The first multi-chamber heart organoids developed – Drug Target Review

The first multi-chamber cardioids derived from hiPSCs have enabled scientists to investigate heart development and defects.

Researchers, led by Dr Sasha Mendjan at the Institute of Molecular Biotechnology (IMBA) of the Austrian Academy of Sciences, have developed the first multi-chamber heart organoids that reflect the organs intricate structures. This promises advanced screening platforms for understanding heart development, drug development and toxicology studies.

The leading cause of death worldwide is cardiovascular disease, yet there are limited therapies for it. Similarly, one in 50 babies born suffer from a congenital heart defect but scientists have little understanding of why these occur. However, the team at IMBA have produced a new physiological organoid model that comprises the major regions of the human heart, enabling scientists to study cardiac disease and development.

In 2021, the Mendjan lab developed the first chamber-like heart organoid formed from human induced pluripotent stem cells (hiPSCs). hiPSCs have many benefits, such as overcoming the ethical and immune-compatibility issues faced due to the use of human embryonic stem cells (hESCs). hiPSCs can be derived from patient-specific somatic cells (eg, skin fibroblasts and hematopoietic cells) and be directly reprogrammed by defined factors to induce pluripotency. These hiPSCs displayed similarities in morphology, proliferation, feeder dependence, surface markers, gene expression, promoter activities, in vitro differentiation potential, and teratoma formation characteristics to hESCs.1

These heart organoids, named cardioids, were self-organising and mimicked the development of the hearts left ventricular chamber in the very early days of embryogenesis. Dr Mendjan said: These cardioids were a proof-of-principle and an important step forwardWhile most adult diseases affect the left ventricle, which pumps oxygenated blood through the body, congenital defects affect mostly other heart regions essential to establish and maintain circulation.

For the new study, the IMBA scientists furthered this work and derived organoid model of each developing heart structure individually. Dr Mendjan explained: Then we asked: If we let all these organoids co-develop together, do we get a heart model that co-ordinately beats like the early human heart?

The researchers grew the left and right ventricular and the atrial organoids together. Dr Mendjan remarked: Indeed, an electrical signal spread from the atrium to the left and then the right ventricular chambers just like in early foetal heart development in animalsWe now observed this fundamental process in a human heart model for the first time, with all its chambers.

We now observed this fundamental process in a human heart model for the first time, with all its chambers.

This model allowed the team to investigate how regional gene expression differences led to specific chamber contraction patterns and the intricate communication between them.

Also, insight was gained into early heart development, especially how the human heart starts beating, which was previously unknown. One of the studys first authors Alison Deyett, a PhD student in the Mendjan group detailed: At first, the left ventricular chamber leads the budding right ventricular and atrium chambers at its rhythm. Then, as the atrium develops two days later the ventricles follow the atrial lead. This mirrors what is seen in animals before the final leaders, the pacemakers, control the heart rhythm.

Multi-chamber cardioids also allowed the scientists to study chamber-specific defects. The team established a screening platform for defects for a proof-of-principle experiment, in which they investigated how teratogens and mutations affect hundreds of heart organoids simultaneously.

Thalidomide, a well-known teratogen in humans, as well as retinoid derivatives, that are used in treatments against leukaemia, psoriasis, and acne, are known to cause severe heart defects in the foetus. Both teratogens induced similar, serious compartment-specific defects in the heart organoids. Similarly, mutations in three cardiac transcription factor genes resulted in chamber-specific defects observed in human development. Dr Mendjan summarised: Our tests show that multi-chamber cardioids recapitulate embryonic heart development and can uncover disruptive effects on the whole heart with high specificity. We do this using a holistic approach, looking at multiple readouts simultaneously.

Someday, multi-chamber heart organoids could be used for toxicology studies and to develop novel drugs with heart chamber-specific effects. Drug-induced cardiotoxicity is the leading cause of drug attrition during the development process,2 so these organoids are promising for the future.

Dr Mendjan said: For example, atrial arrhythmias are widespread, but we currently dont have good drugs to treat it. One reason is that no models existed comprising all regions of the developing heart working in a coordinated manner so far.

Developing heart organoids from patient-derived stem cells may provide insight into developmental defects and its potential treatment and prevention, which the Mendjan lab hope to understand further.

This study was published in Cell.

1 Ho Beatrice Xuan, Pek Nicole Min Qian, Soh Boon-Seng. Disease Modeling Using 3D Organoids Derived from Human Induced Pluripotent Stem Cells. International Journal of Molecular Sciences (IJMS) [Internet]. 2018 March 21 [2023 December 7];19(4)936. Available from: https://doi.org/10.3390/ijms19040936

2 Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, et al. Lessons Learned from the Fate of AstraZenecas Drug Pipeline: a Five-Dimensional Framework. Nature Review Drug Discovery. 2014 May 16 [2023 December 7];13(6)419-431. Available from: https://www.nature.com/articles/nrd4309

View post:
The first multi-chamber heart organoids developed - Drug Target Review

Adult stem cell activity in naked mole rats for long-term tissue maintenance – Nature.com

Ethics

This study involved undertaking animal procedures in four different countries: U.K, USA, Austria, and the Republic of South Africa. Animal procedures were carried out in accordance with Home Office, UK regulations and the Animals (Scientific Procedures) Act, 1986 of UK, the Institutional Animal Care and Use Committee (IACUC) of USA, Act 7, 1991 of South Africa, and the Directive 2010/63/EU of the European Parliament.

Normal human colonoscopy samples were collected under the research tissue bank ethics 16/YH/0247 supported by NIHR Biomedical Research Centre, Oxford, U.K. and under the London Dulwich Research Ethics Committee (reference number 15/LO/1998). Written informed consent was obtained from all participants undergoing routine bowel cancer or IBD screening. All samples were anonymized.

Wild-caught mice (F1) were acquired from a founder population trapped in lower Austria and Vienna (2016) and housed at the Konrad Lorenz Institute of Ethology, University of Vienna, Austria. All C57BL/6J mice used in this study were purchased from Charles River (Kent, UK) or the Jackson Laboratory (USA) and housed at Biomedical Services Unit in John Radcliffe Hospital, Oxford, UK or at Rutgers University Animal Facility in Newark, New Jersey, USA. Mice were housed in individually ventilated cages under specific pathogen-free conditions and maintained at 1923C temperature with 45-65% relative humidity, in an alternating 12-h light/12-h dark cycles and fed with food and water ad libitum.

Naked mole rats (NMRs) were housed at the Animal Facility of the Department of Zoology and Entomology, University of Pretoria. The NMRs were kept in tunnel systems consisting of several Perspex chambers containing wood shavings as nestling material. The NMR room was maintained at temperatures ranging between 2932C, with relative humidity around 40-60%. NMRs were fed chopped fresh fruits and vegetables (apple, sweet potato, cucumber, and capsicum) daily ad libitum along with weekly supplement of ProNutro (Bokomo). Since NMRs obtain all their necessary water from food sources, no drinking water was provided to the animals. All scientific procedures on NMRs were conducted under ethics approval (NAS046-19 and NAS289-2020) by the Animal Ethics Committee, University of Pretoria. In addition, DAFF section 20 approval was granted (SDAH-Epi-20111909592).

For all analyses, both male and female mice, NMRs, and humans were included in the study.

15mg/mL solution of BrdU (5-bromo-2-deoxyuridine, Abcam, ab142567) and 12.3mg/mL solution of EdU (5-ethynyl-2-deoxyuridine, Merck, 900584) were prepared in sterile 1 PBS (Gibco, 10010023) and filtered through a 0.2m strainer. Using a 27-gauge needle and 1mL syringe, 100mg per kg bodyweight BrdU and 82.14mg per kg bodyweight EdU were administered intraperitoneally. Animals were checked regularly for signs of discomfort (hunched back, shivering, low mobility) after the injection.

For cumulative labelling protocol using BrdU, the first injection in naked mole rats was administered between 14:00 to 15:00. Subsequent BrdU injections were given every 8h for a duration of 5 days and intestinal tissues were collected every 8h after the first injection. In C57BL/6J mice, the first BrdU injection was also given between 14:00 to 15:00, with further injections given every 6h for a total of 2.25 days. Mouse intestinal tissues were collected 1h after each injection. The rationale for the frequency and total number of injections in the two species is discussed in Supplementary Note1.

Dextran sulphate sodium (DSS) salt (Merck, 42867) was dissolved in sterile ddH2O to prepare 0 to 8.75% (w/V) solution. Using a 2mL syringe fitted with a plastic feeding tube (Prime Bioscience, FTP-20-38), 50mL per kg bodyweight of DSS solution in NMRs or 12mL per kg bodyweight in mice was administered orally at specific intervals for 3 days. Body mass was monitored daily and stool samples collected while animals were also checked for signs of discomfort (e.g. hunched back, shivering, low mobility) every 3h.

After sacrificing the animals by approved procedures, the intestine was immediately isolated from the abdominal cavity and fatty tissue was removed. The small intestine was then divided into three equal sections: SB1 (duodenum), SB2 (jejunum) and SB3 (ileum). All three parts of the small intestine and colon were then flushed with 1 PBS (Phosphate Buffered Saline) solution using a P1000 pipette to clean all the faecal material. Each tissue section was then cut open longitudinally using a gut cutting device86 and the edges pinned down onto a 3MM filter paper such that the luminal side was facing upward. The tissue was then fixed in 10% neutral buffered formalin overnight at room temperature. The following day fixed intestinal tissues were rolled using the Swiss-rolling technique87 and stored in 70% ethanol at 4C. Next, formalin-fixed Swiss-rolls were dehydrated through increasing concentrations of ethanol, cleared through xylene, and embedded in paraffin. The paraffin blocks were sectioned at 4m thickness using a microtome (Anglia Scientific).

Tissue sections on SuperFrost Plus slides (VWR, 6310108) were deparaffinized by submerging slides in xylene (2 times, 10min each) and rehydrated in 100% ethanol (2 times, 5min each), 95% ethanol (2min), 70% ethanol (2min), 50% ethanol (2min), and distilled water (5min). Sections were then stained with Harris Haematoxylin (Merck, HHS32) for 2min 45s followed by washing in running tap water for 5min. Next, slides were dipped in 95% ethanol ten times before sections were counter-stained with Eosin solution (Merck, 117081) for 3min. This was followed by tissue sections being dehydrated in 95% ethanol (15s) and 100% ethanol (2 times, 15s each), dipped in xylene (2 times, 5min each), and finally coverslipped using DPX Mountant (Merck, 06522).

Tissue sections on SuperFrost Plus slides (VWR, 6310108) were first deparaffinized with xylene (2 times, 5min each). They were rehydrated in 100%, 90%, 70% ethanol (5min each) and tap water (2min), dipped in 3% acetic acid solution (3min) before staining with Alcian blue 8GX (Merck, A5268) solution (pH 2.5) for 30min. Tissue sections were then washed (5min) in running tap water and counterstained (5min) with Nuclear Fast Red (Merck, N3020). After 1min wash in running tap water again, tissue sections were dehydrated in ethanol, dipped in xylene and finally coverslipped using DPX Mountant (Merck, 06522).

To preserve the mucus layer of the colonic epithelium, contact with any aqueous solution was avoided after the excision of the intestinal tissue. Without removing the faecal matter, several segments of the colon were cut using a scalpel and fixed overnight at room temperature in methacran/Carnoys solution which was composed of 60% methanol, 30% chloroform, and 10% glacial acetic acid. On the second day, fixed tissues were processed in 100% methanol (2 times, 30min each), 100% ethanol (3 times, 60min each) and xylene (2 times, 60min each). Processed tissues were embedded in paraffin and 4m thick sections cut and stained with Alcian blue as described above. Stained tissues were photomicrographed at 60 magnification on an Olympus BX51 brightfield microscope. For both NMRs and mice, 30 independent measurements of the mucus layer were taken from 3 animals using the measure tool in Fiji package88.

Tissue sections on SuperFrost Plus slides (VWR, 6310108) were deparaffinized in xylene (2 times, 5min each) and rehydrated in 100%, 90%, 70% ethanol (5min each) and distilled water (5min). A hydrophobic barrier was drawn around the tissue sections using a PAP pen (Vector Lab, H-4000) before incubating in the AB solution (AP Staining kit, SystemBio, AP100B-1) for 20min at room temperature in the dark. All sections were then washed in 1 PBS (5min, on a shaker), counterstained with Nuclear Fast Red (5min), washed in running tap water (1min), dehydrated in ethanol, dipped in xylene and finally coverslipped with DPX Mountant (Merck, 06522).

4m thick formalin-fixed paraffin-embedded (FFPE) sections were cut using a microtome and dried overnight on SuperFrost Plus slides (VWR, 6310108). Tissue sections were baked at 60C for 1h the next day, deparaffinized in 3 rounds of xylene (5min each) and rehydrated in 100%, 90%, 70% ethanol and distilled H2O (5min each). Endogenous peroxidase activity was quenched by incubating sections in 3% H2O2 (Merck, 8222871000) for 20min. A heat mediated antigen retrieval was performed by boiling sections in 10mM sodium citrate buffer (pH 6.0) for 10min which was followed by 20min of cooling down in the same solution. This was followed by incubating the tissue sections in 1 PBSTX (0.1% Triton X) for 10min. All sections were then blocked for 1h at room temperature using 5% serum which matched the species of the secondary antibody. Next, primary antibodies were diluted in antibody diluent (1% BSA dissolved in 1 PBS) which was applied to the tissue sections and incubated overnight at 4C. The primary antibodies used in this study were Chromogranin A (Abcam, ab15160) at 1:2000 and BrdU (Abcam, ab6326) at 1:500. It is noteworthy that in our BrdU staining, we did not use HCl-mediated DNA denaturation and only performed heat-mediated antigen retrieval (98-100C) which has been shown to produce a brighter signal than acid hydrolysis89. After 3 rounds of washes (5min each) with 1 PBST (0.1% Tween20 in 1 PBS), tissue sections were then incubated for 1h at room temperature with biotinylated secondary antibodies diluted at 1:300. For our study specifically, we used goat anti-rabbit IgG (Vector Laboratories, BA-1000) and goat anti-rat IgG (Abcam, ab207997). To detect the biotinylated target, we used the Avidin/Biotinylated enzyme Complex (ABC) kit (Vector Laboratories, PK-6101) and developed the signal using the DAB (3,3-diaminobenzidine) solution (R&D systems, 4800-30-07). The tissue sections were then counterstained with Harris Haematoxylin (Merck, HHS32) for 5s, dehydrated in 70%, 90% and 100% ethanol for 15s each, dipped in xylene and coverslipped using DPX Mountant (Merck, 06522).

Species-specific RNAscope probes from ACD Bio-techne were used to detect Lgr5 mRNA expression in NMR (584631), mouse (312171) and human (311021) intestinal tissues. We used the RNAscope Multiplex Fluorescent Detection Kit v2 (ACD Bio-techne, 323110) and followed the instructions of the manufacturer (document number 323100-USM, ACD Bio-techne) to detect Lgr5 mRNA targets at a single cell level in FFPE tissue sections mounted on SuperFrost Plus slides (VWR, 6310108).

To enable multiplexing of mRNA and proteins, we adapted the manufacturers instructions (document number 323100-USM, ACD Bio-techne) for RNAscope Multiplex Fluorescent Detection Kit v2 (ACD Bio-techne, 323110) to exclude the step involving protease treatment. Once the mRNA signal was developed, we proceeded to detect proteins by first washing tissue sections (2 times, 2min each) in 1 TBST (0.1% Tween20 in 1 Tris-buffered saline). This was followed by blocking for 1h at room temperature with 10% serum which matched the species of the secondary antibodies. Multiple primary antibodies (diluted in 1% BSA in 1 TBS) were then applied to the tissue sections and incubated overnight at 4C. The dilutions of various primary antibodies used in our study were 1:500 for EpCAM (Abcam, ab71916), 1:500 for Ki67 (Cell Signaling, 12202), 1:200 for p27Kip1 (Cell Signaling, 3686 and 2552), 1:500 for BrdU (Abcam, ab6326) and 1:2000 for PHH3-S28 (Abcam, ab32388). Following primary antibody incubation, the next day we washed the sections thrice in 1 TBST (5min each) before incubating them with fluorophore-linked secondary antibodies (at 1:500 dilution) for 1h at room temperature. Fluorescent secondary antibodies used in our study included goat anti-rabbit Alexa 488 (Invitrogen, A11008), goat anti-rat Alexa 488 (Invitrogen, A11006), goat anti-rabbit Alexa 555 (Invitrogen, A21428) and goat anti-rabbit Alexa 633 (Invitrogen, A21070). Following the secondary antibody incubation, tissue sections were washed three times in 1 TBST (5min each) and counterstained with DAPI (Invitrogen, D1306) for 15min at room temperature before mounting with coverslips (VWR, 631-0138) using Diamond Antifade Mountant (Invitrogen, P36961).

Click-iT Plus TUNEL Assay Kit (Invitrogen, C10617) was used following the manufacturers instructions to detect apoptotic cells FFPE tissue sections.

EdU-Click 488 kit (Base Click, BCK-EdU488-1) was used according to the instructions provided by the manufacturer to detect EdU-positive cells in FFPE tissue sections.

Plasma BrdU concentration was determined following the protocol described by Barker et al.90. In brief, 100L naked mole rat blood was collected by a tail vein puncture after 8hand 16h of BrdU injection. The blood was mixed with heparin to stop clotting and centrifuged at 13,000g for 15min to separate all blood cells. Plasma was collected from the top layer and stored at 80C.

HEK293T cells (ATCC, CRL-3216) were cultured in high-glucose DMEM (Merck, D6546) containing 10% FBS (Gibco, 10270), 1 Penicillin-Streptomycin (Merck, P4333-100ML), and 2mM l-glutamine (Gibco, 25030-024) at 37C with 5% CO2. Cells were plated on a 13mm sterile glass coverslip precoated with poly l-lysine (VWR, 631-0149) in a 24-well plate (Starlab, CC7682-7524) and cultured overnight. The media was replaced with 500L fresh culture media containing 10L plasma or standard BrdU solution (3, 10, 20, 30, 40, 50g/ml) and incubated at 37C for 4h. Cells were then washed with 1 PBS and fixed in 4% paraformaldehyde for 20min at room temperature. Fixed cells were kept in 1 PBS at 4C before proceeding to immunocytochemical detection of BrdU.

Fixed cells on coverslips in 24 well plates were incubated with 3% H2O2 for 10min at room temperature. After washing with 1 PBS, cells were incubated in 2N HCl for 1h at room temperature to denature DNA strands. Fixed cells were then incubated in 0.1M Borate buffer (pH 8.5) for 30min at room temperature and in 1 PBSTX (0.1% Triton X) for 10min. Cells were blocked with 5% goat serum for 1h at room temperature and incubated with rat anti-BrdU primary antibody (Abcam, ab6326, 1:2000) overnight at 4C. The next day, cells were washed three times in 1 PBST and incubated with goat anti-rat-biotin-linked secondary antibody (Abcam, ab207997, 1:400) for 1h at room temperature. The biotinylated signal was developed using the ABC Kit (Vectastain, PK-6101) following the manufacturers instructions and detected with DAB solution (R&D systems, 4800-30-07). Gills No. 3 Haematoxylin (Merck, GHS316-500ML) was used for counterstaining and cells on the coverslips were mounted on glass slides using Aquatex mounting agent (Merck, 108562).

Intestinal tissue was washed with PBS, cut open longitudinally and laid flat on a glass slide with the luminal side facing upward. The small intestinal villi were scrapped off the flat tissue by a glass slide and collected in cold 1 PBS. The remaining tissue containing crypts was chopped into <2mm pieces using a scalpel, washed three times with ice-cold 1 PBS and incubated in chelation medium (2mM EDTA in 1 PBS without Ca2+ and Mg2+, Gibco 10010023) for 40min with agitation at 4C. The digested tissue was shaken vigorously for 30s in 1 PBS to release crypts and villi. To separate out crypts and villi of the small intestine, the solution was passed through a 100m cell strainer. The isolated crypts in the flow through were pelleted and transferred to RLT Buffer (Qiagen, 79216). RNeasy microkit (Qiagen, 74004) was used for RNA extraction. Extracted RNAs were incubated with DNase1 (ThermoFisher, EN0521) at 37C for 30min, followed by a 10min incubation with EDTA at 65C. High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, 4368814) was used to generate complementary DNA from total RNA. Quantitative real-time-PCR (qRT-PCR) was performed on LightCycler96 (Roche) with mouse and naked mole rat Gapdh used as an endogenous control. The IDs of Taqman Gene expression assays (Applied Biosystems) used in this study are Gapdh (Mm99999915_g1, Hg05064520_gH), Muc2 (Mm01276681_m1, Hg05250665_g1), Synaptophysin (Mm00436850_m1, Hg05249763_m1), and Aldolase B (Mm00523293_m1, Hg05103981_m1). The 2-Ct method was used to calculate the relative gene expression levels.

Brightfield images of tissue sections were captured using an Olympus BX51 microscope coupled with an Olympus DP70 camera system using DP controller software. Villi were imaged using 10 objective while crypts were imaged with 20 (for colon) or 60 (for small intestine) objective lens. Histopathological scoring in this study was performed based on the digital images obtained on Hamamatsu (Nanozoomer HT) scanner at 40 magnification.

To quantify cell numbers in crypt-villous structures from brightfield images, cell counter plugin of Fiji software was used. The dimensions of crypt-villous structure were calculated using the measure tool in Fiji.

Fluorescent images of intestinal crypts were acquired from 4m thick tissue sections with a Plan Apochromat 63 or 100 1.4 oil objective on a Zeiss LSM 780 upright or inverted confocal microscope. Images were acquired in Zen SP7 FP3 (black) software using 405nm, 488nm, 561nm, and 633nm laser lines in sequential tracks. Z-stacks of 6-12 optical sections with 50% overlap between subsequent planes were captured within the span of a single cell at 0.3m z-distance, 0.087m pixel dimension, and 12-bit depth.

For generating the RGB images used in the figures (Figs.1a, b, 2ad, 3d, 4d, 7a, d, Supplementary Figs.14, 5d, e, 11b), the original.czi raw files were imported into Fiji software package and a maximum intensity z-projection was created from the stacks. Using the split channel option of Fiji, the multicolour fluorescent images were separated into individual channels (DAPI, Alexa 488, Cy3, Alexa 633). The maximum and minimum displayed pixel values of individual channels were adjusted across the entire image set including in negative controls (i.e. linear adjustment) to correct for autofluorescence that had been introduced in the image stacks during acquisition. Then, using merge channel option in Fiji, two/more channels were combined to create a composite image (Lgr5/Ki67 or LGR5/KI67, Lgr5/EpCAM or LGR5/EPCAM, Lgr5/p27 or LGR5/P27, Lgr5/BrdU, Lgr5/pHH3 or LGR5/PHH3) while keeping the individual channels intact. Finally, all the individual and composite images were converted into RGB color type and saved in TIFF format. These images (TIFF) were compiled in Adobe Illustrator 2020 software to produce the panels presented in the figures.

Z-stack images were processed in batch mode of Fiji package. Firstly, a maximum intensity projection was created to generate a 2D image from the stacks. Next, each channel of the image was separated, and maximum and minimum displayed pixel values were adjusted across the entire image set including negative controls. To quantify the number of rodent Lgr5 or human LGR5 mRNA expressed in a single cell, all the ISH dots were manually counted within the cell periphery demarcated by EpCAM staining. As the Lgr5 or LGR5 signal was captured using confocal microscopy at a resolution of 237nm, overlapping/merged Lgr5 or LGR5 mRNA signal dots were rarely observed. To calculate the distribution of Lgr5+ or LGR5+ cells relative to other cells along the crypt axis, the cell present at the crypt apex was assigned position 0 and we counted cells on each side of this cell to acquire datapoints in our quantifications. Any cell containing more than three Lgr5 or LGR5 mRNA puncta was considered positive for Lgr5 or LGR5 expression (Lgr5+ or LGR5+).

We observed significant variation in autofluorescence levels between mouse, human and NMR intestinal tissues, with mouse tissue emitting the most and naked mole rats the least. This variation necessitated adjusting the laser powers of the confocal microscope during image acquisition so that maximal image contrast was achieved while also reducing the autofluorescence signals. The maximum and minimum displayed pixel values of individual channels were adjusted across the entire image set (i.e. linear adjustment), including in negative controls, to correct for autofluorescence. These adjustments resulted in varying intensities for specific signals in the three species and, therefore, we took a binary approach for the quantification of the antibody-based signals. The presence of any specific signal in the target compartment inside a cell was considered positive regardless of the staining intensity.

We determined the length of the cell cycle (TT) and S-phase (TS) in CBC cells (Lgr5+CBC) of naked mole rats by counting the fraction of BrdU-labelled Lgr5+CBC cells after successive pulsing over 5 days in NMRs and 2.25 days in mice. As the CBC cells (Lgr5+CBC) cells are on average asynchronously and asymmetrically dividing45, the labelling index (LI) which provides the ratio of labelled cells to the total population (LI=Lgr5+CBCBrdU+/Lgr5+CBC) at any given time (t) can be modelled by Eq.1 below where TT is the total cell division time33.

$${{{{{rm{LI}}}}}}= (1/{{{{{rm{T}}}}}}_{{{{{rm{T}}}}}}){{{{{rm{X}}}}}}t+({{{{{rm{T}}}}}}_{{{{{rm{S}}}}}}/{{{{{rm{T}}}}}}_{{{{{rm{T}}}}}}),{{{{{rm{for}}}}}},{t}{{{{{rm{le }}}}}}{{{{{{rm{T}}}}}}}_{{{{{{rm{T}}}}}}}-{{{{{{rm{T}}}}}}}_{{{{{{rm{S}}}}}}}\ {{{{{rm{LI}}}}}}= 1,{{{{{rm{for}}}}}},t > {{{{{{rm{T}}}}}}}_{{{{{{rm{T}}}}}}}-{{{{{{rm{T}}}}}}}_{{{{{{rm{S}}}}}}}$$

(1)

Equation1 assumes that there are no or only very few stem cells (based on p27 negativity in NMR and mouse Lgr5+CBC cells) that remain quiescent for the duration of the BrdU experiment. The lfit tool in STATA was used to calculate the least square fit of the data by considering the time points before LI reached saturation. We derived TT from the slope of the regression (TT=1/slope). When t=0, LI0=TS/TT which is the y-intercept of the graph. Thus, the duration of S-phase (TS) was estimated from the y-intercept of the regression line.

For human LGR5+CBC cells, we assumed KI67 is undetectable at G1/S transition and detected in the S to M phases of the cell cycle46. We determined the fraction of LGR5+CBC cells that expressed KI67 and calculated the length of S, G2 and M-phase (T(S, G2, M)) using Eq.2:

$${{{{{{rm{T}}}}}}_{{({{{{{rm{S}}}}}},{{{{{rm{G}}}}}}2,{{{{{rm{M}}}}}})}}}{{{{{rm{KI}}}}}}67^{+}={{{{{{{rm{T}}}}}}}_{{{{{rm{T}}}}}}^{({{{{{{rm{Ref}}}}}}},31)}}{{{{{rm{X}}}}}},{{{{{{rm{LGR}}}}}}5}^{+{{{{{rm{CBC}}}}}}}{{{{{rm{KI}}}}}}67^{+}/{{LGR}5}^{+{{{{{rm{CBC}}}}}}}$$

(2)

The time in mitosis (TM) was calculated after quantifying the fraction of rodent (mouse or NMR) Lgr5+CBC or human LGR5+CBC cells positive for phospho-histone H3 using Eq.3:

$${{{{{{rm{T}}}}}}{{{{{rm{M}}}}}}}^{{{{{{rm{Ki}}}}}}67+}={{{{{{{rm{T}}}}}}}_{{{{{{rm{T}}}}}}}}^{({{{{{rm{linear}}}}}},{{{{{rm{regression}}}}}})}{{{{{rm{X}}}}}},{{Lgr}5}^{+{{{{{rm{CBC}}}}}}}{{{{{rm{pHH}}}}}}3+({{{{{rm{Ser}}}}}}28)/{{Lgr}5}^{+{{{{{rm{CBC}}}}}}}$$

(3)

or

$${{{{{{{rm{T}}}}}}}_{{{{{{rm{M}}}}}}}}^{{{{{{rm{KI}}}}}}67+}={{{{{{{rm{T}}}}}}}_{{{{{{rm{T}}}}}}}}{({{{{{rm{ref}}}}}}31)}{{{{{rm{X}}}}}},{{LGR}5}^{+{{{{{rm{CBC}}}}}}}{{{{{{rm{PHH}}}}}}3}^{+}({{{{{rm{Ser}}}}}}28)/{{LGR}5}^{+{{{{{rm{CBC}}}}}}}$$

Using TS estimated by Ishikawa et al.31 previously, the length of G2-phase (TG2) was calculated using Eq.4:

$${{{{{{{rm{T}}}}}}}_{{{{{{rm{G}}}}}}2}}^{{{{{{rm{KI}}}}}}67+}={{{{{{rm{T}}}}}}}_{({{{{{rm{S}}}}}},{{{{{rm{G}}}}}}2,{{{{{rm{M}}}}}})}{{{{{{rm{KI}}}}}}67}^{+}-left({{{{{{{rm{T}}}}}}}_{{{{{{rm{S}}}}}}}}^{{{{{{rm{KI}}}}}}67+}+{{{{{{{rm{T}}}}}}}_{{{{{{rm{M}}}}}}}}^{{{{{{rm{KI}}}}}}67+}right)$$

(4)

After quantifying the fraction of LGR5+CBC cells expressing P27, we calculated the time spent in G0 and G1 (T(G1, G0)P27+) using Eq.5:

$${{{{{{{rm{T}}}}}}}_{({{{{{rm{G}}}}}}1,{{{{{rm{G}}}}}}0)}}^{{{{{{rm{P}}}}}}27+}={{{{{{{rm{T}}}}}}}_{{{{{{rm{T}}}}}}}}{({{{{{rm{ref}}}}}}31)}{{{{{rm{X}}}}}},{{LGR}5}^{+{{{{{rm{CBC}}}}}}}{{{{{rm{P}}}}}}27+/{{LGR}5}^{+{{{{{rm{CBC}}}}}}}$$

(5)

We took the fraction of LGR5+P27+ cells in G0 phase (QF) from Ishikawa et al. 31 to calculate the length of G0 in human LGR5+CBC cells using Eq.6:

$${{{{{{{rm{T}}}}}}}_{{{{{{rm{G}}}}}}0}}^{{{{{{rm{P}}}}}}27+}={{{{{{rm{QF}}}}}}}{({{{{{rm{ref}}}}}}31)}{{{{{rm{X}}}}}},{{{{{{{rm{T}}}}}}}_{(G1,G0)}}^{{{{{{rm{P}}}}}}27+}$$

(6)

Finally, using Eq.7, we quantified the time human colonic LGR5+CBC cells spend in G1 (TG1):

$${{{{{{rm{T}}}}}}}_{{{{{{rm{T}}}}}}}={{{{{{{rm{T}}}}}}}_{{{{{{rm{G}}}}}}0}}^{{{{{{rm{P}}}}}}27+}+{{{{{{{rm{T}}}}}}}_{{{{{{rm{G}}}}}}1}}^{{{{{{rm{P}}}}}}27+}+{{{{{{{rm{T}}}}}}}_{{{{{{rm{S}}}}}}}}^{{{{{{rm{KI}}}}}}67+}+{{{{{{{rm{T}}}}}}}_{{{{{{rm{G}}}}}}2}}^{{{{{{rm{KI}}}}}}67+}+{{{{{{{rm{T}}}}}}}_{{{{{{rm{M}}}}}}}}^{{{{{{rm{KI}}}}}}67+}$$

(7)

In NMR and mouse, Lgr5+CBC cells are negative for p27 such that TG0=0. For these species, we derived the combined length of time spent in G1 and G2 (TG1+TG2) from Eq.7.

Using the length of TS from cumulative BrdU labelling in Lgr5+CBC cells and assuming no change in TS in Lgr5+ cells located at different positions within the crypt31, we measured the total cell division time (TT) of Lgr5+above crypt base cells using Eq.1 by measuring the labelling index (LI) at a single time point (t) after pulsing animals with BrdU in vivo. More specifically, in C57BL/6 mice (n=3 animals, 4 months old), we administered BrdU once and analysed intestinal tissue at t=0.5h. In NMRs (n=3 animals, 6-24 months-old), we pulsed the animals with BrdU every 8h and analysed the intestine after t=1 day.

We used Microsoft Excel (v16.77.1) for inputting raw data after collection. All statistical tests and graphs displayed in this paper were generated using StataMP 14.1. Details of statistical tests performed are described in figure legends. P-values are generated by conducting two-tailed t-tests, F-test and Wilcoxon rank sum test as indicated in each figure legend. No blinding and randomization were performed during the analysis.

All the figures presented in this manuscript were prepared using Adobe Illustrator 2020 (version 24.1). Vector line arts shown in Figs.1c, d, 3a, h, 4a, h, 6a, Supplementary Figs.5d, e, and 9a, b were created using the curvature tool of Adobe Illustrator.

Further information on research design is available in theNature Portfolio Reporting Summary linked to this article.

Read the original here:
Adult stem cell activity in naked mole rats for long-term tissue maintenance - Nature.com

Platelet-Rich Plasma Injections | What It’s Like Getting PRP Injections Into The Knee as a Skier – SnowBrains

In November 2021, I hiked up and skied Little Chute at Alta, UT.

When skiing down my knee swelled up and I could only bend it 90.

After arriving home the swelling went down.

This happened on a daily repeat cycle until February 2022 when the pain upon skiing became so intense I had to stop skiing altogether.

I talked to my surgeon.

He quickly diagnosed me with osteoarthritis caused by my knee surgery in 2015 to replace my ACL.

I had also vaporized my meniscus and ripped off a square centimeter of cartilage on the end of my femur resulting in a microfracture surgery (performed at the same time as my ACL replacement).

The first course of action was a Non-Steroidal Anti-Inflammatory Drug (NSAID) regimen (ibuprofen, etc).

That didnt work.

In February 2022, I had my first Platelet-Rich Plasma (PRP) injection.

I coupled the injection with 4 weeks off from skiing and physical therapy.

I returned to skiing in March 2022 and had a decent March, April, and May 2022.

I surfed great waves with no issues in the Maldives all of June and July (surfing is not hard on knees).

I returned to snow in August 2022 in Patagonia and on day #1 my knee swelled up and was painful again.

I was only able to ski 22 out of 60 days that summer

I went back in for another PRP injection in October.

This time it worked.

I had a strong 2022/23 ski season with record snowfall across the west and I skied 305 out of 365 days that year without issue, swelling, nor pain.

Since then, Ive officially drunk the Kool-Aid.

I found a doctor who will do PRP injections for $450 a pop and Im doing them every 3 months.

This week, I got my 5th PRP injection in the lateral compartment of my left knee (where there is no meniscus and no cartilage).

I plan on continuing with these PRP injections indefinitely.

In general, its advised to take it easy for a day or two after the injection.

I sometimes experience swelling for 24 hours after the PRP injection.

I believe that these injections are working for me and hopefully, theyll help me put off getting a knee replacement until Im at least 55.

Fingers crossed.

Im 45 years old, 61, 165lbs and Ive been skiing full time since I was 22 and Ive skied year round the last 13 years.

PRP treatment is not yet fully proven by science and therefore, health insurance generally wont cover it.

I also do a lot of physical therapy (building up the muscles in my legs) and I have a custom-made DonJoy knee brace that I wear anytime I ski that is called an unloader knee brace because it bends my knee bowlegged taking pressure off the lateral compartment of my left knee where I lack cartilage.

Unloader knee braces are also not well-proven in science.

You may have to get an MRI before you can get into a doctor to get PRP injections and the doctor you see (orthopedic surgeon) will also most likely take an x-ray of your knees.

All info below from Johns Hopkins University

Platelet-rich plasma consists of two elements: plasma, or the liquid portion of blood, and platelets, a type of blood cell that plays an important role in healing throughout the body. Platelets are well-known for their clotting abilities, but they also contain growth factors that can trigger cell reproduction and stimulate tissue regeneration or healing in the treated area. Platelet-rich plasma is simply blood that contains more platelets than normal.

To create platelet-rich plasma, clinicians take a blood sample from the patient and place it into a device called a centrifuge that rapidly spins the sample, separating out the other components of the blood from the platelets and concentrating them within the plasma.

After creating platelet-rich plasma from a patients blood sample, that solution is injected into the target area, such as an injured knee or a tendon. In some cases, the clinician may use ultrasound to guide the injection. The idea is to increase the concentration of specific bioproteins or hormones, called growth factors, in a specific area to accelerate the healing process.

The mechanism behind PRP injections is not completely understood. Studies show that the increased concentration of growth factors in platelet-rich plasma may stimulate or speed up the healing process, shortening healing time for injuries, decreasing pain, and even encouraging hair growth.

PRP injections are used for a range of conditions,* from musculoskeletal pain and injuries to cosmetic procedures.

Tendon, Ligament, Muscle and Joint Injuries

PRP injections may be able to treat a range of musculoskeletal injuries and conditions. For example, chronic tendon injuries such as tennis elbow or jumpers knee can often take a long time to heal, so adding PRP shots to a treatment regimen can help to stimulate the healing process, decrease pain, and enable a return to activities sooner.

Post-surgical Healing

Clinicians first used PRP to accelerate healing after jaw or plastic surgeries. Now, post-surgical PRP injections have expanded to help heal muscles, tendons, and ligaments, as procedures on these tissues have notoriously long recovery times.

Osteoarthritis

Early studies indicate that PRP injections may help treat osteoarthritis pain and stiffness by modulating the joint environment and reducing inflammation, but research is growing.

Hair Loss

PRP injections can be effective in treating male pattern baldness, both in preventing hair loss and promoting new hair growth. PRP can also aid in the stimulation of hair growth after hair transplants.

Skin Rejuvenation

PRP injections are sometimes used as an anti-aging treatment, but there is little evidence to show that PRP reduces wrinkles and other signs of aging.

PRP Therapy Risks and Side Effects

A PRP injection is a low-risk procedure and does not usually cause major side effects. The procedure involves a blood draw, so you should make sure you are hydrated and have eaten beforehand to prevent feeling lightheaded. After the procedure, you may experience some soreness and bruising at the injection site.

Because PRP injections are made up of your own cells and plasma, the risk of an allergic reaction is much lower than with other injectable medications like corticosteroids. Less common risks of PRP injections include:

If you are considering PRP injections, be sure to talk with your healthcare provider about all the benefits and risks.

View post:
Platelet-Rich Plasma Injections | What It's Like Getting PRP Injections Into The Knee as a Skier - SnowBrains

Half of pediatric patients with aHUS benefit from Soliris after… – AHUS News

Treatment withSoliris (eculizumab) helped about half of the children diagnosed with atypical hemolytic uremic syndrome (aHUS) after they received a stem cell transplant, according to a small study.

Among 13 patients who started taking Soliris, seven survived and saw their disease-associated biomarkers normalize. Six died due to complications related to the stem cell transplant.

The study, Eculizumab treatment in paediatric patients diagnosed with aHUS after haematopoietic stem cell transplantation: a HSCT-TMA case series from Japanese aHUS post-marketing surveillance, was published in Bone Marrow Transplantation. The analysis was sponsored by Alexion, now a part of AstraZeneca Rare Disease, which developed Soliris.

aHUS is a type of thrombotic microangiopathy (TMA), a group of diseases wherein blood clots form in small blood vessels. In aHUS, this is caused by abnormal activity of the immune systems complement cascade. While there are often genetic factors, another trigger is typically needed for symptoms to manifest.

One such trigger can be a hematopoietic stem cell transplant (HSCT), which is used to treat a range of blood and autoimmune disorders. It involves transplanting blood cell precursors into a patient to help repopulate their body with healthy blood cells.

Certain immune complications can occur that drive TMA symptoms, however, especially in people with underlying genetic risk factors. TMA is associated with high mortality rates after a stem cell transplant and the appropriate treatment strategy remains to be established. Soliris is approved for aHUS and other complement-mediated diseases, and inhibits the C5 protein to prevent the complement cascades activation, potentially making it an appropriate treatment for managing HSCT-TMA.

Here, scientists retrospectively analyzed clinical data from 13 pediatric patients in Japan who received Soliris after being diagnosed with aHUS following an HSCT procedure, whichwas intended to treat various forms of cancer or immune system diseases. TMA symptoms emerged about a month later, on average.

Three children had existing risk factors for aHUS, including a history or family history of TMA. Eleven patients had complications from the procedure that could have caused the complement cascade to overact, according to the scientists.

All the children were treated with other therapies before starting Soliris, which was initiated a median of 16 days after the onset of TMA. Soliris was infused into the vein, or intravenously, in a similar regimen as that approved for aHUS, with patients receiving a median of three doses.

Over a median observation period of nearly six months, seven patients survived, leading to a predicted survival of 53.8% six months after the onset of HSCT-TMA.

Among the survivors, Soliris decreased levels of lactate dehydrogenase, a marker of organ damage, after 22 days. It also increased the count of platelets, blood clotting cells that are lost as a cardinal aHUS symptom, after a median of 280 days (about nine months).

Median blood levels of creatinine, a marker of kidney damage, didnt change, but four of the seven survivors did see improvements. Three children who previously required dialysis, a blood cleaning procedure for when the kidneys are failing, discontinued it by the last follow-up visit.

None of the survivors had a TMA recurrence over a median of 111.5 days (around 3.6 months) after stopping Soliris.

Three patients died due to ongoing side effects that started before Soliris was initiated and two died due to infection-related side effects that arose after immunosuppressive therapy to treat an HSCT-associated immune complication. One patient didnt respond well to Soliris and died due to TMA.

The survival rates in those treated with Soliris after HSCT-TMA were lower than in a previous report. That could be because in that study, Soliris was started early as a first-line treatment and at individualized dosing regimens.

Early treatment with complement inhibitor and early evaluation of treatment response can be a preferential treatment strategy once complement dysfunction is suspected in HSCT-TMA, the researchers wrote.

No significant prognostic risk factors were identified by comparing survivors with non-survivors, which could be attributed in part to the small nature of the study.

Further research into the risk stratification of HSCT-TMA and the use of C5 inhibitors are needed to confirm appropriate use in HSCT-TMA and to identify factors that might predict patients responses to therapy, the researchers said.

See the rest here:
Half of pediatric patients with aHUS benefit from Soliris after... - AHUS News

Three Win Grant for Pediatric AML NK Cell Therapy Trial – Mirage News

Fehniger (left), Bednarski and Pfeiffer

Todd A. Fehniger, MD, PhD, a professor of medicine, and co-principal investigators Jeffrey J. Bednarski, MD, PhD, an associate professor of pediatrics, and Thomas Pfeiffer, MD, an assistant professor of pediatrics, all at Washington University School of Medicine in St. Louis, have received a total of $1.4 million from the Leukemia & Lymphoma Society, the Rising Tide Foundation for Clinical Cancer Research and Siteman Kids. The funding will support a phase 2 clinical trial of a novel cell-based immunotherapy used in conjunction with a stem cell transplant. Developed at Washington University, the immunotherapy is for children with acute myeloid leukemia (AML), a type of blood cancer.

With the goal of reducing relapse after stem cell transplantation, the researchers will treat patients with memory-like natural killer (NK) cells, a type of immune cell, that have been exposed to a protein cocktail that reprograms them to better identify and eliminate cancer cells. The memory-like NK cells will come from the same donor who provided the patient's stem cells and be given about one week after a patient's stem cell transplant.

See the rest here:
Three Win Grant for Pediatric AML NK Cell Therapy Trial - Mirage News

Promising new sickle cell therapy trialed at Children’s Hospital of Richmond at VCU and VCU Medical Center – VCU Health

By Kate Marino

The U.S. Food and Drug Administration recently approved two gene therapies to treat sickle cell disease in people ages 12 and older a gigantic step toward curing this often-debilitating disease. Children's Hospital of Richmond at VCU and VCU Medical Center were involved in the clinical trial that led to the approval of these products.

India Sisler, M.D., is interim division chief and clinical director of the Division of Hematology and Oncology and medical director of the pediatric comprehensive sickle cell center at CHoR. She explains how the new therapy works and why its approval is monumental.

Using a process called peripheral blood stem cell collection, we isolate and pull out the patients stem cells. We then send those cells to a company that specializes in gene modification so they can change the way those cells produce hemoglobin. We work to knock the patients bone marrow out completely and give them their own stem cells back with the modified hemoglobin gene.

This is a multi-step process that involves meticulous planning and special care for the patient along the way.

While treatment is only currently available to this certain subset of patients, our hope is that it will become more widely available in the near future. Clinical trials in children under 12 are still in process.

As we see in many patients, as Walter got older he was having a lot of complications and pain, which made daily life difficult. What made him a great candidate was his motivation to improve his health in order to be the social, active young man he wanted to be. He was all-in and hes doing so well now. Every time we see him, hes almost in tears telling us the things hes able to do and the new lease on life he feels like he has.

At this point Walter is done with treatment and cured of sickle cell. He will follow up with our transplant team so we can monitor him for any side effects every couple months for now, then well space out to a year. We dont really know what to expect 20 years from now since this a new treatment, but its so promising and were thrilled to see Walter thriving.

Beth Krieger, M.D, a pediatric hematology and oncology specialist at CHoR, conducted advanced fellowship training with leading experts in the field of bone marrow transplant and cellular therapies at University of Minnesota, where she did a significant amount of transplant work for non-cancerous conditions. Her knowledge and expertise have been instrumental in Walters care during this study and in bringing curative therapy to CHoR.

Our sickle cell teams work hand-in-hand on a daily basis to help patients transition seamlessly from pediatric to adult care, so were accustomed to collaborating and know it is beneficial for everyone.

Two different sickle cell gene therapy products were actually FDA approved the one we trialed and another one and we are working to begin offering both of them here. Our hope is also that many more patients will qualify in the near future and that we can begin curing people of sickle cell disease without them having to experience the extreme pain and other side effects of vaso-occlusive crises.

Similar gene therapy has also been FDA approved for beta thalassemia, another red blood cell disease. Its much less common than sickle cell, but we have patients undergoing this clinical treatment and it has the potential to be equally life changing.

Go here to see the original:
Promising new sickle cell therapy trialed at Children's Hospital of Richmond at VCU and VCU Medical Center - VCU Health

Scientists Grew ‘Mini Brains’ From Stem Cells. Then, The Brains Sort-of Developed Eyes. – ScienceAlert

Mini brains grown in a lab from stem cells spontaneously developed rudimentary eye structures, scientists reported in a fascinating paper in 2021.

On tiny, human-derived brain organoids grown in dishes, two bilaterally symmetrical optic cups were seen to grow, mirroring the development of eye structures in human embryos.

This incredible result will help us to better understand the process of eye differentiation and development, as well as eye diseases.

"Our work highlights the remarkable ability of brain organoids to generate primitive sensory structures that are light sensitive and harbor cell types similar to those found in the body," said neuroscientist Jay Gopalakrishnan of University Hospital Dusseldorf in Germany.

"These organoids can help to study brain-eye interactions during embryo development, model congenital retinal disorders, and generate patient-specific retinal cell types for personalized drug testing and transplantation therapies."

Brain organoids are not true brains, as you might be thinking of them. They are small, three-dimensional structures grown from induced pluripotent stem cells - cells harvested from adult humans and reverse engineered into stem cells, that have the potential to grow into many different types of tissue.

In this case, these stem cells are coaxed to grow into blobs of brain tissue, without anything resembling thoughts, emotions, or consciousness.

Such 'mini brains' are used for research purposes where using actual living brains would be impossible, or at the very least, ethically tricky - testing drug responses, for example, or observing cell development under certain adverse conditions.

This time, Gopalakrishnan and his colleagues were seeking to observe eye development.

In previous research, other scientists had used embryonic stem cells to grow optic cups, the structures that develop into almost the entire globe of the eye during embryonic development. And other research had developed optic cup-like structures from induced pluripotent stem cells.

Rather than grow these structures directly, Gopalakrishnan's team wanted to see if they could be grown as an integrated part of brain organoids. This would add the benefit of seeing how the two types of tissue can grow together, rather than just growing optic structures in isolation.

"Eye development is a complex process, and understanding it could allow underpinning the molecular basis of early retinal diseases," the researchers wrote in their paper.

"Thus, it is crucial to study optic vesicles that are the primordium of the eye whose proximal end is attached to the forebrain, essential for proper eye formation."

Previous work in the development of organoids showed evidence of retinal cells, but these did not develop optic structures, so the team changed their protocols. They didn't attempt to force the development of purely neural cells at the early stages of neural differentiation, and added retinol acetate to the culture medium as an aid to eye development.

Their carefully tended mini brains formed optic cups as early as 30 days into development, with the structures clearly visible at 50 days. This is consistent with the timing of eye development in the human embryo, which means these organoids could be useful for studying the intricacies of this process.

There are other implications, too. The optic cups contained different retinal cell types, which organized into neural networks that responded to light, and even contained lens and corneal tissue. Finally, the structures displayed retinal connectivity to regions of the brain tissue.

"In the mammalian brain, nerve fibers of retinal ganglion cells reach out to connect with their brain targets, an aspect that has never before been shown in an in vitro system," Gopalakrishnan said.

And it's reproducible. Of the 314 brain organoids the team grew, 73 percent developed optic cups. The team hopes to develop strategies for keeping these structures viable on longer time-scales for performing more in-depth research with huge potential, the researchers said.

"Optic vesicle-containing brain organoids displaying highly specialized neuronal cell types can be developed, paving the way to generate personalized organoids and retinal pigment epithelial sheets for transplantation," they wrote in their paper.

"We believe that [these] are next-generation organoids helping to model retinopathies that emerge from early neurodevelopmental disorders."

The research has been published in Cell Stem Cell.

A version of this article was first published in August 2021.

Follow this link:
Scientists Grew 'Mini Brains' From Stem Cells. Then, The Brains Sort-of Developed Eyes. - ScienceAlert

Candel Therapeutics Receives FDA Fast Track Designation for CAN-2409 in Pancreatic Cancer

NEEDHAM, Mass., Dec. 12, 2023 (GLOBE NEWSWIRE) -- Candel Therapeutics, Inc. (Candel or the Company) (Nasdaq: CADL), a clinical stage biopharmaceutical company focused on developing multimodal biological immunotherapies to help patients fight cancer, today announced that the U.S. Food and Drug Administration (FDA) granted Fast Track Designation for its lead investigational adenovirus asset CAN-2409 plus prodrug (valacyclovir) for the treatment of patients with pancreatic ductal adenocarcinoma (PDAC) to improve overall survival.

More here:
Candel Therapeutics Receives FDA Fast Track Designation for CAN-2409 in Pancreatic Cancer

electroCore Expands Intellectual Property Portfolio for Nerve Stimulation Technology

ROCKAWAY, N.J., Dec. 12, 2023 (GLOBE NEWSWIRE) -- electroCore, Inc. (Nasdaq: ECOR), a commercial-stage bioelectronic medicine and wellness company, today announced the United States Patent and Trademark Office has issued a patent related to a system for stimulating a nerve target in the head or neck of a patient for treatment of various disorders, such as primary headache.

Continue reading here:
electroCore Expands Intellectual Property Portfolio for Nerve Stimulation Technology