Embryo Patrol. Artificial Embryos Are Not Human Babies | by Karen Marie Shelton | ILLUMINATION-Curated | Jan, 2024 – Medium

Artificial Embryos Are Not Human Babies Didactic Model of Human Embryonic Development Wagner Souza Esilva Wikimedia

Artificial embryos are not human. Theyre simply a cluster of cells. To be legally human, they must meet the definition of an in vitro fertilized human ovum.

As of the end of 2023, artificial embryos couldnt be successfully implanted into mammals or humans. They couldnt lead to pregnancies, and there is no plan for that to happen in the future.

Synthetic embryos utilize stem cells groundbreakingly, sidestepping the need for sperm or eggs. Ongoing breakthroughs might eventually aid research into genetic disorders and improve babies health, including reducing the risk of problem pregnancies and miscarriages.

Artificial embryos are not related to in vitro fertilization (IVF), which can lead to a human pregnancy.

The term is misleading. These structures arent really synthetic, nor are they exactly embryos. But theyre similar. They are tiny balls of cells arising from a sperm fertilizing an egg but created from stem cells grown in the lab.

Synthetic human embryos, or SHEEFs (synthetic human entities with embryo-like features), are created from very early (actually pre-embryonic) zygotic cells called stem cells.

The stem cells are called pluripotent because they have the potential to develop into almost every cell of the body.

The lab-created embryos are not connected to a beating heart or a brain. They do include cells that would typically go on to form a version of a placenta, yolk sac, and embryo itself.

The model embryos, which resemble human versions, recreate the earliest stages of human development. They could provide a crucial window into genetic disorders and the underlying biological causes of recurrent miscarriage.

Robin Lovell-Badge, headent head of stem cell biology and developmental genetics at Francis Crick Institute in London, reported project advancements. She explained weve cultivated embryos to a specific stage just beyond what is equivalent to 14 days of development for a natural embryo.

View original post here:
Embryo Patrol. Artificial Embryos Are Not Human Babies | by Karen Marie Shelton | ILLUMINATION-Curated | Jan, 2024 - Medium

Enhancing Stem Cell Tracking with Nanoparticle Imaging Agents – AZoNano

Sponsored by MerckJan 23 2024Reviewed by Emily Magee

Stem cell therapies show promise in tissue engineering, regenerative medicine, and their homing effect. They offer hope for treating numerous incurable diseases like Parkinsons disease, liver failure, ischemic heart disease, and cancer.

However, understanding the cell distribution, migration behaviors, and functionality post-engraftment remains challenging due to the absence of effective in vivo tracing methods. This lack impedes the clinical progress of stem cell therapies.

To overcome this hurdle, it is crucial to develop advanced imaging strategies that can label and track transplanted stem cells without disrupting their normal functions. These strategies are necessary to uncover therapeutic mechanisms and assess safety before clinical trials.

Imaging techniques suited for stem cells should prioritize non-toxicity, high resolution, longevity, and the dynamic provision of cell fate information. Over recent years, several bioimaging approaches have emerged.

One particularly promising tactic involves leveraging well-designed nanoparticles as contrast agents to monitor stem cells. These nanoparticles boast unique physicochemical properties and offer versatility and customizability.

However, existing imaging methods still fall short of meeting all the necessary in vivo stem cell tracking demands.

This article provides a concise examination of the four commonly utilized nanoparticle-based imaging techniques for the tracking of stem cells: magnetic resonance imaging (MRI), fluorescence imaging, ultrasound imaging (USI), and photoacoustic imaging (PAI) (Figure 1).

Emphasis is placed on the design of contrast nanoagents, the corresponding imaging mechanisms, and the specific challenges these nanoagents have addressed.

The objective is to illuminate potential pathways for future advancements in contrast agent development, aiming to create more sophisticated solutions for the in vivo tracking of transplanted stem cells.

Figure 1.Various contract nanoagents designed for magnetic resonance imaging, fluorescence imaging, ultrasound imaging, and photoacoustic imaging modalities in stem cell trackingin vivo.Image Credit:Merck

The preferred method for in vivo stem cell imaging is MRI, owing to its distinctive attributes such as unrestricted penetration depth, exceptional spatial (ranging from 40 to 100 m), and temporal (ranging from minutes to hours) resolution, and safe operational nature.1

MRI operates in two main categories: T1/T1* and T2/T2*-weighted MRI. These categories are determined by the relaxation time of the longitudinal (T1) and transverse (T2) components of the magnetization vector towards equilibrium below the applied magnetic field.

MRI contrast agents, such as gadolinium (III) chelates and superparamagnetic iron oxide nanoparticles (SIONPs), are introduced to heighten resolution and amplify signal intensity by influencing the relaxation times of nearby water protons.2

Gadolinium (III) chelates are efficient T1 contrast agents as they accelerate the longitudinal relaxation rate (T1), thereby enhancing positive contrast in T1-weighted MRI sequences.

SIONPs function as T2 contrast agents and have found widespread application in MRI-based cell tracking by producing negative contrast through the reduction of T2/T2* relaxation times.

The subsequent section reviews nanoparticle-based contrast agents that have demonstrated clinical or commercial validation for their utility in stem cell tracking applications.

Nanoparticle-based stem cell labeling agents necessitate specific criteria: colloidal stability, non-toxicity, strong magnetism, and efficient labeling. Achieving colloidal stability in aqueous solutions typically involves utilizing hydrophilic polymer coatings like chitosan, dextran, and PEG, among others.

A preclinical study by Margarita Gutova et al. demonstrated the transplantation of neural stem cells (NSCs) labeled with dextran-coated ferumoxytol (FDA-approved SIONPs) into patients with brain tumors.3 The distribution of NSCs was consistently monitored over 12 weeks at various intervals, followed by surveillance serial MRI scans.3

Enhanced labeling efficiency can be achieved by modifying particle surfaces to carry a positive charge or specific ligands.

For instance, compared to ferumoxytol, self-assembling ferumoxytol nano-complexes altered with heparin and protamine sulfatewhich reversed the original particle's negative chargeexhibited increased labeling efficiency and a threefold rise in T2 relaxivity.

This modification approach demonstrated in vivo MRI detection of a minimum of 1000 HPF-labeled cells implanted within rat brains.4

Clinically approved Gd-DTPA enclosed within cationic liposomes ensured highly efficient uptake of Gd and exceptional intracellular retention in mesenchymal stem cells.

The Gd-DTPA-liposomes complex contrast agent rendered 500000 labeled stem cells distinctly visible for a minimum of two weeks on a 3.0 T clinical scanner, effectively overcoming the relatively low MRI sensitivity of Gd-DTPA.5

Monitoring the migration and survival of stem cells is crucial for both therapeutic efficacy and safety evaluations.

For investigating stem cell migration, Lili Jiang et al. observed that T2*-weighted MRI successfully tracked the migration of implanted pluripotent stem cells labeled with SIONPs (from Merck KGaA, Darmstadt, Germany) from the injection site to the injured brain areas for more than four weeks.6

In assessing cell survival, Ashok J. Theruvath et al. observed significant changes in ferumoxytol-labeled apoptotic matrix-associated stem cell implants (MASIs), indicating a substantial loss of iron signals and an extended T2 relaxation time, which persisted up to two weeks post-implantation during a cartilage repair process.7

When coupled with histopathologic examination, a ferumoxytol-based contrast agent could serve as an indicator distinguishing between living and deceased stem cells.

The duration for tracking stem cells varies depending on specific tissues, organs, and diseases, spanning from several days to months. An effective strategy for in vivo stem cell tracking involves evaluating cell distribution, migration, and differentiation, as well as assessing the efficacy and safety of cell implantation.

Despite the rapid advancements in nanoparticle-based MRI for stem cell tracking, certain challenges persist. These include imaging the differentiation and functionality of stem cells, integrating MRI contrast agents with reporter genes, and combining MRI with other noninvasive imaging tools.

Fluorescence imaging, a traditional optical imaging method, is affordable and highly sensitive, but its effectiveness is limited by poor tissue penetration (<1 cm) in comparison to MRI technology.

One potential solution involves fine-tuning fluorophores or fluorescent proteins to be responsive within the near-infrared (NIR) range, enabling deeper tissue penetration. However, challenges like severe photo-bleaching and light scattering restrict tracking efficiency and duration.8

The evolution of fluorescent nanoparticles (NPs) has significantly enhanced the capability of fluorescent imaging for long-term stem cell tracing in vivo.

Quantum dots (QDs), a set of classic inorganic semiconductor NPs used for cell labeling,9 rely on the transition behaviors of excited electrons across various energy levels. These transitions can be adjusted by manipulating material components or compositions, enabling the realization of NIR emission.

For instance, CdSe/ZnS core/shell structures were developed specifically to label and track adipose tissue-derived stem cells (ASCs) in C57BL/6 mice models using NIR emission.10

Modifying the bandgap of materials significantly enhances the optical properties of QDs.11

Wang et al. coupled NIR-II fluorescence QDs of Ag2S with the traditional bioluminescence red firefly luciferase (RFLuc) to label human mesenchymal stem cells (hMSCs).

Using a wide spectrum spanning from 400 to 1700 nm, the researchers investigated the dynamic tracking of survival and osteogenic differentiation of the transplanted hMSCs in a mouse model with calvarial defects.12

Up-conversion nanoparticles (UCNPs) have also been employed to label and track mouse MSCs. UCNPs utilize the anti-Stokes process, absorbing multiple NIR photons to generate a single short-wavelength photon.13,14

Despite this, rare earth metal ions doped in UCNPs and heavy metal ions in QDs pose potential safety risks for tracing clinical stem cells. Aggregation-induced emission (AIE) fluorophores offer a secure method to track stem cells, boasting excellent photostability in comparison to inorganic NPs.15-18

AIE fluorophores remain non-emissive when dispersed but emit robust fluorescence when in an aggregated state due to limited intramolecular rotation, unlike aggregation caused by quenching (ACQ). This unique property makes Dots or AIE NPs present enduring and robust fluorescent signals.

For example, AIE Dots derived from tetraphenyl ethylene have demonstrated the ability to trace the journey of adipose-derived stem cells (ADSCs) over an extended period, outperforming green fluorescent proteins (FPs) and other bioluminescent molecules.18

It is also easy to adjust the AIE monomer for NIR or even NIR-II emission. The advancement of AIE Dots is an encouraging option for in vivo stem cell tracking, offering prolonged stability and reliability.

Due to its remarkable temporal and spatial resolution and substantial tissue penetration depth, ultrasound imaging stands as a potent method for noninvasive and long-term cell tracking in stem cell therapies.19

However, its effectiveness is hindered by the limited contrast between implanted cells and neighboring soft tissues. To address this, ultrasound contrast agents (UCAs), a type of echogenic material, are deployed in clinical settings to enhance contrast and amplify detection signals.20

Traditional UCAs are micro-sized gas-filled bubbles, made of a bioinert heavy gas enveloped by stabilizing shells like proteins, lipids, and biocompatible polymers.21 The potential of these microbubbles for tracking stem cells is curbed by their inadequate structural stability, big microscale size, and short half-life.

Recent research has directed its attention toward scaling down UCAs, leading to the development of nanoscale UCAs such as nanobubbles, silica nanoparticles, and nanotubes tailored for ultrasound imaging.22

Achieving stability in the shell structure is crucial for nanobubbles. Leon et al. described a highly stable nanobubble type by employing a bilayer shell design with varying elastic properties akin to bacterial cell envelopes.23

These ultrastable nanobubbles exhibit minimal signal loss under continuous ultrasound exposure in vitro and boast a prolonged lifespan when tested in vivo. However, due to their small size, nanobubbles might lack sufficient ability to efficiently scatter ultrasonic waves.

To address this, a "small to large" transformation approach has been proposed. This strategy involves nanoscale UCAs transforming into microbubbles upon exposure to ultrasound, thereby amplifying the resulting echo signals.24 Gas-generating nanoparticles exemplify this concept.25,26

Min et al. reported a carbonate copolymer nanoparticle featuring a distinctive gas-generating mechanism. These nanoparticles undergo hydrolysis, yielding microscale CO2 bubbles that effectively absorb ultrasonic energy.27

Silica nanoparticles and similar glass-based nanomaterials significantly boost ultrasound signals due to tissue irregularities caused by the rigid characteristics of these nanoparticles, creating high impedance differences at tissue interfaces.28

Chen et al. detailed an innovative exosome-like silica (ELS) nanoparticle with high ultrasound impedance mismatches designed specifically for stem cell labeling and tracking using ultrasound.29

The researchers found that the nanoparticles' discoid shape, coupled with a positive charge, facilitated cellular uptake, and inherently heightened echogenicity. Silica nanoparticles with varied structures, like hollow and mesoporous configurations, also exhibited substantial ultrasound contrast.30

These silica-based nanoparticles show promise as UCAs for real-time stem cell tracking through ultrasound imaging due to their relatively robust structural integrity, low toxicity, and adaptable size and structure.

Biosafety and precise imaging are crucial for tracking cells in vivo over extended periods. PAI emerges as a promising biomedical imaging technique, based on the principle of the photoacoustic effect.

When a pulsed laser is introduced, it generates heat. The intermittent heat results in expansion, which is identified by PAI as a mechanical wave. This imaging method couples the strong contrast of optical imaging with the deep tissue penetration capabilities of ultrasonic imaging, offering real-time and high-resolution data in vivo.

PAI contrast nano agents are extensively used in vivo for imaging stem cells due to their exceptional capacity for photothermal conversion and the nanoparticles' biocompatibility

Various PAI contrast nano agents have been developed to absorb light across a spectrum, stretching from visible light to the NIR II region. This aims to minimize light absorbance and scattering within tissues.

Tracking methods that solely provide physical information, such as the whereabouts of labeled stem cells, do not meet the demands of studying stem cell engraftment. There is a pressing need for new platforms that can indicate real-time cell function and viability, crucial for clinical applications.

By focusing on the design of contrast nanoagents, new studies of PAI-based stem cell tracking were outlined in the following aspects: shape, component, size, and surface modification, shedding light on the essential factors that require consideration in the design of PAI contrast agents (Table 1).

Gold nanoparticles stand out among the various PAI contrast agents due to their notable advantages, including outstanding photothermal conversion efficiency, stable imaging capabilities, and high biosafety. The inert feature of gold nanoparticles has little effect on cell function.32

The Suggs group employed gold nanospheres of different sizes (20 nm, 40 nm, and 60 nm) in vivo to label and monitor MSCs.33

This study demonstrated the possibility of loading gold nanoparticles into MSCs for imaging purposes while preserving cell functionality post-loading. The research showed that gold nanoparticles remained detectable within stem cells for up to fourteen days, highlighting the potential for long-term and noninvasive cell tracking via PAI.

Anisotropic gold nanoparticles exhibit superior photothermal conversion efficiency. Jokerst et al. highlighted that applying a silica coating notably boosted the uptake rate of gold nanorods into stem cells by up to five times, achieving a minimum detection limit of 100000 labeled cells in vivo.34

The intricate transplant microenvironment in vivo poses challenges. Ricles et al. utilized a dual gold nanoparticle system, comprising gold nanorods and gold nanospheres, to track transplanted stem cells and image infiltrated macrophages.35

These studies introduced a novel approach to differentiate delivered stem cells from infiltrating immune cells, offering insights into the mechanisms of injury healing. Researchers also combined PAI with other imaging techniques to enhance tracking accuracy.

Nam et al. merged PAI with ultrasound imaging to monitor MSCs labeled with citrate-stabilized gold nanospheres.36 This combined approach offered both morphological insights through ultrasound imaging and functional information via photoacoustic imaging, allowing for spatial visualization of labeled MSCs.

Qiao et al. employed magnetic resonance and PAI techniques to track iron oxide (IO)@Au core-shell structure-labeled MSCs for imaging brain tumors.

This system exhibited potential for mapping cell trajectories and visualizing stem cell migration toward brain tumors in real time.37 Additionally, nanoparticles with NIR absorption capabilities were developed for stem cell tracking via PAI due to the superior penetration depth of NIR light.

Kim et al. utilized Prussian blue nanoparticles with robust light absorption at 740 nm for tracking stem cells.38

In vivo, these nanoagents demonstrated detection limits of 200 cells/L, enabling monitoring for up to fourteen days post-injection, attributed to the excellent bio-stability and NIR-I detectability. Yin et al. developed a NIR-II organic semiconductor polymer specifically for stem cell tracking via PAI.39

The researchers observed a significant 40.6-fold and 21.7-fold enhancement in subcutaneous and brain imaging, respectively, primarily due to the deep tissue penetration of NIR-II light.

To monitor stem cell activity post-transplantation, Dhada et al. engineered a ROS-sensitive dye (R775c) coated onto gold nanorods.

The strategy leveraged the fact that stem cells release ROS to degrade dying cells in vivo, enabling the measurement of stem cell viability. Consequently, they could simultaneously visualize cell viability and location in vivo using PAI.40

Table 1.The design of nanoparticle-based contrast agents for PAI.Source:Merck

Table 2. Comparison of different imaging modalities. Source:Merck

Nano agents offer key advantages over organic molecules in PAI imaging, primarily in terms of photostability, water solubility, and biocompatibility.

The limitations of PAI hinge on the penetration depth and photothermal conversion efficiency of contrast nano agents. Therefore, future advancements in PAI imaging are likely to focus on developing nanomaterials with enhanced NIR-II responsiveness and improved biocompatibility.

Advancing stem cell-based therapies heavily relies on the progress of stem cell tracking techniques. Current imaging technologies present both strengths and limitations (Table 2).

Consequently, the choice of imaging modalities depends on specific tissue requirements and imaging depth.

A range of nanoparticles, including iron oxide nanoparticles, quantum dots, aggregative-induced emission nanoparticles, silica nanoparticles, and gold nanoparticles, have been employed for stem cell tracing due to their distinct physical-chemical properties. These properties enhance imaging signals upon irradiation.

By elaborately designing the structures and compositions, nanoparticles play a pivotal role in improving the performance of existing imaging modalities concerning imaging resolution, stability, and lifespan. However, tracking stem cells in vivo remains a significant challenge in the biomedical field.

The amplitude of the image signal typically correlates linearly with contrast nanoagent concentration, but as contrast agents within cells dilute through cell division, challenges arise. This fuels the necessity for a new generation of contrast agents that resist dilution or prompt cells to self-synthesize these agents, addressing this issue.

Additionally, apart from cell division, studies indicate that nanoparticle levels within cells achieve equilibrium via endocytosis and exocytosis, potentially leading to false positive detections in vivo.

Moreover, few contrast agents can capture a cell's live state and normal functional abilities, severely limiting the exploration of stem cell therapy mechanisms. Therefore, concerted efforts are crucial to design multifunctional contrast agents that address these concerns.

Lastly, the advancement of both imaging devices (the "hardware") and imaging contrast agents (the "software") is urgently needed. This development aims to enable precise, long-term, and dynamic tracking of transplanted stem cells, a critical aspect of the field.

This information has been sourced, reviewed, and adapted from materials provided by Merck.

For more information on this source, please visit Merck.

Read the original here:
Enhancing Stem Cell Tracking with Nanoparticle Imaging Agents - AZoNano

1st-of-its-kind therapy blocks immune attack after stem-cell transplant – Livescience.com

A new treatment may be able to prevent a common immune-related complication of lifesaving bone-marrow transplants, a midstage clinical trial shows.

In patients with blood cancer, high doses of chemotherapy or radiation therapy are used to kill the cancerous cells, but these treatments also damage the patients' healthy, blood-forming stem cells. To rectify this, doctors may perform a type of bone-marrow transplant known as an allogeneic hematopoietic stem cell transplant (HSCT), in which blood-forming stem cells from a healthy donor are transplanted into the cancer patient.

But there's a potential catch: Immune cells in the donor's tissue can sometimes attack the recipient's tissue, because the cells see it as foreign. A short-term form of this condition, called graft-versus-host disease (GVHD), affects around 40% of bone-marrow transplant patients, while different studies estimate that between 6% and 80% develop a chronic form of GVHD.

There are ways to reduce a patient's risk of this reaction, for example, by prescribing immune-suppressing drugs, but this reduces their ability to fight pathogens. There are also treatments that target a type of immune cell called T cells in the donor tissue. Although these therapies can be effective, they may come with an increased risk of cancer relapse or infection, the scientists behind the new trial wrote in a study published Jan. 4 in the journal Blood.

Related: Zika virus could potentially treat cancer, another early study hints

In contrast, the new treatment, called CD24Fc, derails the immune response at an earlier stage, by inhibiting the response of the cells responsible for activating T cells.

Specifically, the treatment prevents so-called antigen-presenting cells from activating donor T cells that would go on to attack the cancer patient's cells that have been damaged by radiation and chemotherapy before transplantation. These antigen-presenting cells can differentiate between the damaged host cells and pathogens, such as viruses, so CD24Fc only quietens the unwanted inflammation tied to GVHD and not other, helpful immune activity. As such, CD24Fc has also been trialed as a treatment for other conditions that are exacerbated by an off-the-rails immune response to tissue damage, such as severe COVID-19.

"The molecular targets of the intervention are new and original," Dr. Ivan Maillard, a professor of medicine at the University of Pennsylvania who was not involved in the research, told Live Science in an email. CD24Fc kicks in right as the body senses the tissue damage associated with the transplant procedure, rather than later on, he said.

The new trial included 26 patients with blood cancer who received three doses of CD24Fc in the month before they underwent an allogeneic HSCT. They also received the standard, post-transplant immunosuppressive treatment. Of these patients, only one developed moderate-to-severe GVHD within six months of their surgery.

The researchers cross-referenced the trial participants' data to that of 92 patients who underwent the same procedure but without CD24Fc. These patients' results were pulled from a database, and 68 of the patients, or 74%, developed GVHD within six months.

Related: 'Bionic breast' could restore sensation for cancer survivors

The trial's results are "impressive," Dr. Javier Bolaos Meade, a professor of oncology at Johns Hopkins Medicine who was not involved in the research, told Live Science in an email. However, after a year, the researchers found no significant difference in overall survival rates, risk of chronic GVHD, or relapse rates between the two groups, which was "disappointing," he said.

CD24Fc caused minimal side effects in most patients, the authors wrote in the paper. However, two of the 26 patients developed a rare-but-serious skin disorder called Stevens-Johnson syndrome, which is normally caused by an adverse immune reaction to certain medications.

"Whether this rare complication was related to administration of CD24Fc cannot be excluded and will require careful monitoring and evaluation in future trials," Dr. Paul Martin, a professor emeritus of clinical research at the Fred Hutchinson Cancer Center in Seattle who was not involved in the research, wrote in a commentary accompanying the study.

The authors highlighted several limitations of the study, including that it was small and that the conclusions about CD24Fc's effectiveness stem from comparisons to historic data, rather than to a typical control group you'd expect in a gold-standard trial. This may have skewed the reliability of the findings for several reasons; for instance, the trial patients and historic patients may have differed from each other in key ways that weren't flagged or accounted for.

Nevertheless, Bolaos Meade and Maillard anticipate that there will be more studies testing this approach to GVHD prevention, which could provide more robust data about its effectiveness.

This article is for informational purposes only and is not meant to offer medical advice.

Ever wonder why some people build muscle more easily than others or why freckles come out in the sun? Send us your questions about how the human body works to community@livescience.com with the subject line "Health Desk Q," and you may see your question answered on the website!

Go here to see the original:
1st-of-its-kind therapy blocks immune attack after stem-cell transplant - Livescience.com

Stem cell research project to launch into space – Fox Weather

Science experiments are launching to the space station on NASA's 20th Northrop Grumman mission.

A Mayo Clinic research project focusing on gravitys role in bone loss will be one of several experiments aboard a SpaceX Falcon 9 rocket when it lifts off from Cape Canaveral Space Force Station later this month.

The mission is known as NG-20 and is destined to deliver food, supplies and experiments to the International Space Station.

The stem cell experiment has been a long time in the planning and researchers say theyll be able to learn more about tissue repair and regeneration.

"Weve known for some time that astronauts lose bone density on long-duration space flights," Dr. Abba Zubair, a laboratory medicine and pathology specialist at the Mayo Clinic said in a statement. "We want to understand how this occurs so we can work on solutions that prevent bone loss not only in astronauts while theyre in space but also in patients here on Earth."

Northrop Grummans 20th operational cargo delivery flight

(Northrop Grumman / FOX Weather)

Zubair believes the experiment could have implications on clinical trials and travel to Mars.

"We will use what we learn from this project to advance our research on the road to clinical trials, with the ultimate goal of testing therapeutic agents that can prevent or treat bone loss that comes with osteoporosis, as well as bone loss that occurs in patients who are bedridden for long periods of time," Zubair stated.

2024 ROCKET LAUNCH SCHEDULE SHOWS CONTINUED STEADY PACE OF BLAST-OFFS

If weather or technical matters dont delay the launch, itll lift off from Floridas Space Coast on Jan. 29 with spacecraft named after NASA astronaut Dr. Patricia "Patty" Hilliard Robertson.

Robertson was killed during a private plane crash a year before she was set to arrive at the ISS in 2002.

"It is the companys tradition to name each Cygnus spacecraft in honor of an individual who has made substantial contributions to human spaceflight. Dr. Robertson was an accomplished medical doctor and avid acrobatic pilot prior to her NASA career," Northrop Grumman, the producer of the Cygnus spacecraft, stated.

Dr. Patricia "Patty" Hilliard Robertson

(NASA)

A crew of seven aboard the ISS will be tasked with unloading the Cygnus spacecraft a few days after launch.

The mission is Northrop Grummans 20th cargo flight to the ISS, which is expected to continue through 2026.

SEE THE OBJECT HUMANS LEFT BEHIND ON THE MOON

Other experiments aboard the NG-20 will involve testing a 3D metal printer, semiconductor manufacturing and a thermal protection system.

The Mayo Clinic stated a second space flight could launch by the end of the year, which would analyze bone formation and loss.

The combination of experiments is expected to help researchers study bones healing potential and lead to potential treatments that could be used in space and on Earth.

The International Space Station

(NASA)

Visit link:
Stem cell research project to launch into space - Fox Weather

Gut bacteria can be the key to safer stem cell transplantations, study finds – EURACTIV

A new study shows that adverse effects in stem cell transplantation are less common when certain microbes are present in the patients gut, which opens possibilities to create better conditions synthetically and ensure safer outcomes.

Stem cell transplantations can help cure many haematological conditions such as leukaemia, myeloma, and lymphoma in which the bone marrow is damaged and can no longer produce healthy blood cells.

However, there are still considerable risks associated with them, like graft-versus-host disease (GvHD) and transplant-related mortality (TRM).

GvHD can happen after a stem cell transplantation, when in some cases the donor stem cells, the graft, attack healthy cells in the patient, typically in the skin, the gut or the liver.

It affects up to 30% of patients and can be severe. In some cases, patients respond to steroids, but in many others, they are refractory, reducing the survival outcomes and setting the mortality rate as high as 50%.

Some previous studies have shown that the probability of developing GvHD is related to the recipients microbiome, the community of bacteria, fungi, and viruses that reside in patients guts.

Theres been quite a bit of interest in the microbiome because a few landmark studies have shown a correlation between the microbiome and outcomes in stem cell transplantation, Erik Thiele Orberg from TUM (Technical University of Munich) told Euractiv.

We didnt understand the mechanisms that underlie and confer this effect, he explained.

Along with a team of researchers from the TUM and the Universittsklinikum Regensburg (UKR), Thiele Orberg has tried to fill some of the knowledge gaps in a study.

According to Thiele Orberg, these findings will help identify individuals at risk of developing these adverse reactions during stem cell transplantation.

In the study, researchers analysed stool samples from a cohort of patients undergoing stem cell transplantation and confirmed that patients with a higher bacterial diversity had better outcomes, including reduced mortality, lower transplant-related mortality, and less relapse.

They aimed to identify metabolites substances produced by gut bacteria during metabolism that could influence immune responses in patients undergoing stemcell transplantation and identify the microbiome contributing to their production.

Thiele Orberg explained that they were able to find which consortia of protective bacteria, bacteriophages, and metabolites are highly associated with beneficial outcomes and are useful in identifying their lack in patients, creating a risk of developing GVHD and transplant-related mortality.

New possibilities for future procedures

The researchers next step is to figure out how to create this beneficial landscape in the recipients guts.

The studys findings suggest that it may be possible to use synthetic bacteria consortia to produce the protective metabolites identified in the study to improve the transplantations outcomes.

All these new data, Thiele Orberg added, could also be used to improve other already established procedures, like faecal microbiota transplantation (FMT), the transplant of faecal matter from a donor into the intestinal tract of a recipient to change their microbiome.

It is currently being researched in several advanced clinical trials, but we still have the same burning questions in that field, namely what makes a donor a good donor [for FMT] and why do some patients respond and others dont, he explained.

One of the current hypotheses, backed by early pilot experiments, is that the patients who respond to FMT are those able to kick-start their metabolite production after the procedure.

With these new findings, Thiele Orberg explained that a future standard procedure to ensure better outcomes could go as follows:

A patient undergoing stem cell transplantation would be continuously screened using the immune modulatory metabolite risk index. Once a patient is considered to be at risk, they could be prophylactically treated using metabolite cocktails or precision FMT products from donors that have been previously validated for robust metabolite production.

All these discoveries open new investigative paths not only for stem cell transplantation but also for new microbiome studies in other cell therapies.

[Edited by Zoran Radosavljevic]

The rest is here:
Gut bacteria can be the key to safer stem cell transplantations, study finds - EURACTIV

Stem Cell Research Heading to the ISS on Axiom Mission 3 – ISS National Lab

KENNEDY SPACE CENTER (FL), January 17, 2024 More than 5 million people worldwide are living with neurodegenerative disorders like Parkinsons disease and primary progressive multiple sclerosis (PPMS). Researchers funded by the National Stem Cell Foundation (NSCF) are turning to the microgravity environment of the International Space Station (ISS) to better understand and model what causes these debilitating diseases as part of an ISS National Laboratory-sponsored investigation flying on Axiom Spaces third private astronaut mission.

The mission will mark the fifth flight to the orbiting laboratory for NSCF, which is aiming to study tissue changes within stem cell-derived brain organoids to pinpoint where inflammation begins in the brain. Studies have shown a link between inflammation and these types of neurodegenerative diseases, with specialized immune cells within the bodys central nervous system, called microglia, playing a key role in regulating inflammation.

To that end, NSCF will send human brain organoids derived from patients with two different types of degenerative brain diseasesParkinsons and PPMSto the orbiting laboratory. NSCF CEO Paula Grisanti says that the data collected from this flight is crucial. We send research to space because we can see the cells interacting in ways that are not possible on Earth, she said. By adding microglia, we can begin to see where inflammation begins in those processes.

According to Grisanti, findings from the investigation will inform the foundations next mission set to launch in March. Both flights involve organoids created from induced pluripotent stem cells (IPSCs) from affected patients. Approximately 80 organoids will be studied over the two-week mission before being returned to Earth and to NSCF for further analysis.

The absence of gravity acts as an accelerator, speeding up the aging process we see here on Earth, says Grisanti. We turn to space because cells mature more quickly in microgravity, she said. This means we can see the same changes in cells in a matter of weeks or months in microgravity that might take years to see on the ground.

A follow-on investigation will fly on SpaceXs upcoming 30th Commercial Resupply Services (CRS) mission, currently slated for launch in March. On that flight, organoids from patients with Alzheimers disease will be added, and all three sets of cells will be studied over the course of a month. Results from both investigations will be used to inform drug discovery as well as clinical trial assessment for novel therapeutics designed to treat these types of diseases.

By developing human organoids of neurodegenerative diseases, with microglia in the accelerated environment of microgravity, we have added an important new tool and a new way of looking at and understanding how and why neurodegeneration occurs, said Grisanti.

Through private astronaut missions, Axiom Space and the ISS National Lab partner to expand access to the unique microgravity environment for the benefit of humanity. To learn more about all the payloads launching on this mission, please visit Axiom Spaces Research Overview and our launch page.

Download the high-resolution image for this release:Axiom Mission 3

Media Contact: Patrick ONeill 904-806-0035 PONeill@ISSNationalLab.org

# # #

About the International Space Station (ISS) National Laboratory: The International Space Station (ISS) is a one-of-a-kind laboratory that enables research and technology development not possible on Earth. As a public service enterprise, the ISS National Laboratory allows researchers to leverage this multiuser facility to improve quality of life on Earth, mature space-based business models, advance science literacy in the future workforce, and expand a sustainable and scalable market in low Earth orbit. Through this orbiting national laboratory, research resources on the ISS are available to support non-NASA science, technology, and education initiatives from U.S. government agencies, academic institutions, and the private sector. The Center for the Advancement of Science in Space (CASIS) manages the ISS National Lab, under Cooperative Agreement with NASA, facilitating access to its permanent microgravity research environment, a powerful vantage point in low Earth orbit, and the extreme and varied conditions of space. To learn more about the ISS National Lab, visit ourwebsite.

About Axiom Space:Axiom Space is building for beyond, guided by the vision of a thriving home in space that benefits every human, everywhere. The leading provider of human spaceflight services and developer of human-rated space infrastructure, Axiom Space operates end-to-end missions to the International Space Station today while developing its successor, Axiom Station the worlds first commercial space station in low-Earth orbit, which will sustain human growth off the planet and bring untold benefits back home. For more information visit Axiom Spaceswebsite.

View original post here:
Stem Cell Research Heading to the ISS on Axiom Mission 3 - ISS National Lab

Harnessing Healing Power: A Comprehensive Review on Platelet-Rich Plasma in Compound Fracture Care – Cureus

Specialty

Please choose I'm not a medical professional. Allergy and Immunology Anatomy Anesthesiology Cardiac/Thoracic/Vascular Surgery Cardiology Critical Care Dentistry Dermatology Diabetes and Endocrinology Emergency Medicine Epidemiology and Public Health Family Medicine Forensic Medicine Gastroenterology General Practice Genetics Geriatrics Health Policy Hematology HIV/AIDS Hospital-based Medicine I'm not a medical professional. Infectious Disease Integrative/Complementary Medicine Internal Medicine Internal Medicine-Pediatrics Medical Education and Simulation Medical Physics Medical Student Nephrology Neurological Surgery Neurology Nuclear Medicine Nutrition Obstetrics and Gynecology Occupational Health Oncology Ophthalmology Optometry Oral Medicine Orthopaedics Osteopathic Medicine Otolaryngology Pain Management Palliative Care Pathology Pediatrics Pediatric Surgery Physical Medicine and Rehabilitation Plastic Surgery Podiatry Preventive Medicine Psychiatry Psychology Pulmonology Radiation Oncology Radiology Rheumatology Substance Use and Addiction Surgery Therapeutics Trauma Urology Miscellaneous

Read the rest here:
Harnessing Healing Power: A Comprehensive Review on Platelet-Rich Plasma in Compound Fracture Care - Cureus

Grow Up Conference & Expo Announces Top 50 Cannabis Leaders in Canada Ahead of CannaVision ’24 Executive Summit

Leading cannabis event company celebrates national leaders and launches an exclusive gathering for decision-makers in the global arena Leading cannabis event company celebrates national leaders and launches an exclusive gathering for decision-makers in the global arena

See original here:
Grow Up Conference & Expo Announces Top 50 Cannabis Leaders in Canada Ahead of CannaVision '24 Executive Summit