Page 392«..1020..391392393394..400410..»

Stem-Cell Based Therapy Shows Promise in Treating High-Risk Type 1 Diabetes – Newswise

Posted: June 13, 2022 at 2:31 am

Newswise An investigative stem cell-based therapy called PEC-Direct, designed to act as a replacement pancreas, has the potential to provide blood sugar control in patients with high-risk type 1 diabetes, suggests a clinical study presented Saturday, June 11 at ENDO 2022, the Endocrine Societys annual meeting in Atlanta, Ga.

The study found multiple patients using the new treatment had clinically relevant increases in C-peptide, a substance made in the pancreas along with insulin. C-peptide and insulin are released from the pancreas at the same time and in about equal amounts, so measuring C-peptide can show how much insulin the body is making.

This research represents the first instance in multiple patients of clinically relevant increases in C-peptide, indicative of insulin production, with a stem cell-based therapy delivered in a device, according to Manasi Sinha Jaiman, M.D., M.P.H., Chief Medical Officer of ViaCyte, Inc., in San Diego, Calif., the company that makes PEC-Direct.

Patients with type 1 diabetes eventually lose the ability to produce their own insulin to control blood sugar levels. Patients must frequently check those levels with finger sticks, inject multiple insulin shots or carry around bulky devices. The injection of insulin also carries the risk of accidentally lowering blood sugar to dangerous levels.

The PEC-Direct device is designed to provide a long-term, stable source of insulin to regulate glucose levels. The device comprises a pouch containing stem-cell derived pancreatic cells which mature into insulin-producing cells once implanted into the body to regulate glucose levels. The open device membrane allows blood vessels to grow into the device to contact the cells. To prevent an immune reaction, patients take immunosuppressive drugs.

The treatment is meant for patients with high-risk type 1 diabetes, who may be especially vulnerable to acute complications due to factors such as recurrent severe low blood sugar, or frequent and extreme blood sugar fluctuations that are difficult to control.

The study included 10 adults with type 1 diabetes who had received their diagnosis at least 5 years prior to the start of the study and were not able to tell when their blood sugar went too low (called hypoglycemia unawareness). Initial data from one patient showed clinically relevant levels of stimulated C-peptide and corresponding improvements in blood glucose control within six months after implantation of PEC-Direct. Since then, increased C-peptide levels were seen in multiple patients, along with decreases in HbA1C (a blood test that measures average blood sugar levels over the past three months) by as much as 1.5%, and decreases in the amount of insulin patients needed to administer by as much as 70%.

The results suggest stem cell-based replacement therapy has the potential to provide blood glucose control and could one day eliminate the need for injecting or dosing insulin externally, Jaiman said. The study provides further proof-of-concept that continued optimization of PEC-Direct has promise as a functional cure for type 1 diabetes.

###

Endocrinologists are at the core of solving the most pressing health problems of our time, from diabetes and obesity to infertility, bone health, and hormone-related cancers. The Endocrine Society is the worlds oldest and largest organization of scientists devoted to hormone research and physicians who care for people with hormone-related conditions.

The Society has more than 18,000 members, including scientists, physicians, educators, nurses and students in 122 countries. To learn more about the Society and the field of endocrinology, visit our site atwww.endocrine.org. Follow us on Twitter at@TheEndoSocietyand@EndoMedia.

See the original post here:
Stem-Cell Based Therapy Shows Promise in Treating High-Risk Type 1 Diabetes - Newswise

Posted in Stem Cell Therapy | Comments Off on Stem-Cell Based Therapy Shows Promise in Treating High-Risk Type 1 Diabetes – Newswise

What Is Myelofibrosis? Symptoms, Causes, Diagnosis, Treatment, and Prevention – Everyday Health

Posted: June 13, 2022 at 2:31 am

Your treatment approach will depend on how advanced your cancer is, your overall health, what genetic mutations you have, if any, and other factors.

Myelofibrosis is very heterogenous [diverse], explains Mesa. Plans need to be quite individualized because not everybody is the same.

For example, a small subset of people with myelofibrosis who are low risk and minimally affected by the disease may be candidates for the simplest treatment of all: observation. This means the doctor will watch the cancer carefully, but the patient isnt on any active treatment.

Because of these risks, Mesa says stem cell transplants are performed in less than 10 percent of patients with myelofibrosis.

A stem cell transplant can, even in the best circumstances, have a 10 to 15 percent chance of fatal complications, Mesa says. To put that in perspective, open-heart surgery carries only a 1 percent risk.

Participating in a clinical trial may be an option for some people with myelofibrosis. These studies might allow you to receive a therapy that isnt yet available to the public.

Mesa says there are several drugs being studied in clinical trials that could be beneficial for people with myelofibrosis.

Be hopeful. There are a lot of new medicines in development, a lot of things that are being discovered, and a lot of people out there rooting for you even though this is a rare cancer, he says.

See original here:
What Is Myelofibrosis? Symptoms, Causes, Diagnosis, Treatment, and Prevention - Everyday Health

Posted in Stem Cell Therapy | Comments Off on What Is Myelofibrosis? Symptoms, Causes, Diagnosis, Treatment, and Prevention – Everyday Health

European Commission Approves Mosunetuzumab for Patients with R/R Follicular Lymphoma – Targeted Oncology

Posted: June 13, 2022 at 2:31 am

Conditional marketing authorization has been granted to the CD20xCD3 T-cell engaging bispecific antibody mosunetuzumab (Lunsumio) by the European Commission for the treatment of adult patients with relapsed or refractory follicular lymphoma who have previously received at least 2 prior systemic therapies, according to Roche.1

The basis of the approval is supported by findings of the phase 1/2 GO29781 trial (NCT02500407), which met its primary end point of complete response (CR) per independent review facility (IRF) assessment in 2021, with the bispecific antibody eliciting a CR rate of 60% (95% CI, 49%-70%) at a median follow-up of 18.3 months.

Further data revealed the overall response rate (ORR) achieved with the agent to be 80% (95% CI, 70%-88%), and that the median duration of response (DOR) was 22.8 months (95% CI, 9.7not estimable [NE]). Along with this, favorable tolerability was seen in patients with heavily pretreated follicular lymphoma who received mosunetuzumab.

We are delighted that Lunsumio is the first bispecific antibody approved in Europe for people with relapsed or refractory follicular lymphoma, said Levi Garraway, MD, PhD, Roches chief medical officer and head of global product development, in the press release. Lunsumios high response rates, off-the-shelf availability, and initial outpatient administration could transform how advanced follicular lymphoma is treated.

Within the multicenter, open-label, dose-escalation, and dose-expansion trial, mosunetuzumab was examined in patients with follicular lymphoma with grade 1 to 3a disease.2

Eligibility in the trial was open to patients with an ECOG performance status of 0 to 1 and to those who had previously received 2 or more systemic regimens, including at least 1 anti-CD20 antibody and 1 alkylating agent.

Every 3 weeks, as part of 21-day cycles through a step-up dosing approach in cycle 1, patients (n = 90) were administered mosunetuzumab. Within the first cycle, patients received mosunetuzumab at a dose of 1 mg on day 1, 2 mg on day 8, and 60 mg on day 15. For cycle 2, mosunetuzumab was given at a dose of 60 mg on day 1. In subsequent cycles, the bispecific antibody was given at 30 mg on day 1. For the patients who achieved a CR following cycle 8, they were subjected to 17 cycles of treatment. Additionally, there was no mandatory hospitalization for participants given the initial dose.

The primary end point of the trial was CR rate per IRF assessment, evaluated in comparison with the historical CR rate of 14%, and secondary end points included ORR, DOR, progression-free survival (PFS), safety, and tolerability.

Of those enrolled in the study, the median age was 60 years (range, 29-90) with 61.1% being male. Patients with an ECOG performance status of 0 made up 58.9% of the participants while 41.1% had a status of 1. A total of 23.3% of patients had stage I-II Ann Arbor disease, and 76.7% had stage III-IV disease. Additionally, those enrolled in the trial mostly had received 3 prior lines of therapy (range, 2-10), and all patients had previously received anti-CD20 therapy and an alkylator.

Other prior systemic therapies patients received consisted of PI3K inhibitors (18.9%), immunomodulatory drugs (14.4%), and chimeric antigen receptor (CAR) T-cell therapy (3.3%). Patients who had previously had autologous stem cell transplant made up 21.1% of participants.

Moreover, 68.9% of individuals were refractory to the last prior therapy they received, 78.9% of patients were refractory to a previous anti-CD20 therapy, and 53.3% were double refractory to an anti-CD20 therapy as well as an alkylating therapy. A total of 52.2% of patients also had progression of disease within 24 months (POD24) of starting treatment.

Data presented at the 2021 ASH Annual Meeting revealed that mosunetuzumab had an investigator-assessed CR rate of 60% (95% CI, 49%-70%), and the CR rates produced with the agent in high-risk subgroups were similar to that of the overall study population.

Patients younger than 65 years of age (n = 60) had a CR rate with the bispecific antibody of 55% (95% CI, 42%-68%) compared with a CR rate of 70% (95% CI, 51%-85%) in patients aged 65 years or older (n = 30). With the agent, the CR rate was 74% (95% CI, 56%-87%) for those who previously received 2 lines of therapy (n = 34) and 52% (95% CI, 39%-65%) in patients who received 3 or more prior lines (n = 56). Patients who were relapsed or refractory to their last prior therapy (n = 62) and in those who were not (n = 28) demonstrated CR rates of 52% (95% CI, 39%-65%) and 79% (95% CI, 59%-92%), respectively.

Mosunetuzumab also produced a CR rate of 50% (95% CI, 35%-65%) in the group of individuals who were double refractory (n = 48) vs 71% (95% CI, 55%-84%) in those who were not. Further, the CR rate was shown to be 57% (95% CI, 42%-72%) for those with POD24 disease (n = 47) compared with 63% (95% CI, 47%-77%) in those who did not progress within 24 months (n = 43).

Median time to response was 1.4 months (range, 1.1-8.9), and the median time to first CR was 3.0 months (range, 1.1-18.9). The median PFS with mosunetuzumab was 17.9 months (95% CI, 10.1-NE).

In regard to safety, cytokine release syndrome (CRS) was the most common toxicity, which was observed in 39% of patients. Still, CRS was low grade with 14% of patients experiencing grade 2 CRS, and it was resolved by treatment completion. Other frequently observed adverse effects included neutropenia, hypophosphatemia, pyrexia, and headache.

Two phase 3 clinical studies are exploring the use of mosunetuzumab in the second-line setting: CELESTIMO (NCT04712097), investigating mosunetuzumab plus lenalidomide (Revlimid) for patients with follicular lymphoma, and SUNMO (NCT05171647), investigating mosunetuzumab plus polatuzumab vedotin (Polivy) for patients with diffuse large B-cell lymphoma.

Having additional treatment options for people with follicular lymphoma, where multiple prior lines of therapy have failed, is critical to help them achieve better outcomes, Elizabeth Budde, MD, PhD, a hematologic oncologist and associate professor at the City of Hope Comprehensive Cancer Center, added in the press release. It is exciting to have a new class of immunotherapy like Lunsumio, offering a readily available, chemotherapy-free and fixed-duration treatment, with great potential to provide durable remissions without the need to stay on treatment continuously.

Link:
European Commission Approves Mosunetuzumab for Patients with R/R Follicular Lymphoma - Targeted Oncology

Posted in Stem Cell Therapy | Comments Off on European Commission Approves Mosunetuzumab for Patients with R/R Follicular Lymphoma – Targeted Oncology

Bluebird’s future in balance as FDA weighs gene therapy approvals – BioPharma Dive

Posted: June 13, 2022 at 2:30 am

After years of research and billions of dollars spent, Bluebird bio is on the cusp of a milestone only a few drugmakers have ever reached.

Two gene therapies its developed are under review by the Food and Drug Administration and, if approved, would become only the third and fourth cleared by the agency for use in treating inherited diseases.

But Bluebird, a pioneer in the field, simultaneously faces financial peril. The Massachusetts-based biotech is quickly running out of cash and earlier this year warned investors that it may struggle to stay solvent. Its already cutting costs.

Approvals, should they come, could mean Bluebirds survival. They would also help restore confidence in a field thats been recently shaken by safety scares and regulatory surprises.

Most importantly, FDA clearances would bring new treatment options for patients with few. One of Bluebirds therapies treats the rare blood condition beta thalassemia, which in its severe form requires blood transfusions every few weeks for life. The other is meant to halt the progression of a devastating neurological disorder called CALD, or cerebral adrenoleukodystrophy, that affects young boys.

On Thursday and again on Friday, the FDA will convene a panel of outside experts to review Bluebirds data and advise it on the treatments respective benefits and risks. Documents published Tuesday indicate agency scientists are cautiously supportive of the beta thalassemia treatment, called beti-cel, but are more skeptical of the CALD therapy, eli-cel.

The FDA is due to make its decisions by Aug. 19 and Sept. 16, respectively. Its verdicts could determine Bluebirds fate, shape how other gene therapies are developed and, potentially, change how two genetic diseases are treated.

I think there really is a spotlight on how this is handled by the FDA, said Christine Duncan, a physician at Boston Childrens Hospital and medical director of the gene therapy program there.

Four years ago, Bluebird was riding high. The biotechs progress developing gene therapies for rare genetic diseases early last decade had helped propel the field forward, and Bluebird, as a result, had become one of the industrys most valuable companies.

That January, Nick Leschly, Bluebirds longtime CEO, took the stage at the pharmaceutical industrys most closely watched conference and told the gathered crowd his company would soon ask regulators for approval of three experimental drugs.

It was a noteworthy declaration, meant to signal Bluebirds coming transition into a commercial-stage drugmaker after nearly three decades of laboratory and clinical research. Leschlys forum, the grand ballroom of the Westin St. Francis at that years J.P. Morgan Healthcare Conference, seemed to match the moment, and reflect Bluebirds ascension.

We believe we're in a leadership position in an important field that's emerging, Leschly said, according to a transcript of his 2018 speech by market data firm Koyfin.

But Bluebirds transition has been rockier than anticipated. While the company did win milestone approvals of beti-cel and eli-cel in Europe, manufacturing hurdles and difficulties securing reimbursement forced Bluebird to later pull both from market and shut down its operations there. Only a handful of patients received either treatment.

In the U.S., disagreements with the FDA over production tests twice delayed Bluebirds plans to seek approval of beti-cel, and slowed progress with another gene therapy for the blood condition sickle cell. The FDA also initially refusedBluebird and partner Bristol Myers Squibbs application for the third drug referred to by Leschly, a cell therapy for multiple myeloma, before eventually approving it in early 2021.

Its the challenge of being [on] a frontier. You hit all the waves first, Leschly said in a February interview, a few months after leaving to head 2seventybio, a new biotech that Bluebird created by spinning out its oncology business. (Andrew Obenshain, formerly Bluebirds genetic disease head, succeeded Leschly as Bluebirds CEO.)

Other gene therapy developers have struggled to cross the FDAs finish line as well, or have run into unexpected roadblocks with their products

Whats happening to them is not unique to Bluebird, said Nicole Paulk, an assistant professor of AAV gene therapy at the University of California, San Francisco. Any gene therapy company you can name off the top of your head theyre all facing the same problems around manufacturing and [production controls].

For Bluebird, though, the delays and setbacks have been especially costly. Its market value, near $10 billion in early 2018, has dwindled to just above $250 million currently. The company moved its headquarters from Cambridge to Somerville, Massachusetts to save money and in April said it will lay off about a third of its employees.

The cost-cutting might not be enough, either. Bluebird, which regularly records quarterly losses totaling hundreds of millions of dollars, now has just $267 million in available funds. In March and again in May, the biotech warned there was substantial doubt it will be able to stay afloat for the next year.

Bluebirds survival may come down to whether the FDA clears beti-cel and eli-cel. Approval of either would give Bluebird the chance to make money not only from commercial sales, but also from special regulatory vouchers the FDA awards that typically sell for about $100 million each.

Yet Bluebirds treatments arent a sure bet to win the FDAs blessing. Looming over both are safety concerns that have cropped up over the past year.

Last February, Bluebird suspended testing of its sickle cell gene therapy after a study participant developed a form of leukemia. While no cases were reported for beti-cel, which uses similar technology, the FDA subsequently placed study holds on both therapies.

An investigation by the company found no clear link between treatment and the leukemia case, and in June the FDA lifted its mandated trial pauses.

But then in August Bluebird reported a case of myelodysplastic syndrome, a cancer-like condition of the bone marrow that can evolve into leukemia, in a CALD patient treated with eli-cel.

Two more eli-cel patients were subsequently diagnosed with myelodysplastic syndrome, or MDS. Two of the cases were directly linked to treatment, while the third was judged highly likely to be related.

In documents published Tuesday, FDA scientists made clear their concerns, noting that, as all three cases occurred long after treatment, they expect more to emerge from recently treated study participants. Laboratory testing also revealed troubling genetic signs in many of the other patients that could portend but not necessarily cause future cancer.

Both eli-cel and beti-cel, as well as Bluebirds sickle cell treatment, are based around a similar therapeutic concept. Stem cells are taken from each patient and, in a laboratory, genetically modified using a type of virus known as a lentivirus. Once reinfused, the engineered stem cells mature and express proteins that replace ones mutated or missing in beta-thalassemia, CALD and sickle cell.

The question is whether the insertion of the virus into the genome of the cells has resulted in the clonal expansion of that population of cells and, potentially, evolution towards leukemia, said Paul Orchard, a pediatric transplant specialist at the University of Minnesota who helped run a study of eli-cel.

The FDA and its advisers will have to weigh the therapies safety risks against their benefits. In the case of eli-cel, the treatments effectiveness was primarily measured as the percent of treated patients who survived two years without major functional disabilities like tube-feeding or wheelchair dependence.

Twenty-nine of the 32 study participants assessed reached this goal, a rate thats near what Bluebird found for similar CALD patients treated with donor-derived stem cell transplants in two natural history studies.

Donor, or allogeneic, transplants can be an effective treatment for CALD, but work best when the stem cells come from a sibling. Unfortunately, only about 30% of boys with CALD have matched sibling donors, according to the FDA.

Aside from donor factors, it is unclear if there is a CALD population for whom the benefit of treatment with eli-cel outweighs the significant and unknown long-term risk of MDS, FDA staff wrote in the documents.

Duncan, of Boston Childrens, agrees the tradeoffs are difficult to weigh but argues that, for some at least, eli-cel would be a needed additional option.

Its not black and white, said Duncan, who is an investigator in an eli-cel study and will present to the FDAs advisers Thursday on behalf of Bluebird. This is an area of gray in that for some patients, if they dont have an allogeneic stem cell transplant match, they dont have a choice. Their disease progresses, theyre neurologically devastated and they die.

The FDA appears more supportive of beti-cel, which in testing showed a powerful ability to free beta thalassemia patients from needing regular blood transfusions. Agency staff agreed the data show it to provide a meaningful benefit for those patients and noted treatment could reduce serious health risks associated with chronic transfusions.

Even so, the FDA is asking its advisers to weigh the cancer risk observed in testing of eli-cel and Bluebirds sickle cell therapy, and whether that should impact its decision on beti-cel.

Neither eli-cel or beti-cel, if approved, are expected to become top-selling drugs, despite expectations that Bluebird could price them similarly to other gene therapies available in the U.S. and Europe that cost more than $1 million.

Bluebird estimates there are between 1,000 and 1,300 beta thalassemia patients in the U.S. who require regular blood transfusions. The number of boys with CALD is far less, perhaps as few as 50 in the U.S, according to analysts from RBC Capital Markets.

But the impact of each treatment could still be significant, both for patients that might receive them and for the broader gene therapy field.

Only two gene therapies for inherited diseases are approved in the U.S. Luxturna for a form of childhood blindness and Zolgensma for the neurodegenerative disease spinal muscular atrophy. (Six CAR-T cell therapies, which are sometimes classified as gene therapy, are also approved for a range of blood cancers.)

Previously FDA officials had predicted that, by 2025, theyd be reviewing between 10 and 20 gene and cell therapies each year a figure that now looks likely to be an overestimate. The agency also appears to be moving cautiously as more safety concerns beyond Bluebird emerge, freezing a number of gene therapy studies over the past year.

And while investment in gene therapy has boomed, a large number of companies are now restructuring their research, cutting costs or laying off employees amid a broader biotech market downturn.

We need a catalyst moment to bring us out of this sentiment slump, said Paulk.

FDA approval of beti-cel and eli-cel could provide one. If the agency decides to hold off, expectations for more gene therapy clearances in the near future might be further tempered.

For Bluebird, the consequences are clearer. The FDAs decisions could determine whether the company remains around long enough to provide either therapy to patients.

It would be extremely unfortunate to get to the point where [weve] developed efficacious therapies and then the companies dont have the finances to move forward, said Orchard, of the University of Minnesota.

Read the original here:
Bluebird's future in balance as FDA weighs gene therapy approvals - BioPharma Dive

Posted in Minnesota Stem Cells | Comments Off on Bluebird’s future in balance as FDA weighs gene therapy approvals – BioPharma Dive

Neural stem cells: developmental mechanisms and disease modeling

Posted: June 13, 2022 at 2:29 am

Cell Tissue Res. Author manuscript; available in PMC 2018 May 22.

Published in final edited form as:

PMCID: PMC5963504

NIHMSID: NIHMS967727

1Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA

2Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA

1Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA

1Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA

2Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA

The astonishing progress in the field of stem cell biology during the past 40 years has transformed both science and medicine. Neural stem cells (NSCs) are the stem cells of the nervous system. During development they give rise to the entire nervous system. In adults, a small number of NSCs remain and are mostly quiescent; however, ample evidence supports their important roles in plasticity, aging, disease, and regeneration of the nervous system. Because NSCs are regulated by both intrinsic genetic and epigenetic programs and extrinsic stimuli transduced through the stem cell niche, dysregulation of NSCs due to either genetic causes or environmental impacts may lead to disease. Therefore, extensive investigations in the past decades have been devoted to understanding how NSCs are regulated. On the other hand, ever since their discovery, NSCs have been a focal point for cell-based therapeutic strategies in the brain and spinal cord. The limited number of NSCs residing in the tissue has been a limiting factor for their clinical applications. Although recent advancements in embryonic and induced pluripotent stem cells have provided novel sources for NSCs, several challenges remain. In this special issue, leaders and experts in NSCs summarize our current understanding of NSC molecular regulation and the importance of NSCs for disease modeling and translational applications.

The term stem cells first appeared in the scientific literature in 1868 by the German biologist Ernst Haeckel (Haeckel, 1868). In his writings (Haeckel, 1868), stem cells had two distinct meanings: one is the unicellular evolutionary origin of all multicellular organisms, and the other is the fertilized egg giving rise to all other cell types of the body. The latter definition has evolved into the modern definition of stem cells - cells that can divide to self-renew and to differentiate into other cell types in tissues and organs (Li and Zhao, 2008, Ramalho-Santos and Willenbring, 2007).

The behavior and fate of stem cells are strongly influenced by their specific anatomical locations and surrounding cell types, called the stem cell niche. The niche provides physical support to host or anchor stem cells, and supplies factors to maintain and regulate them (Li and Zhao, 2008). Stem cells are also regulated by intrinsic signaling cascades and transcriptional mechanisms, some of which are common among all stem cells, and others that are unique to specific types. Some of the best known regulators include TGF-, BMP, Smad, Wnt, Notch, EGF fibroblast growth factors (Jobe, et al., 2012, Li and Zhao, 2008). Therefore, stem cells are regulated by complex mechanisms in both temporal- and context-specific manners to maintain their unique characteristics. Understanding stem cell regulation gives us the opportunity to explore mechanisms of development, as well as disorders resulting from their dysfunction.

During development, the central nervous system (CNS) is generated from a small number of neural stem cells (NSCs) lining the neural tube (Kriegstein and Alvarez-Buylla, 2009). A great deal of experimental evidence has demonstrated that radial glia, the NSCs during mammalian CNS development, undergo both symmetric divisions to expand the NSC pool, and asymmetric divisions to give rise to intermediate progenitors (IPCs) and the differentiated cell types. The three major cell types in the CNS arise from NSCs in a temporally defined sequence, with neurons appearing first, followed by astrocytes, and then oligodendrocytes (Okano and Temple, 2009). The technical advancement of live imaging and genomic tools have allowed for the identification of human-specific NSC populations (e.g. outer radial glia, or oRG) located at the outer subventricular zone (SVZ) (Gertz, et al., 2014). These oRG are essential for cortical expansion to achieve the large size of the human cortex. Single-cell genomic technologies have identified specific oRG markers that might be used for further characterization of these cells (Liu, et al., 2016, Pollen, et al., 2014). Investigating the regulatory mechanisms governing the self-renewal and fate specification of NSCs, especially human-specific developmental features, has significantly contributed to our understanding of human brain development and developmental diseases. In addition, this knowledge also has helped scientists refine protocols for pluripotent stem cell differentiation into specific nervous system cell types for both therapeutic goals and disease modeling.

In adult brains, NSCs are reduced and become restricted to specific brain regions. In rodents, both NSCs and ongoing neurogenesis have been widely documented in the SVZ of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus (DG) of the hippocampus (Kempermann, et al., 2015). In humans, experimental evidence has supported ongoing neurogenesis in the hippocampus (Eriksson, et al., 1998, Spalding, et al., 2013). The confirmation of mammalian adult neurogenesis in the 1990s was one of the most exciting moments in science in the 21st century. Not only did it overthrow the prevailing dogma suggesting no neurons were made in the adult brain, but also it hinted that these adult NSCs could be utilized for neural repair in disease and following injury. Forty years later, we have learned a lot about NSCs. In the adult rodent SVZ, neurogenesis has been shown to be important for olfactory function and olfactory learning (Alonso, et al., 2006). During development, a subset of slowly-dividing NSCs are set aside to be the NSCs of the SVZ in the postnatal and adult brain (Fuentealba, et al., 2015, Furutachi, et al., 2015). The majority of neurogenic radial glia, however, become astrocytes and ependymal cells at the end of embryonic neurogenesis (Noctor, et al., 2004). A subset of these astrocytes persist as NSCs in specialized niches in the adult brain and continuously generate neurons that functionally integrate into restricted brain regions (Doetsch, 2003). In the hippocampus, radial glia-like stem cells of the SGZ make newborn neurons throughout life (Goritz and Frisen, 2012). These newborn neurons integrate into the circuity of the DG, contributing to behaviors such as pattern separation (Aimone, et al., 2011) and spatial learning (Dupret, et al., 2008), as well as hippocampus-associated learning, memory, and executive functions (Kempermann, Song and Gage, 2015).

Significant effort has been devoted into understanding the regulation of adult neurogenesis. As a result, we now know that many extrinsic stimuli and intrinsic mechanisms can affect this process. Mouse genetic studies have clearly demonstrated the important role of transcriptional regulation of NSCs through intrinsic genetic mechanisms (Hsieh and Zhao, 2016). Some examples include SOXC family proteins [Kavyanifar et al, in this issue (Kavyanifar, et al., 2018)], Bmi-1 (Molofsky, et al., 2003), Sox2 (Ferri, et al., 2004, Graham, et al., 1999), PTEN (Bonaguidi, et al., 2011), and Notch [Zhang et al, in this issue (Zhang, et al., 2018)]. In addition epigenetic regulation by DNA methylation pathways (e.g. Mbd1, Mecp2, Dnmt, Tet) (Noguchi, et al., 2015, Smrt, et al., 2007, Tsujimura, et al., 2009, Zhang, et al., 2013, Zhao, et al., 2003), chromatin remodeling (e.g. BAF, BRG1) (Ninkovic, et al., 2013, Petrik, et al., 2015, Tuoc, et al., 2017), and noncoding RNAs (Liu, et al., 2010)[Anderson and Lim, in this issue (Anderson and Lim, 2018)] play important roles. Many growth factors, signaling molecules, and neurotransmitters have been shown to regulate neurogenesis (Kempermann, Song and Gage, 2015). Catavero et al [in this issue (Catavero, et al., 2018)] review the role of GABA circuits, signaling, and receptors in regulating development of adult born cells, as well as the molecular players that modulate GABA signaling. Because progenitors with multipotent differentiation potentials have been found in brain regions without active neurogenesis (Palmer, et al., 1997), it is hypothesized that these progenitors might be manipulated to become neuron-competent in vivo so that they can contribute to brain generation [Wang et al, in this issue (Wang and Zhang, 2018)].

A great amount of literature has documented how physiological activities and enriched environment influences adult neurogenesis (Kempermann, Song and Gage, 2015). However, as summarized by Eisinger and Zhao [in this issue (Eisinger and Zhao, 2018)], the genes and gene network involved in these changes within NSCs have not been systematically analyzed at genome wide levels. Adult neurogenesis is also influenced by diseases including epilepsy (Parent and Lowenstein, 1997), stroke (Zhang and Chopp, 2016), depression (Dranovsky and Hen, 2006, Kempermann, et al., 2003), and injury [(Morshead and Ruddy, in this issue (Morshead and Ruddy, 2018) in this issue). Thodeson et al [in this issue (Thodeson, et al., 2018)] further summarize the contribution and dysregulation of adult neurogenesis in epilepsy and discuss how we can translate these findings to human therapeutics by using patient-derived neurons to study monogenic epilepsy-in-a-dish.

Aging affects every individual and is a major risk factor for many diseases. One of the strongest negative regulators of adult neurogenesis is aging. Both intrinsic and extrinsic components regulate the limitations of NSC proliferation and function (Moore and Jessberger, 2017, Seib and Martin-Villalba, 2015). In this issue, Mosher and Schaffer (Mosher and Schafer, 2018) and Ruddy and Morshead (Morshead and Ruddy, 2018) examine factors such as secreted signals, cell contact- dependent signals, and extracellular matrix cues that control neurogenesis in an age-dependent manner, and define these signals by the extrinsic mechanism through which they are presented to the NSCs. Smith et al [in this issue (Smith, et al., 2018)] discuss how age-related changes in the blood, such as blood-borne-factors, and peripheral immune cells, contribute to the age-related decline in adult neurogenesis in the mammalian brain.

Despite the extensive knowledge we have gained regarding adult neurogenesis, critical questions remain. For example, the control of the functional integration of new neurons remains a mystery. It has been shown that adult NSC-differentiated newborn neurons exhibit a critical period for sensitivity to external stimuli (Bergami, et al., 2015), and a heightened sensitivity to seizures (Kron, et al., 2010). It remains unclear how new neurons choose their connections. Jahn and Bergami [in this issue (Jahn and Bergami, 2018)] further discuss the critical period and its regulators during adult newborn neuron development.

Understanding the extrinsic and intrinsic regulation of adult NSCs and their newborn progeny, and their response to both positive and negative stimuli will further illuminate their role in disease, injury, stress, and brain function.

Human pluripotent stem cells (PSCs), including human embryonic stem cells (ESCs) and induced PSCs (iPSCs), offer a model system to reveal cellular and molecular events underlying normal and abnormal neural development in humans. ESCs are pluripotent cells derived from the inner cell mass of blastocyst stage preimplantation embryos, which were first isolated from mouse by Evans and Kaufman in 1981 (Evans and Kaufman, 1981) and later from humans by James Thompson in 1998 (Thomson, et al., 1998). Human ESCs are invaluable in the study of early embryonic development, allowing us to identify critical regulators of cell commitment, differentiation, and adult cell reprogramming (Dvash, et al., 2006, Ren, et al., 2009). iPSCs are reprogrammed from somatic cells by forced expression of stem cell genes and have the characteristics of ESCs (Okita, et al., 2007, Yu, et al., 2007). The development of iPSC technology has allowed us access to cells of the human nervous system through reprogramming of patient-derived cells, revolutionizing our ability to study human development and diseases.

To generate neural cells from either ESCs or iPSCs, the first step is neural induction. Through actions of a number of activators and inhibitors of cell signaling pathways, this process yields neural epithelial cell-like NSCs and then intermediate neural progenitors, resembling embryonic development. Despite many advances, a major hurdle of neural differentiation is lineage control. Using a standard dorsal forebrain neural differentiation protocol, most neural progenitors obtained are forebrain excitatory progenitors that produce mostly forebrain glutamatergic excitatory neurons. However, the purity and layer-specific composition of these progenitors, as well as neurons, vary significantly from experiment to experiment, cell line to cell line, and lab to lab. In addition, differentiation into specific types of neurons with high purity has always been a challenging goal. Much effort has been devoted into improving the efficiency of dopaminergic neuron and GABAergic neuron differentiation with great success (Hu, et al., 2010). However, the brain has many other types of neurons. Vadodaria et al [in this issue (Vadodaria, et al., 2018)] discuss how to generate serotonergic neurons, a type of neuron highly relevant to psychiatric disorders. To better understand the molecular control of human PSC and NSC differentiation, where protocols result in a large amount of cellular heterogeneity in identity and response, analysis must be done at the level of single cells. Harbom et al [in this issue (Harbom, et al., 2018)] summarizes how new state-of-the-art single-cell analysis methods may help to define differentiation from pluripotent cells.

The advancement in iPSC and gene editing technology has transformed the field of human disease modeling. As in many human disorders, especially neuropsychiatric disorders, mouse models have been useful. Yet there are several critical reasons why it is necessary to use human cells to define the underlying mechanisms that lead to human patient characteristics, particularly those affecting the nervous system. For example, in fragile X syndrome (FXS), the epigenetic silencing of the Fragile X Mental Retardation Gene 1 (FMR1) gene that causes FXS occurs only in humans. Mice engineered to mimic the human mutation in the FMR1 gene do not show the same methylation and silencing characteristics of the gene as in humans (Brouwer, et al., 2007). These results indicate that some epigenetic mechanisms in human and mice are different and preclude the ability to study epigenetic mechanisms of FMR1 silencing in mouse models of FXS (Bhattacharyya and Zhao, 2016). In this issue, Li and Shi discuss disease modeling using human PSC-differentiated neural progenitors (Li, et al., 2018), and Brito et al specifically focus on modeling autism spectrum disorder (Brito, et al., 2018).

The use of NSCs as a treatment strategy in CNS disease and injury has been tested for decades. Parkinsons disease specifically has gained the most momentum for potential therapeutic benefits (Studer, 2017); however, similar work has been performed in Huntingtons disease, stroke, and following spinal cord injury [for a review on this topic, see (Vishwakarma, et al., 2014)]. In some of these paradigms, NSCs are expected to differentiate into a specific cell type in the local CNS environment; in other cases, they are in a supportive role. In this issue, Kameda et al explores progress in using NSCs as a therapy following spinal cord injury (Kameda, et al., 2018).

While the development of PSC technologies has been a scientific breakthrough for future studies, there are limitations and risks that may be associated with their use. ESCs, iPSCs, and their differentiated NSCs are dividing cells. Either transplantation of NSCs or in vivo reprogramming of endogenous cells into NSCs could lead to tumorigenesis. In addition, reprogramming somatic cells into iPSCs results in a loss of some epigenetic signatures of disease and aging which are critical for disease modeling (Mertens, et al., 2015, Miller, et al., 2013, Ocampo, et al., 2016). In recent years, direct reprogramming of fibroblasts or other cell types into induced neurons (iN) has been developed (for review see (Mertens, et al., 2016)). Remarkably a growing number of studies have demonstrated that such direct reprogramming also can be effective in vivo. Wang et al [(Wang and Zhang, 2018) in this issue] will summarize recent progress of in vivo reprogramming into new neurons and present how this method can be used for spinal cord injury.

In cellular reprogramming, the cells targeted and the genetic factors used vary; however, the biggest difference is that some protocols push cells through a NSC stage, whereas others skip these stages (Gascon, et al., 2017, Guo, et al., 2014, Wang, et al., 2016). Bypassing this developmental stage has both pros and cons, and may lead to a completely novel path towards lineage commitment [discussed by Falk and Karow (Falk and Karow, 2018) in this issue].

NSCs are fascinating and promising cells because of their capability, flexibility, and multiplicity. Understanding how NSCs function provides important knowledge in development, adaptation, disease, regeneration, and rehabilitation of the nervous system. The studies of cortical development and adult neurogenesis using rodent models have contributed significantly to our knowledge about NSCs and will continually yield important new information, taking advantage of novel genetic and imaging technologies. However, using human NSCs provides us with a window to investigate human-specific aspects of development and disease mechanisms, which is potentiated by the fast advancement of stem cell and gene editing technologies. Challenges still remain regarding cell lineage control, in vivo NSC behavior, three dimensional cellular interactions, and preservation of epigenetic and aging marks.

We thank Klaus Unsicker for his encouragement and support and Jutta Jger for her help with invitations, and communications with authors and reviewers. This work was supported by grants from the US National Institutes of Health (R01MH078972, R56MH113146, R21NS098767, and R21NS095632 to X.Z, U54HD090256 to the Waisman Center), University of Wisconsin (UW)-Madison Vilas Trust (Kellett Mid-Career Award to X.Z.) and UW-Madison and Wisconsin Alumni Research Foundation (WARF to X.Z.), Jenni and Kyle Professorship (to X.Zhao), a Sloan Research Fellowship (to D.L.M.), a Junior Faculty Grant from the American Federation for Aging Research (to D.L.M.), and startup funds from UW-Madison School of Medicine and Public Health, WARF, and the Neuroscience Department (to D.L.M.).

Read more:
Neural stem cells: developmental mechanisms and disease modeling

Posted in Wisconsin Stem Cells | Comments Off on Neural stem cells: developmental mechanisms and disease modeling

The benefits and risks of stem cell technology – PMC

Posted: June 13, 2022 at 2:29 am

Stem cell technology will transform medical practice. While stem cell research has already elucidated many basic disease mechanisms, the promise of stem cellbased therapies remains largely unrealized. In this review, we begin with an overview of different stem cell types. Next, we review the progress in using stem cells for regenerative therapy. Last, we discuss the risks associated with stem cellbased therapies.

There are three major types of stem cells as follows: adult stem cells (also called tissue-specific stem cells), embryonic stem (ES) cells, and induced pluripotent stem (iPS) cells.

A majority of adult stem cells are lineage-restricted cells that often reside within niches of their tissue of origin. Adult stem cells are characterized by their capacity for self-renewal and differentiation into tissue-specific cell types. Many adult tissues contain stem cells including skin, muscle, intestine, and bone marrow (Gan et al, 1997; Artlett et al, 1998; Matsuoka et al, 2001; Coulombel, 2004; Humphries et al, 2011). However, it remains unclear whether all adult organs contain stem cells. Adult stem cells are quiescent but can be induced to replicate and differentiate after tissue injury to replace cells that have died. The process by which this occurs is poorly understood. Importantly, adult stem cells are exquisitely tissue-specific in that they can only differentiate into the mature cell type of the organ within which they reside (Rinkevich et al, 2011).

Thus far, there are few accepted adult stem cellbased therapies. Hematopoietic stem cells (HSCs) can be used after myeloablation to repopulate the bone marrow in patients with hematologic disorders, potentially curing the underlying disorder (Meletis and Terpos, 2009; Terwey et al, 2009; Casper et al, 2010; Hill and Copelan, 2010; Hoff and Bruch-Gerharz, 2010; de Witte et al, 2010). HSCs are found most abundantly in the bone marrow, but can also be harvested at birth from umbilical cord blood (Broxmeyer et al, 1989). Similar to the HSCs harvested from bone marrow, cord blood stem cells are tissue-specific and can only be used to reconstitute the hematopoietic system (Forraz et al, 2002; McGuckin et al, 2003; McGuckin and Forraz, 2008). In addition to HSCs, limbal stem cells have been used for corneal replacement (Rama et al, 2010).

Mesenchymal stem cells (MSCs) are a subset of adult stem cells that may be particularly useful for stem cellbased therapies for three reasons. First, MSCs have been isolated from a variety of mesenchymal tissues, including bone marrow, muscle, circulating blood, blood vessels, and fat, thus making them abundant and readily available (Deans and Moseley, 2000; Zhang et al, 2009; Lue et al, 2010; Portmann-Lanz et al, 2010). Second, MSCs can differentiate into a wide array of cell types, including osteoblasts, chondrocytes, and adipocytes (Pittenger et al, 1999). This suggests that MSCs may have broader therapeutic applications compared to other adult stem cells. Third, MSCs exert potent paracrine effects enhancing the ability of injured tissue to repair itself. In fact, animal studies suggest that this may be the predominant mechanism by which MSCs promote tissue repair. The paracrine effects of MSC-based therapy have been shown to aid in angiogenic, antiapoptotic, and immunomodulatory processes. For instance, MSCs in culture secrete hepatocyte growth factor (HGF), insulin-like growth factor-1 (IGF-1), and vascular endothelial growth factor (VEGF) (Nagaya et al, 2005). In a rat model of myocardial ischemia, injection of human bone marrow-derived stem cells upregulated cardiac expression of VEGF, HGF, bFGF, angiopoietin-1 and angiopoietin-2, and PDGF (Yoon et al, 2005). In swine, injection of bone marrow-derived mononuclear cells into ischemic myocardium was shown to increase the expression of VEGF, enhance angiogenesis, and improve cardiac performance (Tse et al, 2007). Bone marrow-derived stem cells have also been used in a number of small clinical trials with conflicting results. In the largest of these trials (REPAIR-AMI), 204 patients with acute myocardial infarction were randomized to receive bone marrow-derived progenitor cells vs placebo 37 days after reperfusion. After 4 months, the patients that were infused with stem cells showed improvement in left ventricular function compared to control patients. At 1 year, the combined endpoint of recurrent ischemia, revascularization, or death was decreased in the group treated with stem cells (Schachinger et al, 2006).

Embryonic stem cells are derived from the inner cell mass of the developing embryo during the blastocyst stage (Thomson et al, 1998). In contrast to adult stem cells, ES cells are pluripotent and can theoretically give rise to any cell type if exposed to the proper stimuli. Thus, ES cells possess a greater therapeutic potential than adult stem cells. However, four major obstacles exist to implementing ES cells therapeutically. First, directing ES cells to differentiate into a particular cell type has proven to be challenging. Second, ES cells can potentially transform into cancerous tissue. Third, after transplantation, immunological mismatch can occur resulting in host rejection. Fourth, harvesting cells from a potentially viable embryo raises ethical concerns. At the time of this publication, there are only two ongoing clinical trials utilizing human ES-derived cells. One trial is a safety study for the use of human ES-derived oligodendrocyte precursors in patients with paraplegia (Genron based in Menlo Park, California). The other is using human ES-derived retinal pigmented epithelial cells to treat blindness resulting from macular degeneration (Advanced Cell Technology, Santa Monica, CA, USA).

In stem cell research, the most exciting recent advancement has been the development of iPS cell technology. In 2006, the laboratory of Shinya Yamanaka at the Gladstone Institute was the first to reprogram adult mouse fibroblasts into an embryonic-like cell, or iPS cell, by overexpression of four transcription factors, Oct3/4, Sox2, c-Myc, and Klf4 under ES cell culture conditions (Takahashi and Yamanaka, 2006). Yamakana's pioneering work in cellular reprogramming using adult mouse cells set the foundation for the successful creation of iPS cells from adult human cells by both his team (Takahashi et al, 2007) and a group led by James Thomson at the University of Wisconsin (Yu et al, 2007). These initial proof of concept studies were expanded upon by leading scientists such as George Daley, who created the first library of disease-specific iPS cell lines (Park et al, 2008). These seminal discoveries in the cellular reprogramming of adult cells invigorated the stem cell field and created a niche for a new avenue of stem cell research based on iPS cells and their derivatives. Since the first publication on cellular reprogramming in 2006, there has been an exponential growth in the number of publications on iPS cells.

Similar to ES cells, iPS cells are pluripotent and, thus, have tremendous therapeutic potential. As of yet, there are no clinical trials using iPS cells. However, iPS cells are already powerful tools for modeling disease processes. Prior to iPS cell technology, in vitro cell culture disease models were limited to those cell types that could be harvested from the patient without harm usually dermal fibroblasts from skin biopsies. However, mature dermal fibroblasts alone cannot recapitulate complicated disease processes involving multiple cell types. Using iPS technology, dermal fibroblasts can be de-differentiated into iPS cells. Subsequently, the iPS cells can be directed to differentiate into the cell type most beneficial for modeling a particular disease process. Advances in the production of iPS cells have found that the earliest pluripotent stage of the derivation process can be eliminated under certain circumstances. For instance, dermal fibroblasts have been directly differentiated into dopaminergic neurons by viral co-transduction of forebrain transcriptional regulators (Brn2, Myt1l, Zic1, Olig2, and Ascl1) in the presence of media containing neuronal survival factors [brain-derived neurotrophic factor, neurotrophin-3 (NT3), and glial-conditioned media] (Qiang et al, 2011). Additionally, dermal fibroblasts have been directly differentiated into cardiomyocyte-like cells using the transcription factors Gata4, Mef2c, and Tb5 (Ieda et al, 2010). Regardless of the derivation process, once the cell type of interest is generated, the phenotype central to the disease process can be readily studied. In addition, compounds can be screened for therapeutic benefit and environmental toxins can be screened as potential contributors to the disease. Thus far, iPS cells have generated valuable in vitro models for many neurodegenerative (including Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis), hematologic (including Fanconi's anemia and dyskeratosis congenital), and cardiac disorders (most notably the long QT syndrome) (Park et al, 2008). iPS cells from patients with the long QT syndrome are particularly interesting as they may provide an excellent platform for rapidly screening drugs for a common, lethal side effect (Zwi et al, 2009; Malan et al, 2011; Tiscornia et al, 2011). The development of patient-specific iPS cells for in vitro disease modeling will determine the potential for these cells to differentiate into desired cell lineages, serve as models for investigating the mechanisms underlying disease pathophysiology, and serve as tools for future preclinical drug screening and toxicology studies.

Despite substantial improvements in therapy, cardiovascular disease remains the leading cause of death in the industrialized world. Therefore, there is a particular interest in cardiovascular regenerative therapies. The potential of diverse progenitor cells to repair damaged heart tissue includes replacement (tissue transplant), restoration (activation of resident cardiac progenitor cells, paracrine effects), and regeneration (stem cell engraftment forming new myocytes) (Codina et al, 2010). It is unclear whether the heart contains resident stem cells. However, experiments show that bone marrow mononuclear cells (BMCs) can repair myocardial damage, reduce left ventricular remodeling, and improve heart function by myocardial regeneration (Hakuno et al, 2002; Amado et al, 2005; Dai et al, 2005; Schneider et al, 2008). The regenerative capacity of human heart tissue was further supported by the detection of the renewal of human cardiomyocytes (1% annually at the age of 25) by analysis of carbon-14 integration into human cardiomyocyte DNA (Bergmann et al, 2009). It is not clear whether cardiomyocyte renewal is derived from resident adult stem cells, cardiomyocyte duplication, or homing of non-myocardial progenitor cells. Bone marrow cells home to the injured myocardium as shown by Y chromosome-positive BMCs in female recipients (Deb et al, 2003). On the basis of these promising results, clinical trials in patients with ischemic heart disease have been initiated primarily using bone marrow-derived cells. However, these small trials have shown controversial results. This is likely due to a lack of standardization for cell harvesting and delivery procedures. This highlights the need for a better understanding of the basic mechanisms underlying stem cell isolation and homing prior to clinical implementation.

Although stem cells have the capacity to differentiate into neurons, oligodendrocytes, and astrocytes, novel clinical stem cellbased therapies for central and peripheral nervous system diseases have yet to be realized. It is widely hoped that transplantation of stem cells will provide effective therapy for Parkinson's disease, Alzheimer's disease, Huntington's Disease, amyloid lateral sclerosis, spinal cord injury, and stroke. Several encouraging animal studies have shown that stem cells can rescue some degree of neurological function after injury (Daniela et al, 2007; Hu et al, 2010; Shimada and Spees, 2011). Currently, a number of clinical trials have been performed and are ongoing.

Dental stem cells could potentially repair damaged tooth tissues such as dentin, periodontal ligament, and dental pulp (Gronthos et al, 2002; Ohazama et al, 2004; Jo et al, 2007; Ikeda et al, 2009; Balic et al, 2010; Volponi et al, 2010). Moreover, as the behavior of dental stem cells is similar to MSCs, dental stem cells could also be used to facilitate the repair of non-dental tissues such as bone and nerves (Huang et al, 2009; Takahashi et al, 2010). Several populations of cells with stem cell properties have been isolated from different parts of the tooth. These include cells from the pulp of both exfoliated (children's) and adult teeth, the periodontal ligament that links the tooth root with the bone, the tips of developing roots, and the tissue that surrounds the unerupted tooth (dental follicle) (Bluteau et al, 2008). These cells probably share a common lineage from neural crest cells, and all have generic mesenchymal stem cell-like properties, including expression of marker genes and differentiation into mesenchymal cells in vitro and in vivo (Bluteau et al, 2008). different cell populations do, however, differ in certain aspects of their growth rate in culture, marker gene expression, and cell differentiation. However, the extent to which these differences can be attributed to tissue of origin, function, or culture conditions remains unclear.

There are several issues determining the long-term outcome of stem cellbased therapies, including improvements in the survival, engraftment, proliferation, and regeneration of transplanted cells. The genomic and epigenetic integrity of cell lines that have been manipulated in vitro prior to transplantation play a pivotal role in the survival and clinical benefit of stem cell therapy. Although stem cells possess extensive replicative capacity, immune rejection of donor cells by the host immune system post-transplantation is a primary concern (Negro et al, 2012). Recent studies have shown that the majority of donor cell death occurs in the first hours to days after transplantation, which limits the efficacy and therapeutic potential of stem cellbased therapies (Robey et al, 2008).

Although mouse and human ES cells have traditionally been classified as being immune privileged, a recent study used in vivo, whole-animal, live cell-tracing techniques to demonstrate that human ES cells are rapidly rejected following transplantation into immunocompetent mice (Swijnenburg et al, 2008). Treatment of ES cell-derived vascular progenitor cells with inter-feron (to upregulate major histocompatibility complex (MHC) class I expression) or in vivo ablation of natural killer (NK) cells led to enhanced progenitor cell survival after transplantation into a syngeneic murine ischemic hindlimb model. This suggests that MHC class I-dependent, NK cell-mediated elimination is a major determinant of graft survivability (Ma et al, 2010). Given the risk of rejection, it is likely that initial therapeutic attempts using either ES or iPS cells will require adjunctive immunosuppressive therapy. Immunosuppressive therapy, however, puts the patient at risk of infection as well as drug-specific adverse reactions. As such, determining the mechanisms regulating donor graft tolerance by the host will be crucial for advancing the clinical application of stem cellbased therapies.

An alternative strategy to avoid immune rejection could employ so-called gene editing. Using this technique, the stem cell genome is manipulated ex vivo to correct the underlying genetic defect prior to transplantation. Additionally, stem cell immunologic markers could be manipulated to evade the host immune response. Two recent papers offer alternative methods for gene editing. Soldner et al (2011) used zinc finger nuclease to correct the genetic defect in iPS cells from patients with Parkinson's disease because of a mutation in the -Synuclein (-SYN) gene. Liu et al (2011) used helper-dependent adenoviral vectors (HDAdV) to correct the mutation in the Lamin A (LMNA) gene in iPS cells derived from patients with HutchinsonGilford Progeria (HGP), a syndrome of premature aging. Cells from patients with HGP have dysmorphic nuclei and increased levels of progerin protein. The cellular phenotype is especially pronounced in mature, differentiated cells. Using highly efficient helper-dependent adenoviral vectors containing wild-type sequences, they were able to use homologous recombination to correct two different Lamin A mutations. After genetic correction, the diseased cellular phenotype was reversed even after differentiation into mature smooth muscle cells. In addition to the potential therapeutic benefit, gene editing could generate appropriate controls for in vitro studies.

Finally, there are multiple safety and toxicity concerns regarding the transplantation, engraftment, and long-term survival of stem cells. Donor stem cells that manage to escape immune rejection may later become oncogenic because of their unlimited capacity to replicate (Amariglio et al, 2009). Thus, ES and iPS cells may need to be directed into a more mature cell type prior to transplantation to minimize this risk. Additionally, generation of ES and iPS cells harboring an inducible kill-switch may prevent uncontrolled growth of these cells and/or their derivatives. In two ongoing human trials with ES cells, both companies have provided evidence from animal studies that these cells will not form teratomas. However, this issue has not been thoroughly examined, and enrolled patients will need to be monitored closely for this potentially lethal side effect.

In addition to the previously mentioned technical issues, the use of ES cells raises social and ethical concerns. In the past, these concerns have limited federal funding and thwarted the progress of this very important research. Because funding limitations may be reinstituted in the future, ES cell technology is being less aggressively pursued and young researchers are shying away from the field.

Read the original post:
The benefits and risks of stem cell technology - PMC

Posted in Wisconsin Stem Cells | Comments Off on The benefits and risks of stem cell technology – PMC

Hunters, Hearing Loss, and New Tech to Fix it – MeatEater

Posted: June 13, 2022 at 2:29 am

If ever there was an epidemic in the hunting community, itd be hearing loss. When target practice is a way of life, its easy to become blas about earmuffs. When that buck is about to get over the ridge, jamming in ear plugs is the last of our worries. For those reasons, most of us will go through the rest of our lives unable to hear pheasants cackle and elk bugle as well as we should.

It doesnt have to be that way. You can take steps now to stem the loss of auditory ability with simple, cheap solutions or technologically advanced ones.

Dr. Grace Sturdivant is an evangelist for saving hunters eardrums. Starting her career as a Vanderbilt-trained Doctor of Audiology, she spent years researching, diagnosing, and treating hearing loss. Eventually, however, she decided to take a more proactive approach.

I've shifted from the traditional academic medical setting practice, Sturdivant told MeatEater. I want to prevent and delay the problems that I spent so many years treating in people who were debilitated by this problem that was largely preventable. And they acquired this problem by doing these worthwhile things that they love.

Raised in a Mississippian hunting family, she wasnt about to tell shooters to stop shooting: I want you to continue doing what you're doing. I just want to give you some tools to do it safer.

That desire led to the founding of her company, OtoPro Technologies. Through this business, Sturdivant advocates for ear protection while serving as a consultant for shooters, musicians, pilots, builders, machine operators, and others who deal in loud noises for work or pleasure.

I love music. I don't want tell people Don't go stand front and center at your favorite show. Go do it. But let me give you some really cool specially filtered, small, tiny earplugs so you can take the edge off and your ears won't be ringing all night, she said. That's the whole mission. Even if it comes down to me just educating you on how to wear the foam ear plugs correctly, thats cool.

How Do Guns Affect Hearing?

Sturdivant cites a University of Wisconsin study that found men aged 48 to 92 who hunted regularly were more likely to experience high-frequency hearing loss, a risk that increased 7% for every five years a man had been hunting. Of the participants surveyed, 38% of recreational shooters and 95% said they never wore hearing protection while shooting in the last year.

Another more recent study found that the usage of hearing protection while shooting has increased in recent years, but auditory loss is still a big problem. Still, there are plenty of factors to consider in order to mitigate.

High-intensity impulse sounds will permanently damage delicate cochlear structures, and thus individuals who shoot firearms are at a higher risk of bilateral, high-frequency, noise-induced hearing loss (NIHL) than peer groups who do not shoot, the studys authors wrote. In this article, we describe several factors that influence the risk of NIHL, including the use of a muzzle brake, the number of shots fired, the distance between shooters, the shooting environment, the choice of ammunition, the use of a suppressor, and hearing protection fit and use.

But whats really happening when we pull the trigger that makes our ears hurt and function poorly as a result?

When you think about decibels, think about them in terms of sound pressure level, which is exactly what a decibel is, Sturdivant said. And with a gunshot you're typically at around 150 decibels sound pressure level. And when that amount of sound pressure hits your hearing nerve, it's hitting those hair fibers that are just these tiny, delicate hair fibers that send the sound to the brain. It's hitting them really hard and really fast, like an impulse impact. And so the amount that those hair cells are able to withstand instantly from a 150 decibel sound pressure level is not very much. So, oftentimes, you'll see instant damage or at least instant weakening at that level.

In contrast, she said, you could withstand a lower decibel level for much longer. But even an 80-decibel noise for a long duration will weaken those microscopic hair fibers in your ears. Those organs can recover and regenerate, but you need to allow them time to do so after exposure to loud noises. If you let the problem go on long enough, however, you may be faced with even worse issues.

Of course, not only the communication difficulties and just the frustration of not being able to hear; there's really more to it than that, Sturdivant said. And a large reason why I started this business was because of all the connections that we see in the area of cognitive decline and dementia with hearing loss. Because we actually hear with our brains and when those hair cells are able to send the signal in a healthy way to the brain to be processed, it's stimulating some very specific areas of the brain. And when those areas are not stimulated, we see an earlier onset and a faster rate of cognitive decline with various forms of dementia.

Sturdivant said that most people wait seven to 10 years to do anything about their hearing loss. That can allow cognitive decline to take hold sooner than it would otherwise. Like with most medicine, an ounce of prevention is worth a pound of cure.

How to Protect Your Ears While Shooting

Hearing protection technology has advanced rapidly in recent years, especially from military contractors and European firms. While there are many great offerings in the consumer hunting market already, theres a lot more ultra-modern tech you probably havent even heard about yet. Sturdivant stays highly up to date on all the bleeding-edge ear care gadgets available worldwide.

One of the biggest developments recently becoming widely available to consumers is simple and affordable custom fittings.

Custom is very important because we want a tight air seal of your ear, she said. If there's any air leakage, sound waves are passing through.

Its also a major boon if, say, you got punched in the head in high school or otherwise damaged your ear canals in sports or accidentsplacing you among the minority in regard to one-size-fits-most ear plugs.

Some new offerings also allow air passage to help defeat that pressurized-airplane-cabin, stuffed-up feeling you get from air-tight ear plugs.

There's lots of passive filters for hunting and shooting. This is awesome because it allows the most in and it allows some air exchange. When you first wear it, you wonder like, is this really fitting my ear? Because I can feel air, which is strange. But that's the whole point, Sturdivant said. Theres a sound pressure membrane that vibrates to let the sound pass through and then it stiffens up at 95-decibel sound pressure level. So, it's an all-or-nothing filter.

For hunters who may have already beat the hell out of their eardrums, a favorite feature is ambient microphones built into the earplugs that amplify the surrounding noise, making you still able (or even more able) to hear a buck snort or goose wings whoosh. At the gun range, that means you dont need to take off your headphones and yank out your ear plugs between each target session in order to register what your friends are hollering at you. Ambient noise will sound practically normal, but the plugs immediately block out the concussion of a gun blast.

Some of the higher-end models offer Bluetooth streaming capability, so you can play your favorite podcast while plinking at targets or running the circular saw. Some of these also offer wind noise reduction features, or the ability to adjust the volume level for both ears individually. There are solutions for every situation and budget, from OtoPros new proprietary, universal passive filter ear plugs to $300 custom rigs.

Or course, none of this is meant to say that you cant protect your ears well with items you likely already own (or could acquire painlessly). Sturdivant says that the cheap foam ear plugs still make a big difference, so long as theyre inserted correctly. Pinch and roll the tip up tightly then push it in far enough that none of the material is extending outside your ears, creating an air-tight seal against sound. Pair that with an affordable pair of Caldwell earmuffs and youre a long ways toward saving your ears for later in life.

Sturdivant said that she is seeing a tide change in the way hunters view protecting themselves and others. Folks over 60 tend to think the damage is done, she said, but younger generationsthose that grew up with mandatory seatbelt laws, for exampleare beginning to be more proactive and invest in preventative measures. That may be the most true for parents introducing their kids to hunting and shooting. If you start young enough with good ear protection, theres no reason why a person should ever lose their auditory abilities. Sturdivant hopes to keep that trend growing and encourages every hunter and shooter to share the same message with their family and friends.

My job is to motivate you, to protect your hearing. Your job is to become damned and determined to be like, I'm going to get over this hump and this is going to become my new normal. And then it becomes second nature, she concluded. Hearing protection just needs to be another piece of essential hunter safety gear.

Original post:
Hunters, Hearing Loss, and New Tech to Fix it - MeatEater

Posted in Wisconsin Stem Cells | Comments Off on Hunters, Hearing Loss, and New Tech to Fix it – MeatEater

UB-led study presents critical step forward in understanding Parkinson’s disease and how to treat it – University at Buffalo

Posted: June 13, 2022 at 2:27 am

BUFFALO, N.Y. A new study led by a researcher in the Jacobs School of Medicine and Biomedical Sciences at the University at Buffalo has important implications for developing future treatments for Parkinsons disease (PD), a progressive nervous system disorder that affects movement and often includes tremors.

In this study, we find a method to differentiate human induced pluripotent stem cells (iPSCs) to A9 dopamine neurons (A9 DA), which are lost in Parkinsons disease, says Jian Feng, PhD, professor of physiology and biophysics in the Jacobs School and the senior author on the paper published May 24 in Molecular Psychiatry.

These neurons are pacemakers that continuously fire action potentials regardless of excitatory inputs from other neurons, he adds. Their pacemaking property is very important to their function and underlies their vulnerability in Parkinsons disease.

This exciting breakthrough is a critical step forward in efforts to better understand Parkinsons disease and how to treat it, says Allison Brashear, MD, UBs vice president for health sciences and dean of the Jacobs School. Jian Feng and his team are to be commended for their innovation and resolve.

Feng explains there are many different types of dopamine neurons in the human brain, and each type is responsible for different brain functions.

Nigral dopamine neurons, also known as the A9 DA neurons, are responsible for controlling voluntary movements. The loss of these neurons causes the movement symptoms of Parkinsons disease, he says.

Scientists have been trying hard to generate these neurons from human pluripotent stem cells to study Parkinsons disease and develop better therapies, Feng says. We have succeeded in making A9 dopamine neurons from human induced pluripotent stem cells. It means that we can now generate these neurons from any PD patients to study their disease.

Feng notes that A9 DA neurons are probably the largest cells in the human body. Their volume is about four times the volume of a mature human egg.

Over 99 percent of the volume is contributed by their extremely extensive axon branches. The total length of axon branches of a single A9 DA neuron is about 4.5 meters, he says. The cell is like the water supply system in a city, with a relatively small plant and hundreds of miles of water pipes going to each building.

In addition to their unique morphology, the A9 DA neurons are pacemakers they fire action potentials continuously regardless of synaptic input.

They depend on Ca2+ channels to maintain the pacemaking activities. Thus, the cells need to deal with a lot of stress from handling Ca2+ and dopamine, Feng says. These unique features of A9 DA neurons make them vulnerable. Lots of efforts are being directed at understanding these vulnerabilities, with the hope of finding a way to arrest or prevent their loss in Parkinsons disease.

Pacemaking is an important feature and vulnerability of A9 DA neurons. Now that we can generate A9 DA pacemakers from any patient, it is possible to use these neurons to screen for compounds that may protect their loss in PD, Feng notes. It is also possible to test whether these cells are a better candidate for transplantation therapy of PD.

To differentiate human iPSCs to A9 DA neurons, the researchers tried to mimic what happens in embryonic development, in which the cells secrete proteins called morphogens to signal to each other their correct position and destiny in the embryo.

Feng notes the A9 DA neurons are in the ventral part of the midbrain in development.

Thus, we differentiate the human iPSCs in three stages, each with different chemicals to mimic the developmental process, he says. The challenge is to identify the correct concentration, duration, and treatment window of each chemical.

The combination of this painstaking work, which is based on previous work by many others in the field, makes it possible for us to generate A9 DA neurons, Feng adds.

Feng points out there are a number of roadblocks to studying Parkinsons disease, but that significant progress is being made.

There is no objective diagnostic test of Parkinsons disease, and when PD is diagnosed by clinical symptoms, it is already too late. The loss of nigral DA neurons has already been going on for at least a decade, he says.

There was previously no way to make human dopamine neurons from a PD patient so we could study these neurons to find out what goes wrong.

Scientists have been using animal models and human cell lines to study Parkinsons disease, but these systems are inadequate in their ability to reflect the situation in human nigral DA neurons, Feng says.

Just within the past 15 years, PD research has been transformed by the ability to make patient-specific dopamine neurons that are increasingly similar to their counterparts in the brain of a PD patient.

Houbo Jiang, PhD, research scientist in the Department of Physiology and Biophysics, and Hong Li, PhD, a former postdoctoral associate in the Department of Physiology and Biophysics, are co first-authors on the paper.

Other co-authors on the study are: Hanqin Li, PhD, a graduate of the doctoral program in neuroscience and currently a postdoctoral fellow at University of California, Berkeley; Li Li, a trainee in UBs doctoral program in neuroscience; and Zhen Yan, PhD, SUNY Distinguished Professor of physiology and biophysics.

The study was funded by the Department of Veterans Affairs, National Institutes of Health and by New York State Stem Cell Science (NYSTEM).

Read more from the original source:
UB-led study presents critical step forward in understanding Parkinson's disease and how to treat it - University at Buffalo

Posted in California Stem Cells | Comments Off on UB-led study presents critical step forward in understanding Parkinson’s disease and how to treat it – University at Buffalo

Dr. Andrew Weil Receives Integrative Healthcare Leader Award – University of Arizona

Posted: June 13, 2022 at 2:26 am

A world-renowned author and expert in the field of integrative medicine, Andrew T. Weil, MD, founder and director of the Andrew Weil Center for Integrative Medicine at the University of Arizona College of Medicine Tucson, was recognized recently with the top award given by the Integrative Healthcare Symposium.

Im honored to receive the symposiums 2022 Leadership Award, Dr. Weil said. Integrative health care is the way of the future, and the University of Arizona Health Sciences is leading that way. Our Center for Integrative Medicine sets the standard for training physicians, nurses, and allied health professionals to provide this kind of care in order to improve health outcomes and lower health care costs.

The annual award was presented to Dr. Weil, who also gave the symposiums keynote address on The Evolution of Integrative Medicine, earlier this year in New York City.

Presenting the award to Dr. Weil was Aly Cohen, MD, a 2014 graduate of the centers Fellowship in Integrative Medicine, founder of Integrative Rheumatology Associates and The Smart Human LLC, and co-author of the book, Non-Toxic: Guide to Living Healthy in a Chemical World. Dr. Cohen is the guest on the latest Body of Wonder podcast with Dr. Weil and center executive director Victoria Maizes, MD.

In lauding Dr. Weil, Dr. Cohen told the tale of how his car broke down in Tucson in the 1970s while he was traveling across country, he fell in love with the desert and stayed. Well, lucky for us. Combining the number of patients reached through fellowship graduates, health coaches, residents in training, medical students and researchers, some 8 million patients are guided by your teachings and are quite grateful your car broke down in Tucson many moons ago, she said.

Commenting on his 15 books, Dr. Cohen said, In an era of plentiful, often radical guidebooks and scary health news flashes, he has provided an oasis for balance and common sense for readers. Dr. Weil, your long and brave history of challenging the status quo of Western medicine and your ongoing mission to educate others through print, social media, television and lectures is a testament to the beautiful and rich journey youve taken.

After graduating high school in Philadelphia in 1959, Dr. Weil won an American Association for the United Nations scholarship that allowed him to travel abroad for a year where he lived with families in India, Thailand and Greece. Upon his return, he did his undergraduate studies in biology and ethnobotany at Harvard University and earned his medical degree from Harvard in 1968.

He interned at Mount Zion Hospital in San Francisco, then took a post with the National Institute of Mental Health before writing his first book, The Natural Mind. As a fellow of the Institute of Current World Affairs from 1971-75, Dr. Weil traveled widely in the Americas and Africa, studying medicinal plants and alternative treatment methods for disease. From 1971-84, he also was on the research staff of the Harvard Botanical Museum and conducted investigations of medicinal and psychoactive plants.

Dr. Weil founded the Program in Integrative Medicine in 1994 at the UArizona College of Medicine Tucson. Fourteen years later, the program by then a division in the colleges Department of Medicine was designated a center of excellence by the Arizona Board of Regents. In 2019, the center was renamed as the Andrew Weil Center for Integrative Medicine. The center broke ground on its new $23 million, donor-funded building March 16.

A UArizona clinical professor of medicine and public health and the Lovell-Jones Endowed Chair in Integrative Medicine, Dr. Weil also is editorial director of DrWeil.com, a leading online resource for healthy living based on a philosophy of integrative medicine, and a founder and partner of True Food Kitchen restaurants, whose recipes are the basis for his New York Times best-selling book, True Food.

Read more from the original source:
Dr. Andrew Weil Receives Integrative Healthcare Leader Award - University of Arizona

Posted in Integrative Medicine | Comments Off on Dr. Andrew Weil Receives Integrative Healthcare Leader Award – University of Arizona

Should You Avoid Nightshades? A Look at the Research – Forks Over Knives

Posted: June 13, 2022 at 2:26 am

While the name might seem ominous and call to mind the contents of a sorcerers cauldron, nightshades are among the most common fruits and vegetables, and you likely already have some in your kitchen. So, what are nightshades, and are they good for you?

There are 2,500 species of flowering plants known as nightshades within the Solanaceae plant family.

The Summer 2022Magazine Is Here!

Our gorgeous bookazine features expert tips, inspiration, and fresh, healthy recipes for summer.

Some of the most commonly consumed nightshades include:

Edible nightshades are some of the most nutritious foods around and have been consumed as part of healthy diets for centuries, says Sharon Palmer, MSFS, RDN.

Hundreds of studies have found benefits from eating these foods. In particular, tomatoes have garnished a great deal of research documenting benefits, such as antioxidant, anti-inflammatory benefits and reduced risks of diseases such as prostate cancer and heart disease, says Palmer. They have also been linked to skin and bone protection.

Meanwhile capsaicin in chile peppers may promote hair growth and reduce your cardiovascular and cancer risk. And potatoes are loaded with mood-regulating carbohydrates and muscle-building protein.

Members of the Solanaceae plant family contain alkaloids, including solanine, a natural insecticide. Solanines in belladonna, the so-called deadly nightshade, can cause delirium, hallucinations, and even death. However, the nightshades we commonly consume contain nowhere near high enough levels to cause similar harm.

There is not enough scientific support documenting that people need to avoid nightshades due to alkaloid content, says Palmer.

That being said, potato sprouts and areas of the potato that have turned green from sun exposure contain higher concentration of solanine and, therefore, should be avoided. Symptoms of solanine poisoning include abdominal pain, vomiting, diarrhea, fever or hypothermia, headaches, and a slow pulse or breathing.

Search the internet for the word nightshades, and youre bound to stumble on plenty of articles warning about inflammation and arthritis pain. But no research has turned up evidence that nightshades affect the joints.

There is a lot of urban legend and misinformation about nightshades being perpetuated over the internet and social media, says Palmer. Some people believe that they should avoid nightshades to reduce inflammation for arthritis benefits. However, studies have found that many nightshade vegetables reduce inflammation levels in the body.

Its worth noting that the Arthritis Foundation put nightshade vegetables, namely bell peppers, on its list of Best Vegetables for Arthritis. Red and yellow bell peppers contain the carotenoid beta-cryptoxanthin, which could reduce your risk of developing inflammatory disorders like rheumatoid arthritis. Additionally, tomatoes and peppers are excellent sources of bone- and cartilage-preserving vitamin C, with a single bell pepper containing more than 150% of the Food and Nutrition Boards daily recommended amount. Eggplants, meanwhile, are rich in anti-inflammatory anthocyanins as well as the essential trace element manganese, which is important to bone formation.

The scientific evidence [regarding nightshades and inflammation] isnt very strong at this time, says triple board-certified rheumatologist Micah Yu, MD, who also practices integrative medicine. Maybe in 10, 20 years, well have more evidence.

Yu notes that theres no test to determine whether someone might have a sensitivity to nightshades. If you suspect nightshades are an issue for you, he suggests keeping a food diary and seeing whether certain foods correspond with your inflammatory symptoms or other adverse reactions. You can try avoiding a food to see if symptoms improve, then reintroducing the food to see if symptoms return. If they return, its reasonable to continue avoiding the food, and consult with a registered dietitian.

At least two studies have suggested potatoes could aggravate inflammatory bowel disease. Both were performed using mice, not humans.

In one study from 2002, researchers isolated solanine and the glycoalkaloid chaconine, present in potatoes, to test intestinal permeability and function. They concluded that levels of solanine and chaconine typically found in potatoes can adversely affect a mammals intestine and exacerbate IBD.

In a 2010 study, mice were fed deep-fried potato skins. Researchers found that deep-frying the potato skins increased glycoalkaloid content and that glycoalkaloid consumption significantly aggravated intestinal inflammation in mice representing two models mimicking human IBD (interleukin 10 gene deficiency and dextran sodium sulfate-induced colitis).

But the data are limited, says Vanita Rahman, MD, clinic director of Barnard Medical Center. We know animal studies dont always translate into meaningful results in humans, so its really hard to draw any conclusions about human health, as far as inflammatory bowel disease.

Before eliminating nightshades altogether, Rahman recommends talking to a health care provider and exploring whether anything else could be contributing to IBD symptoms. Keep in mind that certain nightshadespotatoes and eggplantsare rich in fiber, which has been linked to a reduced risk of developing IBD and greater quality of life in patients with ulcerative colitis.

The bottom line is [nightshades] really are nutritious vegetables that contain a lot of important nutrients for us, says Rahman. They have a lot of health benefits. So, most people should consume them in ways that they find enjoyable.

There are plenty of opportunities to reap the health benefits of these delightful fruits and vegetables. Check out these roundups of favorite recipes from Forks Over Knives to get you started.

From baba ghanoush flatbreads to vegan eggplant parm to ratatouille, these recipes showcase eggplants melt-in-your-mouth deliciousness.

Its easy to see why potatoes are so universally beloved. Transform humble taters into impressive entrees, savory side dishes, delectable vegan cheese sauce, wholesome homemade bread, and more.

Brighten your kitchen and delight your taste buds with these colorful and creative bell pepper recipes.

Harness the ripe, juicy goodness of fresh tomatoes for full-flavored soups, bruschetta, grain bowls, marinara, and more.

For more guidance in healthy cooking, check out Forks Meal Planner, FOKs easy weekly meal-planning tool to keep you on a plant-based path. To learn more about a whole-food, plant-based diet, visit our Plant-Based Primer.

See original here:
Should You Avoid Nightshades? A Look at the Research - Forks Over Knives

Posted in Integrative Medicine | Comments Off on Should You Avoid Nightshades? A Look at the Research – Forks Over Knives

Page 392«..1020..391392393394..400410..»