, Kuja-Halkola R, Larsson H, Landn M, Lichtenstein P. Bipolar disorder and its relation to major psychiatric disorders: a family-based study in the Swedish population. Bipolar Disord. 2015;17:18493.

Article PubMed Google Scholar

Hou L, Bergen SE, Akula N, Song J, Hultman CM, Landn M, et al. Genome-wide association study of 40,000 individuals identifies two novel loci associated with bipolar disorder. Hum Mol Genet. 2016;25:338394.

Article CAS PubMed PubMed Central Google Scholar

Mullins N, Forstner AJ, OConnell KS, Coombes B, Coleman JRI, Qiao Z, et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat Genet. 2021;53:81729.

Article CAS PubMed PubMed Central Google Scholar

Sul JH, Service SK, Huang AY, Ramensky V, Hwang SG, Teshiba TM, et al. Contribution of common and rare variants to bipolar disorder susceptibility in extended pedigrees from population isolates. Transl Psychiatry. 2020;10:110.

Article Google Scholar

Toma C, Shaw AD, Allcock RJN, Heath A, Pierce KD, Mitchell PB, et al. An examination of multiple classes of rare variants in extended families with bipolar disorder. Transl Psychiatry. 2018;8:112.

Article Google Scholar

Stahl EA, Breen G, Forstner AJ, McQuillin A, Ripke S, Trubetskoy V, et al. Genome-wide association study identifies 30 loci associated with bipolar disorder. Nat Genet. 2019;51:793803.

Article CAS PubMed PubMed Central Google Scholar

Clifton NE, Hannon E, Harwood JC, di Florio A, Thomas KL, Holmans PA, et al. Dynamic expression of genes associated with schizophrenia and bipolar disorder across development. Transl Psychiatry. 2019;9:74.

Zeng B, Bendl J, Kosoy R, Fullard JF, Hoffman GE, Roussos P. Multi-ancestry eQTL meta-analysis of human brain identifies candidate causal variants for brain-related traits. Nat Genet. 2022;54:1619.

Article CAS PubMed PubMed Central Google Scholar

Ament SA, Szelinger S, Glusman G, Ashworth J, Hou L, Akula N, et al. Rare variants in neuronal excitability genes influence risk for bipolar disorder. Proc Natl Acad Sci USA. 2015;112:357681.

Article CAS PubMed PubMed Central Google Scholar

Kataoka M, Matoba N, Sawada T, Kazuno AA, Ishiwata M, Fujii K, et al. Exome sequencing for bipolar disorder points to roles of de novo loss-of-function and protein-altering mutations. Mol Psychiatry. 2016;21:88593.

Article CAS PubMed PubMed Central Google Scholar

Sullivan PF, Daly MJ, ODonovan M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet. 2012;13:53751.

Article CAS PubMed PubMed Central Google Scholar

Falk A, Heine VM, Harwood AJ, Sullivan PF, Peitz M, Brstle O, et al. Modeling psychiatric disorders: from genomic findings to cellular phenotypes. Mol Psychiatry. 2016;21:116779.

Article CAS PubMed PubMed Central Google Scholar

Ishii T, Ishikawa M, Fujimori K, Maeda T, Kushima I, Arioka Y, et al. In vitro modeling of the bipolar disorder and schizophrenia using patient-derived induced pluripotent stem cells with copy number variations of PCDH15 and RELN. eNeuro. 2019;6:ENEURO.040318.2019.

Article PubMed Google Scholar

Zoghbi AW, Dhindsa RS, Goldberg TE, Mehralizade A, Motelow JE, Wang X, et al. High-impact rare genetic variants in severe schizophrenia. Proc Natl Acad Sci USA. 2021;118:e2112560118.

Lopez-Larson MP, Shah LM, Weeks HR, King JB, Mallik AK, Yurgelun-Todd DA, et al. Abnormal functional connectivity between default and salience networks in pediatric bipolar disorder. Biol Psychiatry Cogn Neurosci Neuroimaging. 2017;2:8593.

PubMed Google Scholar

The Mini-International Neuropsychiatric Interview (M.I.N.I.). the development and validation of a structured diagnostic psychiatric interview for DSM-IV and ICD-10. J Clin Psychiatry. 1998;59:2233.

Google Scholar

Achenbach TM, Rescorla LA. Manual for the ASEBA school-age forms & profiles. Burlington, VT: University of Vermont, Research Center for Children, Youth, & Families; 2001.

Google Scholar

Wechsler D. Wechsler abbreviated scale of intelligence. New York, NY: The Psychological Corporation: Harcourt Brace & Company; 1999.

Google Scholar

Jo HJ, Gotts SJ, Reynolds RC, Bandettini PA, Martin A, Cox RW, et al. Effective preprocessing procedures virtually eliminate distance-dependent motion artifacts in resting state FMRI. J Appl Math. 2013. https://doi.org/10.1155/2013/935154.

Article PubMed PubMed Central Google Scholar

Cox RW. AFNI: software for analysis and visualization of functional magnetic resonance neuroimages. Comput Biomed Res. 1996;29:16273.

Article CAS PubMed Google Scholar

Anderson JS, Ferguson MA, Lopez-Larson M, Yurgelun-Todd D. Reproducibility of single-subject functional connectivity measurements. Am J Neuroradiol. 2011;32:54855.

Article CAS PubMed PubMed Central Google Scholar

Saad ZS, Gotts SJ, Murphy K, Chen G, jo HJ, Martin A, et al. Trouble at rest: how correlation patterns and group differences become distorted after global signal regression. Brain Connect. 2012;2:2532.

Article PubMed PubMed Central Google Scholar

Murphy K, Birn RM, Handwerker DA, Jones TB, Bandettini PA. The impact of global signal regression on resting state correlations: are anti-correlated networks introduced? Neuroimage. 2009;44:893905.

Article PubMed Google Scholar

Power JD, Barnes KA, Snyder AZ, Schlaggar BL, Petersen SE. Spurious but systematic correlations in functional connectivity MRI networks arise from subject motion. Neuroimage. 2012;59:2142.

Article PubMed Google Scholar

Yeo BTT, Krienen FM, Sepulcre J, Sabuncu MR, Lashkari D, Hollinshead M, et al. The organization of the human cerebral cortex estimated by intrinsic functional connectivity. J Neurophysiol. 2011;106:112565.

Article PubMed Google Scholar

Han DH, Kim SM, Bae S, Renshaw PF, Anderson JS. Brain connectivity and psychiatric comorbidity in adolescents with Internet gaming disorder. Addict Biol. 2017;22:80212.

Article PubMed Google Scholar

Shah LM, Cramer JA, Ferguson MA, Birn RM, Anderson JS. Reliability and reproducibility of individual differences in functional connectivity acquired during task and resting state. Brain Behav. 2016;6:e00456.

Article PubMed PubMed Central Google Scholar

Curtis BJ, Williams PG, Jones CR, Anderson JS. Sleep duration and resting fMRI functional connectivity: examination of short sleepers with and without perceived daytime dysfunction. Brain Behav. 2016;6:e00576.

Freed D, Aldana R, Weber J, Edwards J. The sentieon genomics tools - a fast and accurate solution to variant calling from next-generation sequence data. BioRxiv 2017 https://doi.org/10.1101/115717.

Faust GG, Hall IM. SAMBLASTER: fast duplicate marking and structural variant read extraction. Bioinformatics. 2014;30:25035.

Article CAS PubMed PubMed Central Google Scholar

Ewels P, Magnusson M, Lundin S, Kaller M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics. 2016;32:30478.

Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 2018;34:i88490.

Here is the original post:
Neurite outgrowth deficits caused by rare PLXNB1 mutation in ... - Nature.com

Researchers Created "Embryos" From Monkey Stem Cells For The … – Inverse

The early beginnings of an embryo as it transforms from a clump of cells to a tiny human is a bit of a mystery scientifically speaking. We know many of the broad strokes, but there are a lot of intricate steps involved in the transformation that we arent privy to simply because we cant see inside the uterus as everything unfolds.

Not having front-row seats hasnt stopped scientists from trying to parse together an IKEA instruction manual for embryonic development. Over the last several decades, a variety of studies conducted in animals like zebrafish, mice, and fruit flies have identified some of the genetic switches underpinning embryogenesis a fertilized eggs journey to becoming a multicellular organism. There have also been studies using human and human-animal hybrid embryos to various degrees, of which theres been terse progress due to ethical concerns constraining such research.

The goal, therefore, is to find workarounds and proxies that can mimic a human uterus or resemble human embryonic development. Theres been some effort on that front in recent years when in August 2022, scientists successfully created a synthetic mouse embryo using stem cells instead of the usual mishmash of eggs and sperm and incubated the embryo in a mechanical womb.

Now, in a study published Thursday in the journal Cell Stem Cell, researchers in China have created embryo-like structures from embryonic stem cells taken from the crab-eating macaque. This structure called a blastoid, was similar to a crucial embryonic structure called a blastocyst, and possessed the transformative ability that eventually gives rise to the different cells and tissues in the body. However, when implanted into the uteri of female macaques, the blastoids didnt survive past a week (nearly three weeks in total from creation), although they did develop gestational sacs.

I wouldnt call this a breakthrough study, Jianping Fu, professor of biomedical engineering at the University of Michigan, who wasnt involved in the study, tells Inverse. But it points to an exciting direction to bypass the existing constraints [set] by human and animal models.

Stem cells, particularly embryonic and induced pluripotent stem cells (which are derived from adult cells and rewired to resemble their embryonic counterparts), have become a hotbed of interest simply for one reason: They have the potential to change into any cell type in the body, kind of like a cellular Animorph. This means they can be used to generate a wide range of cell types for research and clinical therapies, such as targeting neurodegenerative diseases like Parkinsons to diabetes and even dental issues.

The embryonic stem cells used in this new study came from crab-eating macaques, a species of long-tailed, brown-gray Old World monkeys native to Southeast Asia. These primates are widely used in medical research due to their physiological and genetic similarities to humans (thus their classification as near-human primates), particularly in areas such as neuroscience, infectious diseases, and reproductive biology.

To get growing and transforming, embryonic stem cells regardless if theyre human or monkey need chemicals called growth factors. This jumpstarts and nudges the cell down a certain career path (think your high school counselor on career day), which prompts certain genes to turn on and off depending on the desired cell type.

For their study, the researchers across various academic research institutions in China bathed their macaque embryonic stem cells in growth factors known from past studies to be involved in embryonic development. (Its important to note, though, we dont have an expansive knowledge of all the growth factors present in an embryo.) After about a week simmering in this chemical cocktail, the embryonic stem cells started to take on the appearance of a blastocyst a hollow sphere-like structure, parts of which will eventually develop into the placenta when viewed under the microscope (hence the name blastoid).

Also, under the microscope, these researchers noticed the blastoids appeared to have reached a stage in embryonic development called gastrulation. This is when three cells, or germ, layers the ectoderm, mesoderm, and endoderm start to form, ultimately giving rise to all the different types of cells and tissues in the body. This seemed to be corroborated by single-cell RNA sequencing, a technique used to photograph gene expression with resolution down to the single cell (around 6,000 in this study). The gene expression snapshots showed that different cells within the blastoid shared a nearly similar gene expression to natural blastocysts or embryos right after implantation, when the fertilized egg attaches to the uterine wall in early pregnancy.

So if it looks like a blastoid, does it act like a blastoid? Specifically, can it become an embryo? Not exactly.

To see how the blastoids fared in a more natural habitat namely inside a surrogate mama the early embryonic structures (about two weeks old at this point) were surgically implanted into eight female macaques. Of the eight, the blastoids appeared to successfully tether in three primates. The cells seemed to trigger pregnancy, indicated by the presence of hormones progesterone and chorionic gonadotropin, both crucial for sustaining pregnancy in monkeys as well as humans. This demonstrated that the blastoids were able to mimic some of the critical functions of a developing embryo, albeit on a limited scale.

While the blastoids did form gestational sacs fluid-filled structures that serve as an early sign of pregnancy and potentially a yolk sac in one (another early embryonic structure that produces blood and germ cells) seven to 10 days after implantation, these structures didnt progress any further. Roughly twenty days after they were first created, the blastoids disappeared without a trace.

Growing these embryo-like structures outside the uterus (at least initially) is among one of many modes of exploration the researchers hope will provide us insight into the molecular mechanisms behind-the-scenes of embryonic development.

[This research] provides new tools and perspectives for the subsequent exploration of primate embryos and reproductive medical health, Qiang Sun, the studys co-author and director of Suzhou Non-human Primate Facility at the Chinese Academy of Sciences, said in a statement.

Especially for reproductive health, this research could lend to a better understanding of why early miscarriages happen, which occur in 10 percent to 20 percent of pregnancies. Theres no exact cause, but there are a variety of reasons, such as random chromosomal abnormalities or structural deficits, whether in the moms uterus or in the baby, that prevent implantation or proper embryonic development.

Such monkey models may be very useful [for] toxicity screening applications to identify chemicals that have potential toxic effects on pregnancy, says Fu of the University of Michigan. Theres also a lot of hope such animal models, especially related to primate development, might guide us to better understand early development so we can [create] better protocols which might be very useful for [in vitro fertilization].

But hes skeptical of exactly how much these findings can contribute to our understanding of early development. Previous studies fusing mouse and human embryonic stem cells show that even these hybrids can develop into blastoids, so demonstrating the same with monkey embryonic stem cells isnt altogether new. Not only that, hes not convinced the monkey blastoids actually achieved gastrulation since, judging from the data provided, they still look very much disorganized.

Fu warns that a monkey model workaround may still toe the line of whats ethically permissible since, evolution-wise, macaques are pretty close to humans. This may make their blastoids nearly equivalent to human cells. Ethical concerns about primate models for embryonic development have been raised in the past.

The researchers acknowledge the potential ethical conundrum but note that the blastoids theyve created are still very different and not functionally on par with human blastocysts.

This research still has a long way to go, and likely many more years before we can completely pick apart the black box that is embryonic development.

The rest is here:
Researchers Created "Embryos" From Monkey Stem Cells For The ... - Inverse

Softer and more gelatinous: taste testing Australias first lab-grown pork – The Guardian

Food science

Advocates of cultivated meat say its better for animal welfare and the environment, but the jury is out on whether the industry is headed for greener pastures

The liability waiver does not inspire confidence. I am not a natural thrill seeker, but in my limited experience, sitting down in an empty Melbourne cafe to eat a snack is not a typical activity that may cause serious or grievous injuries, including bodily injury, and/or death.

I am here to taste a product that is much hyped but not yet commercially available in Australia (and most other jurisdictions): meat that has been grown in a lab.

The meat in question is a soupon of pork, which has been cultivated from the cells of a pigs ear. The pig, I have been assured by Paul Bevan, the chief executive of cultivated meat startup Magic Valley, is still alive and well, continuing to live its happy, healthy, normal life.

Lab-grown meat or cultivated meat, as it is known in the industry is purported by its proponents to be better for animal welfare and the environment. It exited the realm of science fiction in 2013, when a research team at Maastricht University presented the first prototype, a lab-grown beef burger patty.

Since then, the cellular agriculture industry has yielded just a single commercially available product cell-based chicken launched in Singapore in 2020 by the American firm Eat Just. A cultivated chicken product from Eat Just subsidiary Good Meat is now making its way through US regulatory approval.

Magic Valley is hoping to apply for regulatory approval in Australia by the end of the year, and to sell their cultivated meat products lamb and pork, so far by the end of 2024.

I am a meat eater and dont consider myself particularly squeamish, but as I wait to taste Magic Valleys pork I try to suppress a mental image of muscle fibres growing in a Petri dish, la science experiments from university biology class.

The morsel of lab-grown meat is served in a silky wonton skin, doused with chilli oil, spring onion and black vinegar. The recipe has been cooked up by Wendy Chua, a Magic Valley scientist and their in-house gourmet.

The pork is, in a word, delicious. But then again, what generously seasoned dumpling isnt?

The texture of the meat is perhaps slightly softer and more gelatinous than regular mince. Any differences in taste would have been easier to parse with a more sizeable portion, but I suspect the meagre serving is a deliberate logistical (and economic) choice.

Magic Valley is not yet operating at industrial scale, so all their meat is still cultivated in a lab, rather than in 20,000-litre bioreactors that they hope to eventually use. A facility with two bioreactors of such size would eventually be able to produce 300,000kg of meat annually, Bevan says. (It is unclear whether bioreactors this large are commonly used in the industry, but in 2022 Eat Just announced it was building 250,000-litre vats, set to be operational in 2024.)

The process of making the pork begins with reprogramming cells taken from a pigs ear to create induced pluripotent stem cells. These stem cells, which are not yet specialised, have an essentially unlimited ability to generate other cell types. From there, were able to direct the cells to become muscle, fat, connective tissue, bone whatever we choose, Bevan says.

Cells are brewed in nutrient media a liquid of glucose and amino acids that enables the cells to grow. Currently, muscle and fat are grown separately and combined at the end to form the final product. The process takes about three weeks, Bevan says.

Magic Valley describes its meat as slaughter-free, but other cellular agriculture companies use foetal bovine serum a byproduct of the meatpacking industry which is harvested from the blood of cow foetuses as a growth medium.

In terms of input costs at the moment, for us it costs around $50 a kilo to produce, Bevan says. His hope is that once production is scaled up, the costs may drop to $5 per kilogram.

While the current price tag is steep, it is far cheaper than it once was the first lab-grown patty cost US$330,000 to create. But some critics are sceptical that cultivated meat can achieve cost parity with traditional agriculture.

If your interest is maximising profitability in the early years, you should never start a cultivated meat company, Eat Justs chief executive, Josh Tetrick, told the Financial Times in June last year. The article highlighted that despite Eat Justs 2020 commercial milestone in Singapore, the lossmaking companys products are not in shops.

Though lab-grown meat has high energy requirements, analyses suggest the production process involves less carbon emissions and a smaller land-use footprint per kg of meat than traditional agriculture.

From a greenhouse gas perspective, and from a water use and a land use perspective, were looking at between a 70% and 90% reduction compared to conventional meat, Bevan says.

Nutritionally, our cultivated pork products are identical, Bevan says.

Independent studies are unclear on whether cultivated meat provides the same essential minerals, such as iron and vitamin B12, as regular meat. One 2020 review found that vitamins are necessary in the [growth] media for optimal cell proliferation, but it is not clear whether the uptake from media results in levels of vitamins in cultured meat comparable to traditional meat.

But unlike a pork cutlet, which is what it is, Bevan says with cultivated meat we can remove things like saturated fat, add additional protein content, vitamins, minerals, etcetera.

Critics of alternative proteins such as lab-grown meat have suggested the industry could jeopardise the livelihoods of food producers globally, rather than supporting transformational changes in the way we eat. Nonetheless, the demand for meat is rising worldwide, and cultured meat companies have proliferated accordingly.

In March, another Australian startup, Vow, unveiled a meatball engineered from the tissue of the long-extinct woolly mammoth, which nobody has yet tasted. In addition to traditional livestock animals, the company is making meat from the stem cells of at least 13 other animal species, such as alpaca and water buffalo.

As the technology advances, Bevan says the ultimate goal for Magic Valley is to create structured meat products such as steaks and chops. Last year, one Israeli firm used 3D printing techniques to create a 110g steak, but while the technology is impressive, the end product hardly resembles the real thing.

Replacing the chicken in a nugget or the pork filling in a wonton with cultivated meat is beginning to seem like a real possibility. But whether lab-grown chops wont smack of the uncanny valleys pastures is a question thats yet to be answered.

{{topLeft}}

{{bottomLeft}}

{{topRight}}

{{bottomRight}}

{{.}}

Original post:
Softer and more gelatinous: taste testing Australias first lab-grown pork - The Guardian

Pathology and Astrocytes in Autism | NDT – Dove Medical Press

Introduction

Autism spectrum disorder (ASD) is as a neurodevelopmental disorder that presents with disturbances in social communication and repetitive behaviors.1 One in every 54 children suffers from ASD in the United States with a prevalence 4.3 times higher in males than in females.2 Global prevalence around the world is approximately of 1/100 children varying based on geographic, ethnic, and socioeconomic factors.3 The etiology of ASD is not well understood, however genetic, environmental, and immune factors have been reported to be the cause.4 Many genes linked to ASD have also been associated to other neurodevelopmental disorders, indicating a etiological heterogeneity and genetic pleiotropy in ASD.5 Candidate genes linked to ASD include DISC1, DYX1C1, RELN, AVPR1a, ITGB3, RPL10, and SHANK3, among many others.6 These genes regulate development, metabolism, plasticity, synapsis, and other important functions.7,8 Although numerous genes have been involved in ASD, a genetic diagnosis is not possible in most of the cases because the established genetic causes of ASD account for only a small portion of cases.9 Environmental factors such as hypoxia or trauma at birth, heavy metal exposure, maternal obesity, vitamin D deficiency, and maternal diabetes, have been associated with ASD.10 There is also a significant link between ASD with increase in reactive oxygen species (ROS) and a reduction in antioxidant capacity in the brain of ASD patients. ROS accumulation can directly enhance neuroinflammation and cytokine release.11 Accordingly, immune system impairment with elevated expression of pro-inflammatory cytokines and chemokines and microglia activation have been reported in postmortem ASD brains.12,13 Genetic, environmental, and immune factors are involved in the ASD phenotype, but how exactly is poorly understood.

The pathology of ASD is yet to be determined. However, the anatomy of several brain areas such as cerebellum, amygdala, hippocampus and cerebral cortex have been reported to be affected.1416 Increased brain size and disorganization of white and grey matter have been identified in patients with ASD.17,18 MRI studies showed abnormalities in gyral cortical anatomy, especially in the sylvian fissure, superior temporal sulcus,intraparietal sulcus,and inferior frontal gyrus in ASD patients.19,20 Multiregional dysplasia is present in 92% of ASD cases.21 It has been hypothesized that focal dysplasia in ASD may result from abnormalities in progenitor cell division, and/or migration and maturation of newly generated cells during prenatal brain development.21,22 Cortical dysplasia in ASD could explain high seizure prevalence and sensory disturbance in ASD.23 Mini-columnar abnormalities have also been reported in ASD.24,25 Mini-columns contain oriented arrays of pyramidal cells and GABAergic interneurons that modulate pyramidal cells input and output. Mini-columns are considered the basic functional unit in the neocortex. In ASD, there are more mini-columns but they are smaller in size. There is also less neuropil space resulting in cells more compacted.24,25 The increased number of mini-columns may result from additional division of progenitor cells during prenatal development, while the deficits in peripheral neuropil space may result from lack of inhibitory cells.26 White matter is also affected in ASD. In particular, there is a reduction in the number of long axons that are connected to long distance areas, and an increase in thin axons that communicate neighboring areas. This indicates a disconnection between long distance pathways and short distance over-connection. This is the case for the white matter in the anterior cingulate cortex, an area associated with attention, social interaction and emotion, functions altered in ASD. Moreover, there is also a reduction in axonal myelin thickness in some areas such as the white matter of the orbitofrontal cortex.27

The ASD brain also presents with alterations in the number of specific cell types. However, most of the cell types and regions of the brain have not been studied, and some of the data collected do not agree. Alteration in cerebellar cortex including a decrease in size and number of Purkinje cells and abnormality in functional connectivity between the cerebellum and other areas of the brain was reported in postmortem ASD brains. Decrease in Purkinje cells number was more noticeable in posterior lobe (lobule VIIA) of the cerebellum. Accordingly, a reduction in grey matter volume and a smaller vermis lobules VIVII were present in ASD children.28 Children with ASD had a bigger amygdala than typically developing children.29 A study on non-neuronal cell population numbers in the amygdala, reported no changes in number, however there was a strong microglial activation in two of eight ASD brains. In addition, there was a reduced number of a oligodendrocytes in the amygdala of adult ASD cases aged 20 and older.30 In the fusiform gyrus in seven postmortem ASD subjects, there was a decrease in number of neurons in layers III, V and VI, and in the mean perikaryal neuronal volumes in layers V and VI.31 An increase in the pyramidal cell population32,33 and a reduction in oligodendrocyte and astrocyte numbers (Figure 1AB) have also been reported in the prefrontal cortex of ASD postmortem brains.33,34 Also, a reduction in parvalbumin+ chandelier GABAergic interneurons was found in the dorsolateral and ventral prefrontal cortex.17,35,36 Decreased dendrite numbers in the dorsolateral prefrontal cortex and reduced dendrite branching in the CA4 and CA1 have been reported in individuals with ASD.37 Overall, abnormalities in different cell type populations and their morphology may lead to the disturbed neuronal function characteristic of ASD.

Figure 1 (A and B) GFAP+ astrocytes in prefrontal cortical plate (CP) and the white matter (WM) (A) control (CT) and (B) ASD. (A and B) Reconstruction of an average case depicting GFAP+ astrocyte location in the CP and WM. (A) control (CT) and (B). (CF) Astrocytes activation state. (C) Resting astrocyte with few processes and small cell body, (D) mild reactive astrocyte with slightly enhanced staining of glial processes and minor enlargement of cell body, (E) moderate reactive astrocyte with significant increase of cell body size and glial cell ramifications with dark stained processes and (F) severe reactive astrocyte with gemistocytic cell body and degraded processes that present as dark stained puncta.105 Scale bar in A, A, B, B: 500 m; C, D, E, F: 20 m.

Astrocytes are key elements for neuronal metabolic and structural support in the brain. They control ion concentration, modulate neurotransmitter release, maintain the bloodbrain barrier, and regulate blood flow in the nervous system, among many other functions.38 They also have crucial roles in neurodevelopment including in neurogenesis, neuronal migration, and synaptic plasticity.39,40 In addition, with pre- and postsynaptic neurons, perisynaptic astrocytes form tripartite synapses to modulate synaptic transmission.41 Together with microglia, astrocytes are regulators of the inflammatory responses. Innate immune responses are mediated through activation of microglia and astrocytes that produce cytokines, chemokines, and other immune mediators.38,42,43 Astrocyte activation could be either neurotoxic, by accelerating inflammatory responses and tissue damage, or neuroprotective by promoting neuronal survival and tissue repair, though this classification is not clear cut. Pro-inflammatory astrocytes secrete pro-inflammatory factors, such as tumor necrotic factor (TNF) and nitric oxide (NO), whereas neuroprotective astrocytes upregulate neurotrophic factors and thrombospondins to control neuroinflammation. Excessive neuroinflammation with increased reactive astrocytes and pro-inflammatory cytokines has been reported in ASD. Given the role of astrocytes in higher cognitive functions, any alteration in their number, distribution, morphology, and/or function, could lead to major neuronal dysfunction that could contribute to neurodevelopmental disorders such as ASD.44

Glial fibrillary acidic protein (GFAP) is a type III intermediate filament that is mainly expressed in astrocytes. It is also known as a marker for reactive astrocytes (Figure 1CF).42,45 GFAP is reported to be elevated in the cerebrospinal fluid of ASD subjects.46,47 Increased GFAP is correlated with astrogliosis and reactive damage that might result in immune response and further cytokines release.13,48 Data regarding GFAP gene expression in different regions of the ASD brain is controversial. Some studies reported upregulation of GFAP gene expression in the prefrontal cortex and cerebellum,49,50 whereas others reported no significant changes in GFAP gene expression in anterior cingulate cortex and anterior prefrontal cortex in ASD brains.51,52 Rats treated with propionic acid showed increased GFAP gene expression in the hippocampus, and presented ASD-like behaviors including aggressive behavior during adjacent interactions.53 At the protein level, several studies reported an increase in GFAP protein in superior frontal cortex, parietal cortex, cerebellum, and anterior cingulate cortex white matter.13,48,51 There was also an increase in GFAP protein in the cerebellum of postmortem brains whereas vimentin was decreased in both cerebellum and prefrontal cortex.54 In a valproic acid (VPA) animal model of ASD, there was an increase in the number of astrocytes and GFAP in medial prefrontal cortex and primary somatosensory cortex on postnatal day 30.55 In contrast, some other studies showed no change in GFAP protein in anterior cingulate grey matter, amygdala, and anterior and dorsolateral prefrontal white matter of postmortem ASD brains.30,51,56 Other proteins expressed by astrocytes are also changed in ASD. There was decreased amount of aquaporin 4 (AQP4), a water channel protein located in astrocytes, in the medial prefrontal cortex, but an elevation in the primary somatosensory area in the VPA animal model of ASD. AQP4 is mainly responsible for eliminating water from the cerebral parenchyma as well as supporting potassium buffering.55,55 In addition, there was a reduction in AQP4 protein in the cerebellum and an increase of connexin (cnx) 43, a gap junction protein located in astrocytes, in BA9 of postmortem ASD brains.57 Beside buffering ions and neurotransmitters concentration, cnx43 is responsible for regulating cellular growth and cell-cell adhesion. Increased cnx43 expression in ASD subjects could signify enhancement of glial-neuronal communication in frontal lobe that is in charge of executive functions.57

Data regarding the number of astrocytes in the brain with ASD are scarce (Table 1). We previously reported a decrease in the number of astrocytes, labeled with GFAP and S100, and a mild activation in GFAP+ astrocytes in the prefrontal areas BA9, BA46, and BA47 of postmortem ASD brains compared to control individuals.34 Figure 1 depicts representative images of astrocytes labeled with GFAP antibody and their location in control and ASD prefrontal cortex and astrocytes in different stages of activation. In another study from our laboratory, using Nissl staining, we showed a generalized reduction in astrocytes number with an increase in the neuronal population in layer II in the same areas.33 A reduced number of astrocytes could result from a reduced production and/or increased cell death. Increased overall glial cell densities, including astrocytes, oligodendrocytes and microglial cells, in layer II of olfactory cortex was reported, that may correlate with sensory deficits including damaged olfactory identification observed in patients with ASD. This increased glial cell density was correlated positively with the scores for restricted and repetitive behavior domain in the autism diagnostic interview revised (ADI-R) questionnaire.58 Using clustering nuclear profiles, genetic studies showed upregulated protoplasmic astrocyte gene expression in the prefrontal cortex and anterior cingulate cortex of postmortem ASD brains.59 In addition, upregulation of a gene set that was enriched in astrocytes and microglia was observed in frontal and temporal cortex of 251 postmortem samples from 48 ASD cases and 49 control subjects.60 These data contrast with anatomical studies demonstrating a decreased number of astrocytes in the prefrontal cortex. This may be because anatomical landmarks were not taking into account and the number of astrocytes was quantified using homogenated tissue.

Table 1 Summarizing Astrocyte Abnormalities in ASD Human Studies

Astrocytes play a critical role in neurotransmitter homeostasis, and in regulating the excitation/inhibition balance that is disturbed in the ASD cortex. Disturbance in astrocyte calcium signaling through inositol 1,4,5-trisphosphate 6 receptor 2 (IP3R2), that regulates neurotransmitter release, leads to ASD-like behaviors including repetitive behaviors and abnormal social interaction in mice.6163 Also, elevated level of glutamine synthetase (GS), an adenosine triphosphate-dependent enzyme that maintains glutamate levels located in astrocytes, was reported in the plasma of ASD patients.64 Increased mRNA expression of excitatory amino acid transporter 1 (EAAT1), located in astrocytes and responsible for glutamate uptake, and glutamate receptor AMPA 1, were found in the cerebellum of postmortem ASD brains. However, the density of AMPA glutamate receptor protein was decreased in the cerebellum. These findings reveal abnormalities in glutamatergic system in ASD.50 Some other studies reported a correlation between the glutamate transporter single gene polymorphism and the severity of anxiety and repetitive behaviors in ASD children.65 Furthermore, excessive electrical activity resulting from an abnormal glutamatergic function has been reported in ASD patients that can lead to pathologic behaviors.66 In VPA animal model of ASD, there was a decrease of 40% in glutamate transporter 1 (GLT1) at P15, but an increase of 92% in GLT1 with an increase of 160% in glutamate uptake at P120. The amount of glutathhione (GSH) was also increased 27% at P120 suggesting a disturbance in astrocytic glutamate clearance from the synaptic cleft in an animal model of ASD.67

Some report ASD as a hypo-glutamatergic disorder because of the symptoms produced by glutamate antagonists in ASD.68 Accordingly, a hypo-glutamatergic animal model displayed behavioral phenotypes that overlapped with the features observed in ASD69, indicating an alteration in the glutamatergic function in ASD.

Astrocytes also participate in gamma-aminobutyric acid (GABA) clearance. Some studies have shown a relationship between astrocyte abnormalities and the GABAergic system dysfunction in ASD. Wang et al showed a reduction in astrocyte-derived ATP that impaired GABAergic system and lead to ASD-like behaviors in the PFC of the IP3R2 mutant mice. ATP can modulate GABAergic synaptic transmission via P2X2 receptors located at the GABAergic interneuron terminals.63 In an in vitro study, cultured astrocytes exposed to VPA showed impairment in GABAergic inhibitory synapses but the excitatory synapses remained unchanged. This indicates that VPA can alter E/I balance in neural network by affecting the astrocyte-neuron interaction, highlighting the impact of astrocyte dysfunction in ASD pathology.70 Overall, there is evidence that astrocyte regulating of both glutamate and GABA neurotransmitters is altered in the ASD brain.

Neuroinflammation plays a main role in ASD pathology and many studies reported activation of astrocytes in postmortem ASD brains.13,71,72 Reactive astrocytes are the major source of releasing cytokines. The macrophage chemoattractant protein (MCP-1), that is in charge of monocyte/macrophage recruitment to the areas of inflammation, and pro-inflammatory cytokine interleukin-6 (IL-6), are altered in cortical and subcortical white matter in ASD.13 The expression of the translocator protein 18 kDa (TSPO), that is a marker for brain inflammation, and the amount of activated microglia in the frontal cortex and cerebellum are increased in reactive astrocytes in ASD.73 Monocyte chemoattractant protein-1 (MCP-1/CCL2) is a chemokine that has been reported to be elevated in the brain and blood of ASD cases.13,74 CCL2 is produced by astrocytes and microglia in the brain and is necessary for proliferation, migration and activation of microglia and astrocytes.75,76 Elevated level of CCL2 could also increase bloodbrain barrier (BBB) permeability and allow more T-lymphocytes to enter the brain during neuroinflammation.77 Multifocal perivascular lymphocytic cuffs are associated with astrocytes blebs that represents a cytotoxic reaction to lymphocyte attack, suggesting a dysregulation in cellular immunity that could damage astrocytes in ASD brains.78 Although many studies reported immune system dysfunction in ASD, it is not clear whether it is a cause or a consequence of the pathology.72

Astrocytes perform a critical role in synaptic formation, maturation, function, and elimination. An alteration in astrocyte structure and function alters neuronal activity.79 Astrocytes secrete platelet responsive protein (TSP) that works through its neuronal receptor calcium channel subunit 2-1, to control excitatory synaptogenesis.80 The synaptic signaling protein Rho GTPase Ras-related C3 Botulinum toxin substrate 1 (RAC1), is downstream of the TSP-2-1 pathway and has an important role in regulating synaptic and spinal growth.81 Disturbed RAC1 signaling is strongly associated with ASD and epilepsy pathology.82,83 The fact that astrocytes control the TSP-2-1-RAC1 pathway, is an example of the role of astrocytes on synaptic formation in ASD.84 Astrocytes secrete cytokines, such as transforming growth factor 1 (TGF-1) to regulate synaptogenesis. TGF-1 enhances phosphorylation of calcium/calmodulin dependent protein kinase II (CaMK II), downstream of NMDA receptors, to induce the formation of inhibitory synapses.85 TGF-1, with the NMDA coactivator D-serine, encourages the formation of excitatory synapses through NMDA receptor-dependent mechanisms.86 Supporting a role of TGF-1 in the formation of inhibitory synapses suggest that a relationship between the TGF-1 dysfunction and inhibitory synapse disturbance in ASD.87 Hevin is another protein secreted by astrocytes that is essential for maintaining synaptogenesis. Hevin bridges the presynaptic protein Neurexin-1 (NRX1) and postsynaptic Neuroligin-1B (NL1B) to assemble excitatory synapses.88 Mutations in Hevin, Neurexins and Neuroligins are strongly related to ASD pathology suggesting a critical role of these proteins in normal brain development.89

Astrocyte abnormalities have also been reported in other neurodevelopmental disorders, such as schizophrenia (SZ), bipolar disorder (BD) and major depressive disorder (MDD). A reduction in astrocyte densities was present in some brain areas of postmortem brains with SZ including cingulate and motor cortex, medial and ventrolateral regions of the nucleus accumbens, basal nuclei and substantia nigra.90 In an electron microscopic morphometric study of astrocytes in hippocampal CA3 region of 19 SZ cases, mitochondrial volume fraction and area density was negatively correlated with the duration of disease. However, the volume fraction of lipofuscin granules was positively associated with the duration of illness suggesting progressive astrocyte dysfunction due to the mitochondrial deficit.91 An increased expression of GFAP mRNA with astrogliosis was also observed in SZ patients with neuroinflammation.92 Furthermore, in animal studies of SZ, transgenic mice that expressed a mutant form of the disrupted in schizophrenia 1 (DISC1) gene in astrocytes, showed behavioral abnormalities related to SZ supporting the role of astrocytes in SZ pathology.93,94

In BD, astrocytic density was also reduced supporting astrocyte dysfunction in regulating glutamate homeostasis, calcium signaling, circadian rhythms and metabolism. Beneficial therapeutical effects of many BD drugs such as lithium, valproic acid (VPA) and carbamazepine (CBZ) are partly due to their positive actions on astrocytes by affecting the gene expression in astrocytes and regulating astroglia homeostatic pathways.95,96 There is also an elevation reported in the expression profile of cortical astrocytes in the postmortem BD subjects generated from eight different cohorts of subjects.97 In an in vitro study, astrocytes derived from induced pluripotent stem cells (iPSCs) generated from BD individuals showed alteration in transcriptome and a decrease in neuronal activity when they were co-cultured with neuronal cells. BD astrocytes also increased IL-6 secretion in the blood of BD patients highlighting the role of astrocytes in inflammatory signaling in BD pathology.98

A reduction in the astrocyte density in various regions of the brain including the prefrontal cortex, cingulate cortex and amygdala is an important feature in MDD pathology.99101 Golgi staining showed astrocytic hypertrophy in cell bodies and processes in the white matter of cingulate cortex of depressed patients that died by suicide. The presence of hypertrophic astrocytes could reflect local inflammation supporting the neuro-inflammatory hypothesis in depressed patients.102 In addition, the protein and mRNA level of pro-inflammatory cytokines secreted by reactive astrocytes were increased in the prefrontal cortex of suicide victims.103 However, other astrocytic proteins and markers such as GFAP, AQP4, cnx43, cnx30, glutamate transporters, and glutamine synthetase were reduced in MDD.104

Astrocytes play an important role in neurodevelopment and neuronal function in the brain, including higher cognitive functions. Available data indicates that astrocyte number is decreased in the cerebral cortex, while their state of activation and GFAP expression is increased in the ASD brain. This dysfunction and other astrocytic alterations may contribute to the ASD pathology. More research is needed to help our understanding of the mechanisms involved in astrocytic-related pathophysiology in ASD, and to introduce astrocytes as one of the promising targets for ASD treatment. Future research should answer questions as if the decreased in astrocyte number found in cortex occurs in other brain areas, if there are areas where astrocytic activation is more pronounced that others, what is the role of astrocytes on development, plasticity, and inflammation, and what other astrocytic functions are altered in ASD.

The authors report no conflicts of interest in this work.

1. Baio J. Prevalence of autism spectrum disorders: autism and developmental disabilities monitoring network, 14 sites, United States, 2008. Morbid Mortal Wkly Rep. 2012;61(3):157.

2. Maenner MJ, Shaw KA, Baio J, et al. Prevalence of autism spectrum disorder among children aged 8 years autism and developmental disabilities monitoring network, 11 sites, United States, 2016. MMWR Surveill Summ. 2020;69:112. doi:10.15585/mmwr.ss6904a1

3. Zeidan J, Fombonne E, Scorah J, et al. Global prevalence of autism: a systematic review update. Autism Res. 2022;15:778790. doi:10.1002/aur.2696

4. Mandy W, Lai M-C. Annual research review: the role of the environment in the developmental psychopathology of autism spectrum condition. J Child Psychol Psychiatry. 2016;57:271292. doi:10.1111/jcpp.12501

5. Vorstman JAS, Parr JR, Moreno-De-Luca D, Anney RJL, Nurnberger JI, Hallmayer JF. Autism genetics: opportunities and challenges for clinical translation. Nat Rev Genet. 2017;18:362376. doi:10.1038/nrg.2017.4

6. Cardoso IL, Almeida S. Genes involved in the development of autism. Int Arch Commun Disord. 2019;2:19. doi:10.23937/iacod-2017/1710011

7. Ansel A, Rosenzweig JP, Zisman PD, Melamed M, Gesundheit B. Variation in gene expression in autism spectrum disorders: an extensive review of transcriptomic studies. Front Neurosci. 2017;10. doi:10.3389/fnins.2016.00601

8. Chaste P, Leboyer M. Autism risk factors: genes, environment, and gene-environment interactions. Dialogues Clin Neurosci. 2012;14:281292. doi:10.31887/DCNS.2012.14.3/pchaste

9. Voineagu I. Gene expression studies in autism: moving from the genome to the transcriptome and beyond. Neurobiol Dis Assess Gene Expres Neuropsychiatr Dis. 2012;45:6975. doi:10.1016/j.nbd.2011.07.017

10. Modabbernia A, Velthorst E, Reichenberg A. Environmental risk factors for autism: an evidence-based review of systematic reviews and meta-analyses. Mol Autism. 2017;8:13. doi:10.1186/s13229-017-0121-4

11. Pangrazzi L, Balasco L, Bozzi Y. Oxidative stress and immune system dysfunction in autism spectrum disorders. Int J Mol Sci. 2020;21:3293. doi:10.3390/ijms21093293

12. Morgan JT, Chana G, Pardo CA, et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry. 2010;68:368376. doi:10.1016/j.biopsych.2010.05.024

13. Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2005;57:6781. doi:10.1002/ana.20315

14. Bauman ML, Kemper TL. Neuroanatomic observations of the brain in autism: a review and future directions. Int J Dev Neurosci. 2005;23:183187. doi:10.1016/j.ijdevneu.2004.09.006

15. Garbett K, Ebert PJ, Mitchell A, et al. Immune transcriptome alterations in the temporal cortex of subjects with autism. Neurobiol Dis. 2008;30:303311. doi:10.1016/j.nbd.2008.01.012

16. Teffer K, Semendeferi K. Chapter 9 - human prefrontal cortex: evolution, development, and pathology. In: Hofman MA, Falk D, editors. Progress in Brain Research, Evolution of the Primate Brain. Elsevier; 2012:191218. doi:10.1016/B978-0-444-53860-4.00009-X

17. Hashemi E, Ariza J, Rogers H, Noctor SC, Martnez-Cerdeo V. The number of parvalbumin-expressing interneurons is decreased in the prefrontal cortex in autism. Cereb Cortex. 2017;27:19311943. doi:10.1093/cercor/bhw021

18. Scuderi C, Verkhratsky A. Chapter eleven - the role of neuroglia in autism spectrum disorders. In: Ilieva M, Lau -WK-W, editors. Progress in Molecular Biology and Translational Science, Autism. Academic Press; 2020:301330. doi:10.1016/bs.pmbts.2020.04.011

19. Levitt JG, Blanton RE, Smalley S, et al. Cortical sulcal maps in autism. Cereb Cortex. 2003;13:728735. doi:10.1093/cercor/13.7.728

20. Nordahl CW, Dierker D, Mostafavi I, et al. Cortical folding abnormalities in autism revealed by surface-based morphometry. J Neurosci. 2007;27:1172511735. doi:10.1523/JNEUROSCI.0777-07.2007

21. Wegiel J, Kuchna I, Nowicki K, et al. The neuropathology of autism: defects of neurogenesis and neuronal migration, and dysplastic changes. Acta Neuropathol. 2010;119:755770. doi:10.1007/s00401-010-0655-4

22. Casanova MF. Autism as a sequence: from heterochronic germinal cell divisions to abnormalities of cell migration and cortical dysplasias. Med Hypotheses. 2014;83:3238. doi:10.1016/j.mehy.2014.04.014

23. Casanova MF, El-Baz AS, Kamat SS, et al. Focal cortical dysplasias in autism spectrum disorders. Acta Neuropathol Commun. 2013;1:67. doi:10.1186/2051-5960-1-67

24. Casanova MF, Buxhoeveden DP, Switala AE, Roy E. Minicolumnar pathology in autism. Neurology. 2002;58:428432. doi:10.1212/WNL.58.3.428

25. Casanova MF, van Kooten IAJ, Switala AE, et al. Minicolumnar abnormalities in autism. Acta Neuropathol. 2006;112:287303. doi:10.1007/s00401-006-0085-5

26. Casanova MF. The Minicolumnopathy of Autism. In: Casanova MF, Opris I, editors. Recent Advances on the Modular Organization of the Cortex. Netherlands, Dordrecht: Springer; 2015:225237. doi:10.1007/978-94-017-9900-3_13

27. Zikopoulos B, Barbas H. Changes in prefrontal axons may disrupt the network in autism. J Neurosci. 2010;30:1459514609. doi:10.1523/JNEUROSCI.2257-10.2010

28. Palermo S, Morese R. Behavioral Neuroscience. BoD Books on Demand; 2019.

29. Schumann CM, Hamstra J, Goodlin-Jones BL, et al. The amygdala is enlarged in children but not adolescents with autism; the hippocampus is enlarged at all ages. J Neurosci. 2004;24:63926401. doi:10.1523/JNEUROSCI.1297-04.2004

30. Morgan JT, Barger N, Amaral DG, Schumann CM. Stereological study of amygdala glial populations in adolescents and adults with autism spectrum disorder. PLoS One. 2014;9. doi:10.1371/journal.pone.0110356

31. van Kooten IAJ, Palmen SJ, von Cappeln P, et al. Neurons in the fusiform gyrus are fewer and smaller in autism. Brain. 2008;131:987999. doi:10.1093/brain/awn033

32. Courchesne E, Mouton PR, Calhoun ME, et al. Neuron number and size in prefrontal cortex of children with autism. JAMA. 2011;306:20012010. doi:10.1001/jama.2011.1638

33. Falcone C, Mevises N-Y, Hong T, et al. Neuronal and glial cell number is altered in a cortical layer-specific manner in autism. Autism Int J Res Pract. 2021;13623613211014408. doi:10.1177/13623613211014408

34. Vakilzadeh G, Falcone C, Dufour B, Hong T, Noctor SC, Martnez-Cerdeo V. Decreased number and increased activation state of astrocytes in gray and white matter of the prefrontal cortex in autism. Cereb Cortex. 2022;32:49024912. doi:10.1093/cercor/bhab523

35. Amina S, Falcone C, Hong T, et al. Chandelier cartridge density is reduced in the prefrontal cortex in autism. Cereb Cortex. 2021;31:29442951. doi:10.1093/cercor/bhaa402

36. Ariza J, Rogers H, Hashemi E, Noctor SC, Martnez-Cerdeo V. The number of chandelier and basket cells are differentially decreased in prefrontal cortex in autism. Cereb Cortex. 2018;28:411420. doi:10.1093/cercor/bhw349

37. Mukaetova-Ladinska EB, Arnold H, Jaros E, Perry R, Perry E. Depletion of MAP2 expression and laminar cytoarchitectonic changes in dorsolateral prefrontal cortex in adult autistic individuals. Neuropathol Appl Neurobiol. 2004;30:615623. doi:10.1111/j.1365-2990.2004.00574.x

38. Kwon HS, Koh S-H. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl Neurodegener. 2020;9:42. doi:10.1186/s40035-020-00221-2

39. Farhy-Tselnicker I, Allen NJ. Astrocytes, neurons, synapses: a tripartite view on cortical circuit development. Neural Dev. 2018;13:7. doi:10.1186/s13064-018-0104-y

40. Wegiel J, Brown WT, Fauci GL, et al. The role of reduced expression of fragile X mental retardation protein in neurons and increased expression in astrocytes in idiopathic and syndromic autism (duplications 15q11.2-q13). Autism Res. 2018;11:13161331. doi:10.1002/aur.2003

41. Perea G, Navarrete M, Araque A. Tripartite synapses: astrocytes process and control synaptic information. Trends Neurosci. 2009;32:421431. doi:10.1016/j.tins.2009.05.001

42. Colombo E, Farina C. Astrocytes: key regulators of neuroinflammation. Trends Immunol. 2016;37:608620. doi:10.1016/j.it.2016.06.006

43. Pardo-Villamizar CA. Can neuroinflammation influence the development of autism spectrum disorders? In: Zimmerman AW, editor. Autism: Current Theories and Evidence, Current Clinical Neurology. Totowa, NJ: Humana Press; 2008:329346. doi:10.1007/978-1-60327-489-0_15

44. Jacobs S, Nathwani M, Doering LC. Fragile X astrocytes induce developmental delays in dendrite maturation and synaptic protein expression. BMC Neurosci. 2010;11:132. doi:10.1186/1471-2202-11-132

45. Liedtke W, Edelmann W, Bieri PL, et al. GFAP is necessary for the integrity of CNS white matter architecture and long-term maintenance of myelination. Neuron. 1996;17:607615. doi:10.1016/S0896-6273(00)80194-4

46. Ahlsn G, Rosengren L, Belfrage M, et al. Glial fibrillary acidic protein in the cerebrospinal fluid of children with autism and other neuropsychiatric disorders. Biol Psychiatry. 1993;33:734743. doi:10.1016/0006-3223(93)90124-V

47. Rosengren LE, Wikkels C, Hagberg L. A sensitive ELISA for glial fibrillary acidic protein: application in CSF of adults. J Neurosci Methods. 1994;51:197204. doi:10.1016/0165-0270(94)90011-6

48. Laurence JA, Fatemi SH. Glial fibrillary acidic protein is elevated in superior frontal, parietal and cerebellar cortices of autistic subjects. Cerebellum. 2005;4:206210. doi:10.1080/14734220500208846

49. Edmonson C, Ziats MN, Rennert OM. Altered glial marker expression in autistic post-mortem prefrontal cortex and cerebellum. Mol Autism. 2014;5:3. doi:10.1186/2040-2392-5-3

50. Purcell AE, Jeon OH, Zimmerman AW, Blue ME, Pevsner J. Postmortem brain abnormalities of the glutamate neurotransmitter system in autism. Neurology. 2001;57:16181628. doi:10.1212/wnl.57.9.1618

51. Crawford JD, Chandley MJ, Szebeni K, Szebeni A, Waters B, Ordway GA. Elevated GFAP protein in anterior cingulate cortical white matter in males with autism spectrum disorder. Autism Res. 2015;8:649657. doi:10.1002/aur.1480

52. Sciara AN, Beasley B, Crawford JD, et al. Neuroinflammatory gene expression alterations in anterior cingulate cortical white and gray matter of males with autism spectrum disorder. Autism Res. 2020;13:870884. doi:10.1002/aur.2284

53. Choi J, Lee S, Won J, et al. Pathophysiological and neurobehavioral characteristics of a propionic acid-mediated autism-like rat model. PLoS One. 2018;13:e0192925. doi:10.1371/journal.pone.0192925

54. Broek JA, Guest PC, Rahmoune H, Bahn S. Proteomic analysis of post mortem brain tissue from autism patients: evidence for opposite changes in prefrontal cortex and cerebellum in synaptic connectivity-related proteins. Mol Autism. 2014;5:41. doi:10.1186/2040-2392-5-41

55. Deckmann I, Santos-Terra J, Fontes-Dutra M, et al. Resveratrol prevents brain edema, bloodbrain barrier permeability, and altered aquaporin profile in autism animal model. Int J Dev Neurosci. 2021;81:579604. doi:10.1002/jdn.10137

56. Lee TT, Skafidas E, Dottori M, et al. No preliminary evidence of differences in astrocyte density within the white matter of the dorsolateral prefrontal cortex in autism. Mol Autism. 2017;8:64. doi:10.1186/s13229-017-0181-5

57. Fatemi SH, Folsom TD, Reutiman TJ, Lee S. Expression of astrocytic markers aquaporin 4 and connexin 43 is altered in brains of subjects with autism. Synapse. 2008;62:501507. doi:10.1002/syn.20519

58. Menassa DA, Sloan C, Chance SA. Primary olfactory cortex in autism and epilepsy: increased glial cells in autism. Brain Pathol. 2017;27:437448. doi:10.1111/bpa.12415

59. Velmeshev D, Schirmer L, Jung D, et al. Single-cell genomics identifies cell typespecific molecular changes in autism. Science. 2019;364:685689. doi:10.1126/science.aav8130

60. Parikshak NN, Swarup V, Belgard TG, et al. Genome-wide changes in lncRNA, splicing, and regional gene expression patterns in autism. Nature. 2016;540:423427. doi:10.1038/nature20612

61. Bezzi P, Volterra A. A neuronglia signalling network in the active brain. Curr Opin Neurobiol. 2001;11:387394. doi:10.1016/S0959-4388(00)00223-3

62. Petrelli F, Pucci L, Bezzi P. Astrocytes and microglia and their potential link with autism spectrum disorders. Front Cell Neurosci. 2016;10. doi:10.3389/fncel.2016.00021

63. Wang Q, Kong Y, Wu D-Y, et al. Impaired calcium signaling in astrocytes modulates autism spectrum disorder-like behaviors in mice. Nat Commun. 2021;12:3321. doi:10.1038/s41467-021-23843-0

64. Hamed NO, AlAyadhi L, Osman MA, et al. Understanding the roles of glutamine synthetase, glutaminase, and glutamate decarboxylase autoantibodies in imbalanced excitatory/inhibitory neurotransmission as etiological mechanisms of autism. Psychiatry Clin Neurosci. 2018;72:362373. doi:10.1111/pcn.12639

65. Gadow K, Roohi J, DeVincent C, Kirsch S, Hatchwell E. Brief report: glutamate transporter gene (SLC1A1) single nucleotide polymorphism (rs301430) and repetitive behaviors and anxiety in children with autism spectrum disorder. J Autism Dev Disord. 2010;40:11391145. doi:10.1007/s10803-010-0961-7

66. Choudhury PR, Lahiri S, Rajamma U. Glutamate mediated signaling in the pathophysiology of autism spectrum disorders. Pharmacol Biochem Behav Glutamate Recept. 2012;100:841849. doi:10.1016/j.pbb.2011.06.023

67. Bristot Silvestrin R, Bambini-Junior V, Galland F, et al. Animal model of autism induced by prenatal exposure to valproate: altered glutamate metabolism in the hippocampus. Brain Res. 2013;1495:5260. doi:10.1016/j.brainres.2012.11.048

68. Carlsson ML. Hypothesis: is infantile autism a hypoglutamatergic disorder? Relevance of glutamate serotonin interactions for pharmacotherapy. J Neural Transm. 1998;105:525535. doi:10.1007/s007020050076

69. Nilsson M, Carlsson A, Markinhuhta KR, et al. The dopaminergic stabiliser ACR16 counteracts the behavioural primitivization induced by the NMDA receptor antagonist MK-801 in mice: implications for cognition. Prog Neuropsychopharmacol Biol Psychiatry. 2004;28:677685. doi:10.1016/j.pnpbp.2004.05.004

70. Takeda K, Watanabe T, Oyabu K, et al. Valproic acid-exposed astrocytes impair inhibitory synapse formation and function. Sci Rep. 2021;11:23. doi:10.1038/s41598-020-79520-7

71. Liao X, Liu Y, Fu X, Li Y. Postmortem studies of neuroinflammation in autism spectrum disorder: a systematic review. Mol Neurobiol. 2020;57:34243438. doi:10.1007/s12035-020-01976-5

72. Matta SM, Hill-Yardin EL, Crack PJ. The influence of neuroinflammation in autism spectrum disorder. Brain Behav Immun. 2019;79:7590. doi:10.1016/j.bbi.2019.04.037

73. Fiorentino M, Sapone A, Senger S, et al. Bloodbrain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol Autism. 2016;7:49. doi:10.1186/s13229-016-0110-z

74. Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah IN, Van de Water J. Associations of impaired behaviors with elevated plasma chemokines in autism spectrum disorders. J Neuroimmunol. 2011;232:196199. doi:10.1016/j.jneuroim.2010.10.025

75. He M, Dong H, Huang Y, et al. Astrocyte-derived CCL2 is associated with M1 activation and recruitment of cultured microglial cells. Cell Physiol Biochem. 2016;38:859870. doi:10.1159/000443040

76. Hinojosa AE, Garcia-Bueno B, Leza JC, Madrigal JL. CCL2/MCP-1 modulation of microglial activation and proliferation. J Neuroinflammation. 2011;8:77. doi:10.1186/1742-2094-8-77

77. Song L, Pachter JS. Monocyte chemoattractant protein-1 alters expression of tight junction-associated proteins in brain microvascular endothelial cells. Microvasc Res. 2004;67:7889. doi:10.1016/j.mvr.2003.07.001

78. DiStasio MM, Nagakura I, Nadler MJ, Anderson MP. T lymphocytes and cytotoxic astrocyte blebs correlate across autism brains. Ann Neurol. 2019;86:885898. doi:10.1002/ana.25610

79. Clarke LE, Barres BA. Emerging roles of astrocytes in neural circuit development. Nat Rev Neurosci. 2013;14:311321. doi:10.1038/nrn3484

80. Eroglu , Allen NJ, Susman MW, et al. Gabapentin receptor 2-1 is a neuronal thrombospondin receptor responsible for excitatory CNS synaptogenesis. Cell. 2009;139:380392. doi:10.1016/j.cell.2009.09.025

81. Risher WC, Kim N, Koh S, et al. Thrombospondin receptor 2-1 promotes synaptogenesis and spinogenesis via postsynaptic Rac1. J Cell Biol. 2018;217:37473765. doi:10.1083/jcb.201802057

Go here to see the original:
Pathology and Astrocytes in Autism | NDT - Dove Medical Press

SaaS Spend Management Software Market 2023: Exclusive Insights … – Digital Journal

PRESS RELEASE

Published April 11, 2023

SaaS Spend Management Software Market Research Report 2023 | Pages | presents granular analysis on current and future market growth status with industry revenue and CAGR status across all regions with Top Key Players analysis - Flexera, Cledara, Blissfully, Intello, Binadox, G2 Track, License DashboarD

"Final Report will add the analysis of the impact of COVID-19 on this industry."

The SaaS Spend Management Software Market report 2023 offers a comprehensive and precise analysis of the various aspects related to business growth opportunities, challenges, risk factors, and trends across all geographic regions. The report provides up-to-date information on the latest technological advancements, SWOT and PESTLE analysis, and insightful market size information.

In addition, the SaaS Spend Management Software market report covers an in-depth analysis of growth factors, global trending technologies, and key players profiling with company profiles, and supply-demand scope. The report also gives a holistic overview of the industry revenue, demand status, competitive landscape, and regional segments of the global industry. This report is an indispensable value addition to any company looking to develop its future strategies and plan its path forward.

Get a sample PDF of the report at - https://www.marketresearchguru.com/enquiry/request-sample/19910636

The SaaS Spend Management Software market has witnessed growth from USD million to USD million from 2017 to 2022. With the CAGR of %, this market is estimated to reach USD million in 2029.

Key Players covered in the global SaaS Spend Management Software Market are:

The report covers the competitive landscape of various major players, their current market positions, and key business strategies adopted by players. This SaaS Spend Management Software market report includes information about the product launch, expansion of the production facilities or plants, adoption of new technologies, latest merger and acquisition, partnership, and collaboration of the key players. It furthers provides concrete information about the existing market scope for the new entrants and the current competitive levels and scenario for the emerging players in the global market.

Get a sample PDF of the SaaS Spend Management Software Market Report

Most important types of SaaS Spend Management Software products covered in this report are:

Most widely used downstream fields of SaaS Spend Management Software market covered in this report are:

The report combines extensive quantitative analysis and exhaustive qualitative analysis, ranges from a macro overview of the total market size, industry chain, and market dynamics to micro details of segment markets by type, application and region, and, as a result, provides a holistic view of, as well as a deep insight into the SaaS Spend Management Software market covering all its essential aspects.

For the competitive landscape, the report also introduces players in the industry from the perspective of the market share, concentration ratio, etc., and describes the leading companies in detail, with which the readers can get a better idea of their competitors and acquire an in-depth understanding of the competitive situation. Further, mergers and acquisitions, emerging market trends, the impact of COVID-19, and regional conflicts will all be considered.

In a nutshell, this report is a must-read for industry players, investors, researchers, consultants, business strategists, and all those who have any kind of stake or are planning to foray into the market in any manner.

Inquire or Share Your Questions If Any Before the Purchasing This Report - https://www.marketresearchguru.com/enquiry/pre-order-enquiry/19910636

Following Chapter Covered in the SaaS Spend Management Software Market Research:

Chapter 1 mainly defines the market scope and introduces the macro overview of the industry, with an executive summary of different market segments ((by type, application, region, etc.), including the definition, market size, and trend of each market segment.

Chapter 2 provides a qualitative analysis of the current status and future trends of the market. Industry Entry Barriers, market drivers, market challenges, emerging markets, consumer preference analysis, together with the impact of the COVID-19 outbreak will all be thoroughly explained.

Chapter 3 analyzes the current competitive situation of the market by providing data regarding the players, including their sales volume and revenue with corresponding market shares, price and gross margin. In addition, information about market concentration ratio, mergers, acquisitions, and expansion plans will also be covered.

Chapter 4 focuses on the regional market, presenting detailed data (i.e., sales volume, revenue, price, gross margin) of the most representative regions and countries in the world.

Chapter 5 provides the analysis of various market segments according to product types, covering sales volume, revenue along with market share and growth rate, plus the price analysis of each type.

Chapter 6 shows the breakdown data of different applications, including the consumption and revenue with market share and growth rate, with the aim of helping the readers to take a close-up look at the downstream market.Chapter 7 provides a combination of quantitative and qualitative analyses of the market size and development trends in the next five years. The forecast information of the whole, as well as the breakdown market, offers the readers a chance to look into the future of the industry.

Chapter 8 is the analysis of the whole market industrial chain, covering key raw materials suppliers and price analysis, manufacturing cost structure analysis, alternative product analysis, also providing information on major distributors, downstream buyers, and the impact of COVID-19 pandemic.

Chapter 9 shares a list of the key players in the market, together with their basic information, product profiles, market performance (i.e., sales volume, price, revenue, gross margin), recent development, SWOT analysis, etc.

Chapter 10 is the conclusion of the report which helps the readers to sum up the main findings and points.

Chapter 11 introduces the market research methods and data sources.

Geographically, the report includes the research on production, consumption, revenue, market share and growth rate, and forecast (2018 -2029) of the following regions:

To Understand How Covid-19 Impact Is Covered in This Report - https://marketresearchguru.com/enquiry/request-covid19/19910636

The report delivers a comprehensive study of all the segments and shares information regarding the leading regions in the market. This report also states import/export consumption, supply and demand Figures, cost, industry share, policy, price, revenue, and gross margins.

Key Offerings of SaaS Spend Management Software Market:

Trend and forecast analysis: Market trends, forecast, and Analysis to 2023 by segments and regions

Segmentation analysis: Market size by various applications such as product, material, shape, and end use in terms of value and volume shipment.

Regional analysis: Global SaaS Spend Management Software market breakdown by North and South America, Europe, Asia Pacific, Middle East and the Rest of the World.

Growth opportunities: Analysis of growth opportunities in different applications and regions in the Global SaaS Spend Management Software Market

Strategic analysis: This includes new product development and competitive landscape in the Global SaaS Spend Management Software Market

Reasons to purchase the SaaS Spend Management Software market report:

Purchase this Report (Price 2980 USD for a Single-User License) -https://marketresearchguru.com/purchase/19910636

Detailed TOC of SaaS Spend Management Software Market Forecast Report 2023-2029:

1 SaaS Spend Management Software Market Overview

1.1 Product Overview and Scope of SaaS Spend Management Software Market

1.2 SaaS Spend Management Software Market Segment by Type

1.2.1 Global SaaS Spend Management Software Market Sales Volume and CAGR (%) Comparison by Type

1.3 Global SaaS Spend Management Software Market Segment by Application

1.3.1 SaaS Spend Management Software Market Consumption (Sales Volume) Comparison by Application

1.4 Global SaaS Spend Management Software Market, Region Wise

1.5 Global Market Size of SaaS Spend Management Software

1.5.1 Global SaaS Spend Management Software Market Revenue Status and Outlook

1.5.2 Global SaaS Spend Management Software Market Sales Volume Status and Outlook

1.6 Global Macroeconomic Analysis

1.7 The impact of the Russia-Ukraine war on the SaaS Spend Management Software Market

2 Industry Outlook

2.1 SaaS Spend Management Software Industry Technology Status and Trends

2.2 Industry Entry Barriers

2.2.1 Analysis of Financial Barriers

2.2.2 Analysis of Technical Barriers

2.2.3 Analysis of Talent Barriers

2.2.4 Analysis of Brand Barrier

2.3 SaaS Spend Management Software Market Drivers Analysis

2.4 SaaS Spend Management Software Market Challenges Analysis

2.5 Emerging Market Trends

2.6 Consumer Preference Analysis

2.7 SaaS Spend Management Software Industry Development Trends under COVID-19 Outbreak

2.7.1 Global COVID-19 Status Overview

2.7.2 Influence of COVID-19 Outbreak on SaaS Spend Management Software Industry Development

3 Global SaaS Spend Management Software Market Landscape by Player

3.1 Global SaaS Spend Management Software Sales Volume and Share by Player

3.2 Global SaaS Spend Management Software Revenue and Market Share by Player

3.3 Global SaaS Spend Management Software Average Price by Player

3.4 Global SaaS Spend Management Software Gross Margin by Player

3.5 SaaS Spend Management Software Market Competitive Situation and Trends

3.5.1 SaaS Spend Management Software Market Concentration Rate

3.5.2 SaaS Spend Management Software Market Share of Top 3 and Top 6 Players

3.5.3 Mergers and Acquisitions, Expansion

4 Global SaaS Spend Management Software Sales Volume and Revenue Region Wise

4.1 Global SaaS Spend Management Software Sales Volume and Market Share, Region Wise

4.2 Global SaaS Spend Management Software Revenue and Market Share, Region Wise

4.3 Global SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.4 United States SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.4.1 United States SaaS Spend Management Software Market Under COVID-19

4.5 Europe SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.5.1 Europe SaaS Spend Management Software Market Under COVID-19

4.6 China SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.6.1 China SaaS Spend Management Software Market Under COVID-19

4.7 Japan SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.7.1 Japan SaaS Spend Management Software Market Under COVID-19

4.8 India SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.8.1 India SaaS Spend Management Software Market Under COVID-19

4.9 Southeast Asia SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.9.1 Southeast Asia SaaS Spend Management Software Market Under COVID-19

4.10 Latin America SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.10.1 Latin America SaaS Spend Management Software Market Under COVID-19

4.11 Middle East and Africa SaaS Spend Management Software Sales Volume, Revenue, Price and Gross Margin

4.11.1 Middle East and Africa SaaS Spend Management Software Market Under COVID-19

5 Global SaaS Spend Management Software Sales Volume, Revenue, Price Trend by Type

5.1 Global SaaS Spend Management Software Sales Volume and Market Share by Type

5.2 Global SaaS Spend Management Software Revenue and Market Share by Type

5.3 Global SaaS Spend Management Software Price by Type

5.4 Global SaaS Spend Management Software Sales Volume, Revenue and Growth Rate by Type

Here is the original post:
SaaS Spend Management Software Market 2023: Exclusive Insights ... - Digital Journal

Tenaya Therapeutics Has Been An Early Stage Company For Too … – Seeking Alpha

BorisRabtsevich/iStock via Getty Images

Tenaya Therapeutics (NASDAQ:TNYA) is an early stage developer of heart disease therapies and a relatively new IPO that I covered briefly 2 years ago. They have a set of interesting platforms - cellular regeneration, gene therapy and precision medicine - that they are using to develop a number of molecules targeting various heart diseases. The pipeline is at an early stage, with just one molecule in the clinic, but heart disease companies are rare, and the company looks like it is doing interesting science. So we will take a look.

Tenaya was established in 2016 with IP from Gladstone Institute and UT Southwestern. The company was able to raise $50mn in a Series A financing that year. In 2019, they raised another $90mn in a series B. 2 years later, they were able to raise another $106mn in a series C financing after they published preclinical data from two programs. That same year, they launched their IPO.

The company, like I mentioned, has three platforms. The Cellular Regeneration platform delivers genes to cardiac cells using viral vectors to regenerate them. Diseases like myocardial infarction, chemotherapy-related toxicity, and viral infection which result in loss of cardiomyocytes can be targeted through this platform. The Gene Therapy platform uses AAV vectors to deliver genes to correct functional defects in heart cells. These defects could be congenital or non-genetic forms. The precision medicine platform uses "human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as proprietary disease models and analysis of human genetics for the identification of new targets, validation of known targets, and high-throughput screening for drug discovery."

The company is very early stage and their pipeline looks thus:

TNYA Pipeline (TNYA website)

At the time of this Corporate Presentation, they had 2 INDs approved and a third in the works. The latest stage product candidate seems to be small molecule HDAC6 inhibitor TN-301 for HFpEF or Heart Failure with preserved Ejection Fraction. This is in a phase 1 trial, however there is no listing in the registry. TN-201 also has a phase 1 trial - ongoing? - however, again there's no listing yet. TN-201 is a gene therapy targeting mutations of the MYBPC3 gene in hypertrophic cardiomyopathy (HCM). The third IND-enabled asset is TN-401, another gene therapy targeting PKP2 gene in Arrhythmogenic right ventricular cardiomyopathy.

These gene therapy assets use the AAV9 vector. AAVs have been in use for over 2 decades, and 6 gene therapies using AAVs have been FDA-approved. More than 5500 patients have been treated across 40 countries. In hundreds of trials, they have demonstrated their safety, and their long lasting transgene expression. Other important positives for AAV vectors are their low immunogenicity and ability to penetrate both dividing and nondividing cells, and so on. Some disadvantages include inability to deliver larger molecules, expensive manufacturing and so on.

As to the various diseases, MYBPC3 HCM has some 115k US patients. This genetic mutation is the most common form of inherited cardiomyopathy. There are no treatments for the underlying genetic mutation although the disease can lead to higher risks of sudden heart failures. Tenaya's treatment produces a functional copy of the MYBPC3 gene to the cardiomyocytes. These transgenes produce the MyBP-C protein which carries out normal heart function.

In preclinical trials, TN-201 demonstrated that despite a 5x increase in RNA versus wild type genes, there was no protein overexpression:

In vivo comparison (Company website)

There was also higher selectivity for the heart than other cells elsewhere in the body. Preclinical data also showed signs of efficacy in mice models. A single dose in mice demonstrated reduced hypertrophy, durable improvement in cardiac function and extended survival. The phase 1b study, informed by this preclinical data, will begin dosing in Q3. It is an open label dose escalation and dose expansion study. Initial data is expected in 2024.

The small molecule HDAC6 inhibitor TN-301 targets HFpEF. HDAC6 inhibition is an area of recent research interest in stopping the progression of this disease. In 2021, data published in Nature from a study of CKD-506 showed improvements in exercise capacity, heart function, and quality of life. Standard treatments for HFpEF have not included this modality previously. Tenaya says that in preclinical studies, TN-301 has shown a differentiated mechanism versus SGLT2 inhibitors, which are part of the arsenal against HFpEF. The company will start a randomized, placebo-controlled phase 1 SAD/MAD study with safety and tolerability as primary endpoints and PK/PD as secondary endpoints. The company says that "Dosing Commenced in Multiple-Ascending Dose Stage of First-In-Human Clinical Trial of TN-301; Data Anticipated in Second Half 2023." I still do not see anything on the registry.

TNYA has a market cap of $180mn and a cash balance of $204mn. In November, the company raised $77mn through a secondary offering. R&D expenses were $25.7 million for the fourth quarter and G&A expenses were $8.8 million. At that rate, they have a cash runway extending into 2025.

According to insider data, the company saw heavy insider buying in recent months. I was especially glad to see insiders buying the secondary in the open market.

Insider transactions (openinsider.com)

The company has heavy institutional and PE/VC presence, with over 90% of the shares.

In the two years since I covered it last, TNYA has put together preclinical data for its assets. However, nothing has gone into the clinic, although the company has been in existence for nearly a decade, with hundreds of millions of dollars in funding available. I am afraid there's nothing to see here until we have the first proof of viability from the company. That should happen in 2024. We will take another look at that time.

Thanks for reading. At the Total Pharma Tracker, we offer the following:-

Our Android app and website feature a set of tools for DIY investors, including a work-in-progress software where you can enter any ticker and get extensive curated research material.

For investors requiring hands-on support, our in-house experts go through our tools and find the best investible stocks, complete with buy/sell strategies and alerts.

Sign up now for our free trial, request access to our tools, and find out, at no cost to you, what we can do for you.

See the rest here:
Tenaya Therapeutics Has Been An Early Stage Company For Too ... - Seeking Alpha

Google’s Ray Kurzweil says humans will be immortal by 2045 – Daily Mail

Would you like to live forever? Well, some experts say you might.

Last week, a formerGoogleengineer said he believes that humans will achieve immortality within the next eight years.

Ray Kurzweil - who has an 86 per cent success rate with his predictions - thinks that advances in technology will quickly lead to age-reversing 'nanobots'.

While it sounds far-fetched, scientists have been looking for years into ways we can regenerate our cells, or upload our minds to a computer.

MailOnline takes a look at the strangest ways humanity could attain eternal life.

Electronic immortality

Electronic immortality - Preserving brain after death and uploading the mind to a computer.

Freezing the brain - Cryogenically freezing the brain until technology advances to allow it to be brought back to life.

Cell rejuvenation- Rejuvenating ageing or damaged cells in the body by injecting them with stem cells.

Reanimating the brain- Pumping the brain with artificial blood to keep it alive.

The idea of uploading your mind to a computer has been theorised for many years now, but it has mostly remained the stuff of science fiction.

Nectome, a US-based startup, is trying to change that by devising a way to preserve the human brain so that its memories can be uploaded to the cloud.

The firm has figured out a way to preserve the human brain in microscopic detail using a 'high-tech embalming process,' according to the MIT Technology Review.

It uses a chemical solution that can keep the body intact for hundreds or thousands of years as a statue of frozen glass.

'You can think of what we do as a fancy form of embalming that preserves not just the outer details but the inner details,' said Robert McIntyre, Nectome's cofounder.

Speaking to prospective customers, Nectome positions its service as: 'What if we told you we could back up your mind?'

But the key to being able to recreate a person's consciousness involves accessing the organ's 'connectome.'

A connectome is the complex web of neural connections in the brain, often referred to as the brain's wiring system.

Nectome, which has been referred to as a 'preserve-your-brain-and-upload-it' company, has figured out a way to embalm the connectome as well.

However, in order for the technology to work, participants have to be willing to be euthanized, which led to it losing a contract with theMassachusetts Institute of Technology (MIT) in 2018.

The prestigious institution claimed the technology is in its infancy and there is no guarantee that they can recreate consciousness.

Despite the setback, that same year, a prominent futurist predicted that 'electronic immortality' would be available to humans by 2050.

Dr Ian Pearsonsaid that human intelligence, memory or senses could be connected to external technology.

Rather than creating a backed up copy of your mind, most of your intelligence would simply be running from a place outside of your physical brain.

In a blog post, he wrote: One day, your body dies and with it your brain stops,' he wrote in a blog post.

'But no big problem, because 99 per cent of your mind is still fine, running happily on IT, in the clouds.

Assuming you saved enough and prepared well, you connect to an android to use as your body from now on, attend your funeral, and then carry on as before, still you, just with a younger, highly upgraded body.

He adds that this type of immortality has dangers too, as itwould require the use of a purchased or rented android and cloud space ultimately owned by a tech company.

These companies could thus enslave workers after their deaths, by maintaining ownership of the mind for their own benefit down the line.

Maybe the cloud company could replicate your mind and make variations to address a wide range of markets, the futurist wrote.

Maybe they can use your mind as the UX on a new range of home-help robots. Each instance of you thinks they were once you, each thinks they are now enslaved to work for free for a tech company.

Cryogenics is the art of freezing bodies by preserving a dead body with liquid nitrogen.

Currently, it can only legally happen when someone has just been declared dead.

The freezing process must begin as soon as the patient dies in order to prevent brain damage, with facilities currently available in Russia and the US.

In the procedure, the body is cooled in an ice bath to gradually reduce its temperature bit by bit.

Experts then drain the blood and replace it with an anti freeze fluid to stop harmful ice crystals forming in the body.

Freezing the brain

Some companies offer the opportunity for people to have their brains frozen after they die,in the hope they can be brought back to life in the future.

One of these is Russiancryonics firm KrioRus, which currently has 91 human 'patients'stored at -320.8F (-196C) with the aim of protecting them against deterioration.

This is cold enough to stop all cellular function and preserve a body's state until defrost

This is so that they can potentially be revived in the future when science advances enough to cure any illness they may have had, including death itself, says KrioRus.

Their brains, or full bodies, are all currently floating in large vats of liquid nitrogen and housed in a corrugated metal shed outside Moscow.

It costs at least$28,000 (22,500) to be cryogenically preserved with this company.

It claims the service gives people left behind by dead relatives a 'peace of mind' and hope they will see them again.

They also freeze pets, and currently store 58 dogs, cats, birds, hamsters, rabbits and a chinchilla.

But, the head of the Russian Academy of Sciences's Pseudoscience Commission, Evgeny Alexandrov, described cryonics as 'an exclusively commercial undertaking that does not have any scientific basis', in comments to the Izvestia newspaper.

It is 'a fantasy speculating on people's hopes of resurrection from the dead and dreams of eternal life', the newspaper quoted him as saying.

Valeriya Udalova, KrioRus's director, had her dog frozen when it died in 2008, she says it helps people deal with loss.

She said it is likely that humankind will develop the technology to revive dead people in the future, but that there is 'no guarantee of such technology'.

This is by far the only company offering such a service, and there are thought to at least 500 bodies frozen in this way worldwide.

Another prominent company is the Alcor Life Extension Foundation in Arizona, USA, which had 199 patients as of October 2022.

There, full bodies are stored in large cylindrical chambers alongside three other full bodies. Brains can be stored in shelves, with five fitting into a slot for one body.

After a person dies, doctors must work fast to preserve the body and get it into storage.

Comparing the process to organ harvesting, Max More, CEO of Alcor, said in a 2020 interview that the first step was draining the body of its blood and liquids.

They then pump it full of an antifreeze-like substance. This is to prevent cells from becoming crystallised and damaged during the freezing process.

People who invest in these services are often desperate to reunite with family in the future.

The youngest known Alcor patient is a two-year-old Thai girl who died of brain cancer. Her family hopes to reunite with her down the line.

But cryogenic freezing also attracts the rich and eccentric. Bitcoin pioneer Hal Finney chose to have his body cryopreserved after he died from complications related to ALS in 2014.

There are serious ethical and moral concerns about the practice which has been touted for decades but remains a pipe dream.

The high prices of this preservation can often drain a person's estate, and will often consume a massive portion of their life-insurance payout - which could have instead benefited their family down the line.

Mr More admitted during an interview in February 2020 that his firm does not know when technology needed to wake up their patients will exist.

However, he is hopeful that this technology will exist and cited recent success in stem-cell research and lab-made organ growth as starting to pave the way forward.

Dr Michael Hendricks, a biologist at Canada's McGill University, wrote in 2015 that what makes a person's personality, sense of self, decision making and day-to-day mood are small connections between nerve cells.

But current technology has no way of perfectly storing these cells across the body, and changes to them would fundamentally change who a person is.

Cell rejuvenation

Many scientific breakthroughs have been made with regards to stem cell injections, which have been found to be able to rejuvenate cells.

Stem cells are unique because they can differentiate into different types of cells in the body, such as muscle, bone or nerve cells.

When injected into the body, they can integrate with damaged tissues and help to repair and regenerate them.

In 2016, stem cell injectionsreversed the scar tissuein a trial of 11 seriously ill patients who had suffered heart attacks, reducing scarring by 40 per cent.

Similarly, in 2019,Cambridge University researchersregenerated lost heart muscle and blood vessels in rats with damaged hearts after transplanting stem cells from a human heart.

Stem cells are found everywhere in the body, especially the bone marrow, standing ready to morph into the 200-odd types of cell that make up humans to repair damage.

But their numbers fall as we age, leaving older adults lacking the same regenerative capabilities as their younger peers.

Some creatures, like flatworms and hydras, have stem cells throughout their lives so are always able to regenerate lost body parts.

Dr Steven Cohen, who owns wellness clinics in California andLondon, says that stem cell therapy could be the key to extending the human life expectancy to up to 150.

Last month, he said his technology, which involves injecting people with exosomes, small vesicles that are naturally produced by stem cells, is just five years away.

The hope is that the exosomes - bursting with essential proteins, lipids, nucleic acids and others - will flow into organs and help to 'de-age' them, allowing someone to live longer.

A paper published last year found that more exosomes in the body boosted brain function, while another from the same year suggested they could reduce frailty and help someone live longer.

Other scientists have suggested people could one day live to the age of 200and are exploring technology like pills to flush out 'zombie cells' and ways to tweak DNA to extend someone's lifespan.

These cells stop dividing like others but start to spew a cocktail of harmful chemicals, damaging and degrading those around them.

Pills that flush these out are already in human trials with scientists saying they could hit the market in as little as 10 years.

A 2016 study from theSalk Institute in California claimed that the key tohalting or reversing ageing may lie in cellular reprogramming.

This is a process in which the expression of four genes, known as the Yamanaka factors, is induced, allowing scientists to convert any adult cell into induced pluripotent stem cells (iPSCs).

Like embryonic stem cells, which are derived from early-stage embryos, iPSCs are capable of dividing indefinitely and becoming any cell type present in our body.

The researchers found that when cellular reprogramming was induced in mice, their cells looked and acted younger.

Reanimating the brain

A technology that was developed to help scientists study brains in three dimensionscould also provide the key to eternal life.

In 2019, scientists at Yale Universityrestored the circulation and cellular activity in a pig's brainfour hours after its death by pumping it withoxygen-rich artificial blood.

Neuroscientist and lead author Dr Nenad Sestan said it is possible the brains could have been kept alive indefinitely and that additional steps could be taken to restore awareness, according to the Massachusetts Institute of Technology'sTechnology Review.

But he added that his team chose not to attempt either because 'this is uncharted territory.'

Chemicals added to prevent swelling during the procedure would likely prohibit consciousness indefinitely.

This means it may not be possible for the team to resuscitate brains that can still 'think' using their current methods.

The experiment's success provided a new way of studying the structure and function of the intact large mammalian brain.

'Previously, we have only been able to study cells in the large mammalian brain under static or largely two-dimensional conditions utilising small tissue samples outside of their native environment, said co-first author Stefano Daniele.

'For the first time, we are able to investigate the large brain in three dimensions, which increases our ability to study complex cellular interactions and connectivity.'

The team hoped these future 3D brain studies could help doctors find ways to salvage brain function in stroke patients, or test novel therapies.

But scientists also said it may one day allow humans to become immortal by hooking up our minds to artificial systems after our natural bodies have perished.

Nottingham Trent ethics and philosophy lecturer Benjamin Curtis said that this maylead to humans being locked in an eternal 'living hell' and enduring a 'fate worse than death.

'Even if your conscious brain were kept alive after your body had died, you would have to spend the foreseeable future as a disembodied brain in a bucket, locked away inside your own mind without access to the sense that allow us to experience and interact with the world,' Curtis told The Conversation.

'In the best case scenario you would be spending your life with only your own thoughts for company.'

Brain and memory preservation has been explored at length by futurists, scientists and science fiction junkies alike.

Read the original post:
Google's Ray Kurzweil says humans will be immortal by 2045 - Daily Mail

Global Induced Pluripotent Stem Cell (iPSC) Industry Report 2023 …

DUBLIN, Feb. 13, 2023 /PRNewswire/ --The "Global Induced Pluripotent Stem Cell (iPSC) Industry Report - Market Size, Trends, and Forecasts, 2023" report has been added to ResearchAndMarkets.com's offering.

Since the discovery of induced pluripotent stem cell (iPSC) technology in 2006, significant progress has been made in stem cell biology and regenerative medicine. New pathological mechanisms have been identified and explained, new drugs identified by iPSC screens are in the pipeline, and the first clinical trials employing human iPSC-derived cell types have been initiated. iPSCs can be used to explore the causes of disease onset and progression, create and test new drugs and therapies, and treat previously incurable diseases.

Today, methods of commercializing induced pluripotent stem cells (iPSCs) include:

Since the discovery of iPSCs in 2006, it took only seven years for the first iPSC-derived cell product to be transplanted into a human patient in 2013. Since then, iPSC-derived cells have been used within a rapidly growing number of preclinical studies, physician-led studies, and clinical trials worldwide. There are also over 100 clinical trials underway that do not involve the transplant of iPSCs into humans, but rather, the creation and evaluation of iPSC lines for clinical purposes. Within these trials, iPSC lines are created from specific patient populations to determine if these cell lines could be a good model for a disease of interest.

2013 was a landmark year because it saw the first cellular therapy involving the transplant of iPSCs into humans initiated at the RIKEN Center in Kobe, Japan. Led by Dr. Masayo Takahashi, it investigated the safety of iPSC-derived cell sheets in patients with macular degeneration. In another world first, Cynata Therapeutics received approval in 2016 to launch the first formal clinical trial of an allogeneic iPSC-derived cell product (CYP-001) for the treatment of GvHD. CYP-001 is a iPSC-derived MSC product. In this historic trial, CYP-001 met its clinical endpoints and produced positive safety and efficacy data for the treatment of steroid-resistant acute GvHD.

Given this early success, Cynata is has advanced its iPSC-derived MSCs into Phase 2 trials for the severe complications associated with COVID-19, as well as GvHD and critical limb ischemia (CLI). It is also undertaking an impressive Phase 3 trial that will utilize Cynata's iPSC-derived MSC product, CYP-004, in 440 patients with osteoarthritis (OA). This trial represents the world's first Phase 3 clinical trial involving an iPSC-derived cell therapeutic product and the largest one ever completed. Not surprisingly, the Japanese behemoth FUJIFILM has been involved with the co-development and commercialization of Cynata's iPSC-derived MSCs through its 9% ownership stake in the company.

Many market competitors are also commercializing iPSC-derived products for use in drug development and discovery, disease modeling, and toxicology testing. Across the broader iPSC sector, FUJIFILM CDI (FCDI) is one of the largest and most dominant players. Cellular Dynamics International (CDI) was founded in 2004 by Dr. James Thomson at the University of Wisconsin-Madison, who in 2007 derived iPSC lines from human somatic cells for the first time. The feat was accomplished simultaneously by Dr. Shinya Yamanaka's lab in Japan. FUJIFILM acquired CDI in April 2015 for $307 million. Today, the combined company is the world's largest manufacturer of human cells created from iPSCs for use in research, drug discovery and regenerative medicine applications.

Another iPSC specialist is ReproCELL, a company that was established as a venture company originating from the University of Tokyo and Kyoto University in 2009. It became the first company worldwide to make iPSC products commercially available when it launched its ReproCardio product, which are human iPSC-derived cardiomyocytes. Within the European market, the dominant competitors are Evotec, Ncardia, and Axol Bioscience. Headquartered in Hamburg, Germany, Evotec is a drug discovery alliance and development partnership company. It is developing an iPSC platform with the goal to industrialize iPSC-based drug screening as it relates to throughput, reproducibility, and robustness. Today, Evotec's infrastructure represents one of the largest and most advanced iPSC platforms globally.

Ncardia was formed through the merger of Axiogenesis and Pluriomics in 2017. Its predecessor, Axiogenesis, was founded in 2011 with an initial focus on mouse embryonic stem cell-derived cells and assays. When Yamanaka's iPSC technology became available, Axiogenesis became the first European company to license it in 2010. Today, the combined company (Ncardia) is a global authority in cardiac and neural applications of human iPSCs. Founded in 2012, Axol Bioscience is a smaller but noteworthy competitor that specializes in iPSC-derived products. Headquartered in Cambridge, UK, it specializes in human cell culture, providing iPSC-derived cells and iPSC-specific cell culture products.

Of course, the world's largest research supply companies are also commercializing a diverse range of iPSC-derived products and services. Examples of these companies include Lonza, BD Biosciences, Thermo Fisher Scientific, Merck, Takara Bio, and countless others. In total, at least 80 market competitors now offer a diverse range of iPSC products, services, technologies, and therapeutics.

Key Topics Covered:

1. Report Overview

2. Introduction

3. Current Status of iPSC Industry

4. History of Induced Pluripotent Stem Cells (iPSCs)

5. Research Publications on iPSCs

6. iPSC: Patent Landscape Analysis

7. iPSC: Clinical Trial Landscape

8. Research Funding for iPSCs

9. M&A, Collaborations & Funding Activities in iPSC Sector

10. Generation of Induced Pluripotent Stem Cells: An Overview

11. Human iPSC Banking

12. Biomedical Applications of iPSCs

13. Market Overview

14. Company Profiles

Selection of Companies Mentioned

For more information about this report visit https://www.researchandmarkets.com/r/2q9nbt-induced?w=5

About ResearchAndMarkets.com ResearchAndMarkets.com is the world's leading source for international market research reports and market data. We provide you with the latest data on international and regional markets, key industries, the top companies, new products and the latest trends.

Media Contact:

Research and Markets Laura Wood, Senior Manager [emailprotected]

For E.S.T Office Hours Call +1-917-300-0470 For U.S./CAN Toll Free Call +1-800-526-8630 For GMT Office Hours Call +353-1-416-8900

U.S. Fax: 646-607-1904 Fax (outside U.S.): +353-1-481-1716

Logo: https://mma.prnewswire.com/media/539438/Research_and_Markets_Logo.jpg

SOURCE Research and Markets

Read more:
Global Induced Pluripotent Stem Cell (iPSC) Industry Report 2023 ...

Induced Pluripotent Stem Cells (iPSCs)-Roles in Regenerative Therapies …

Title & authors Abstract Conflict of interest statement Figures Similar articles Cited by References Publication types MeSH terms Related information Grant support LinkOut - more resources

Review

Affiliations Expand

Item in Clipboard

Review

Mourad A M Aboul-Soudet al. Cells. 2021.

Display options

Format Abstract PubMed PMID

Item in Clipboard

Display options

Format Abstract PubMed PMID

The discovery of induced pluripotent stem cells (iPSCs) has made an invaluable contribution to the field of regenerative medicine, paving way for identifying the true potential of human embryonic stem cells (ESCs). Since the controversy around ethicality of ESCs continue to be debated, iPSCs have been used to circumvent the process around destruction of the human embryo. The use of iPSCs have transformed biological research, wherein increasing number of studies are documenting nuclear reprogramming strategies to make them beneficial models for drug screening as well as disease modelling. The flexibility around the use of iPSCs include compatibility to non-invasive harvesting, and ability to source from patients with rare diseases. iPSCs have been widely used in cardiac disease modelling, studying inherited arrhythmias, neural disorders including Alzheimer's disease, liver disease, and spinal cord injury. Extensive research around identifying factors that are involved in maintaining the identity of ESCs during induction of pluripotency in somatic cells is undertaken. The focus of the current review is to detail all the clinical translation research around iPSCs and the strength of its ever-growing potential in the clinical space.

Keywords: disease; drug screening; embryo; induced pluripotent stem cells; modelling.

The authors declare that they have no conflict of interest.

Figure 1

Showing the process of progression

Figure 1

Showing the process of progression and generating iPSC cells. Detailed description of creating

Showing the process of progression and generating iPSC cells. Detailed description of creating iPSCs with reprogramming factors and differentiating them into a variety of cell types.

Figure 2

Schematic representation on derivation and

Figure 2

Schematic representation on derivation and assay for human iPSCs. Detailed schematic representation of

Schematic representation on derivation and assay for human iPSCs. Detailed schematic representation of derivation of iPSC with the various assays to evaluate the developmental efficiency.

Figure 3

Process of liver development and

Figure 3

Process of liver development and hepatic differentiation from hiPSCs. The process of isolated

Process of liver development and hepatic differentiation from hiPSCs. The process of isolated cells from patients can be cultured and reprogrammed into patient-specific hiPSCs and quick comparison from natural liver development.

Skalova S, Svadlakova T, Shaikh Qureshi WM, Dev K, Mokry J. Skalova S, et al. Int J Mol Sci. 2015 Feb 13;16(2):4043-67. doi: 10.3390/ijms16024043. Int J Mol Sci. 2015. PMID: 25689424 Free PMC article. Review.

Ohnuki M, Takahashi K. Ohnuki M, et al. Philos Trans R Soc Lond B Biol Sci. 2015 Oct 19;370(1680):20140367. doi: 10.1098/rstb.2014.0367. Philos Trans R Soc Lond B Biol Sci. 2015. PMID: 26416678 Free PMC article. Review.

Valdimarsdttir G, Richter A. Valdimarsdttir G, et al. Laeknabladid. 2015 Dec;101(12):581-6. doi: 10.17992/lbl.2015.12.56. Laeknabladid. 2015. PMID: 26656400 Review. Icelandic.

Parrotta EI, Scalise S, Scaramuzzino L, Cuda G. Parrotta EI, et al. Int J Mol Sci. 2019 Nov 16;20(22):5760. doi: 10.3390/ijms20225760. Int J Mol Sci. 2019. PMID: 31744081 Free PMC article. Review.

Okano H, Nakamura M, Yoshida K, Okada Y, Tsuji O, Nori S, Ikeda E, Yamanaka S, Miura K. Okano H, et al. Circ Res. 2013 Feb 1;112(3):523-33. doi: 10.1161/CIRCRESAHA.111.256149. Circ Res. 2013. PMID: 23371901 Review.

Ng WC, Lokanathan Y, Baki MM, Fauzi MB, Zainuddin AA, Azman M. Ng WC, et al. Biomedicines. 2022 Nov 30;10(12):3082. doi: 10.3390/biomedicines10123082. Biomedicines. 2022. PMID: 36551838 Free PMC article. Review.

Shakir S, Hackett TL, Mostao-Guidolin LB. Shakir S, et al. Front Bioeng Biotechnol. 2022 Oct 28;10:1011800. doi: 10.3389/fbioe.2022.1011800. eCollection 2022. Front Bioeng Biotechnol. 2022. PMID: 36394026 Free PMC article. Review.

Alberti G, Russo E, Corrao S, Anzalone R, Kruzliak P, Miceli V, Conaldi PG, Di Gaudio F, La Rocca G. Alberti G, et al. Biomedicines. 2022 Nov 5;10(11):2822. doi: 10.3390/biomedicines10112822. Biomedicines. 2022. PMID: 36359342 Free PMC article. Review.

Linkova DD, Rubtsova YP, Egorikhina MN. Linkova DD, et al. Cells. 2022 Aug 29;11(17):2691. doi: 10.3390/cells11172691. Cells. 2022. PMID: 36078098 Free PMC article. Review.

Li XW, Lu YY, Zhang SY, Sai NN, Fan YY, Cheng Y, Liu QS. Li XW, et al. Front Pharmacol. 2022 Jun 22;13:926123. doi: 10.3389/fphar.2022.926123. eCollection 2022. Front Pharmacol. 2022. PMID: 35814256 Free PMC article.

Show all 116 references

Cite

Format: AMA APA MLA NLM

Read more:
Induced Pluripotent Stem Cells (iPSCs)-Roles in Regenerative Therapies ...

Induced pluripotent stem cell technology: a decade of progress

Title & authors Abstract Conflict of interest statement Figures Similar articles Cited by Publication types MeSH terms Grant support LinkOut - more resources

Review

Affiliations Expand

Item in Clipboard

Review

Yanhong Shiet al. Nat Rev Drug Discov. 2017 Feb.

Display options

Format Abstract PubMed PMID

Item in Clipboard

Display options

Format Abstract PubMed PMID

Since the advent of induced pluripotent stem cell (iPSC) technology a decade ago, enormous progress has been made in stem cell biology and regenerative medicine. Human iPSCs have been widely used for disease modelling, drug discovery and cell therapy development. Novel pathological mechanisms have been elucidated, new drugs originating from iPSC screens are in the pipeline and the first clinical trial using human iPSC-derived products has been initiated. In particular, the combination of human iPSC technology with recent developments in gene editing and 3D organoids makes iPSC-based platforms even more powerful in each area of their application, including precision medicine. In this Review, we discuss the progress in applications of iPSC technology that are particularly relevant to drug discovery and regenerative medicine, and consider the remaining challenges and the emerging opportunities in the field.

Conflict of Interest

J.C.W. is a co-founder of Stem Cell Theranostics. S.Y. is a scientific advisor of iPS Academia Japan without salary. The other authors declare no conflict of interest.

Figure 1

A schematic for human iPSC-based

Figure 1

A schematic for human iPSC-based disease modeling. Human iPSCs are derived from individual

A schematic for human iPSC-based disease modeling. Human iPSCs are derived from individual patients and differentiated into specific cell types. To develop new therapies, the resultant cells are used to observe disease-specific phenotypes and identify novel pathological mechanisms,. Human iPSC-based disease modeling with patient-specific cells now provides an exciting new approach for the development of personalized diagnosis and medicine.

Figure 2

A schematic for human iPSCs-based

Figure 2

A schematic for human iPSCs-based cell therapy. Human iPSC-based cell therapy development usually

A schematic for human iPSCs-based cell therapy. Human iPSC-based cell therapy development usually includes the following steps: 1) Collect somatic cells from patients and culture somatic cells from affected patients; 2) Reprogram patient somatic cells into iPSCs; 3) Use genome editing technology or viral transduction method to repair patient iPSCs and turn them into genetically corrected iPSCs; 4) Differentiate the corrected iPSCs into desired cell types to serve as genetically matched healthy donor cells; 5) Perform quality control test for cell identity, purity, activity, and safety; and 6) Transplant the genetically matched healthy cells into patients for cell therapy.

Doss MX, Sachinidis A. Doss MX, et al. Cells. 2019 Apr 30;8(5):403. doi: 10.3390/cells8050403. Cells. 2019. PMID: 31052294 Free PMC article. Review.

Wang F, Kong J, Cui YY, Liu P, Wen JY. Wang F, et al. Chin Med J (Engl). 2018 Apr 5;131(7):852-856. doi: 10.4103/0366-6999.228231. Chin Med J (Engl). 2018. PMID: 29578130 Free PMC article. Review.

Wattanapanitch M. Wattanapanitch M. Stem Cells Int. 2019 May 2;2019:5171032. doi: 10.1155/2019/5171032. eCollection 2019. Stem Cells Int. 2019. PMID: 31191673 Free PMC article. Review.

Nsair A, MacLellan WR. Nsair A, et al. Adv Drug Deliv Rev. 2011 Apr 30;63(4-5):324-30. doi: 10.1016/j.addr.2011.01.013. Epub 2011 Mar 1. Adv Drug Deliv Rev. 2011. PMID: 21371511 Free PMC article. Review.

Cai CY, Meng FL, Rao L, Liu YY, Zhao XL. Cai CY, et al. Yi Chuan. 2020 Nov 20;42(11):1042-1061. doi: 10.16288/j.yczz.20-235. Yi Chuan. 2020. PMID: 33229312 Review. Chinese.

Pilipovi K, Harej Hrka A, Kui N, Mri-Peli J. Pilipovi K, et al. Biomedicines. 2022 Dec 29;11(1):94. doi: 10.3390/biomedicines11010094. Biomedicines. 2022. PMID: 36672601 Free PMC article. Review.

Lien CY, Chen TT, Tsai ET, Hsiao YJ, Lee N, Gao CE, Yang YP, Chen SJ, Yarmishyn AA, Hwang DK, Chou SJ, Chu WC, Chiou SH, Chien Y. Lien CY, et al. Cells. 2023 Jan 4;12(2):211. doi: 10.3390/cells12020211. Cells. 2023. PMID: 36672144 Free PMC article.

Akbarzadeh A, Sobhani S, Soltani Khaboushan A, Kajbafzadeh AM. Akbarzadeh A, et al. Bioengineering (Basel). 2023 Jan 12;10(1):106. doi: 10.3390/bioengineering10010106. Bioengineering (Basel). 2023. PMID: 36671678 Free PMC article. Review.

Han JL, Entcheva E. Han JL, et al. Stem Cell Rev Rep. 2023 Jan 19:1-20. doi: 10.1007/s12015-023-10506-4. Online ahead of print. Stem Cell Rev Rep. 2023. PMID: 36656467 Free PMC article. Review.

Mohd Satar A, Othman FA, Tan SC. Mohd Satar A, et al. World J Stem Cells. 2022 Dec 26;14(12):851-867. doi: 10.4252/wjsc.v14.i12.851. World J Stem Cells. 2022. PMID: 36619694 Free PMC article.

Cite

Format: AMA APA MLA NLM

Read more:
Induced pluripotent stem cell technology: a decade of progress