Podcasts about pi3 kinase

  • 15PODCASTS
  • 18EPISODES
  • 26mAVG DURATION
  • ?INFREQUENT EPISODES
  • May 17, 2023LATEST

POPULARITY

20172018201920202021202220232024


Best podcasts about pi3 kinase

Latest podcast episodes about pi3 kinase

JCO Precision Oncology Conversations
Larotrectinib for TRK Fusion - Positive Lung Cancer

JCO Precision Oncology Conversations

Play Episode Listen Later May 17, 2023 28:56


JCO PO author Alexander E. Drilon, MD, shares insights into his article, “Efficacy and Safety of Larotrectinib in Patients With Tropomyosin Receptor Kinase Fusion–Positive Lung Cancers” and the article's findings of the activity of larotrectinib in patients with advanced lung cancer harboring NTRK gene fusions. Host Dr. Rafeh Naqash and Dr. Drilon discuss drug development, testing for fusions, resistance mechanisms, and cancer metastases. Click here to read the article!   TRANSCRIPT Dr. Rafeh Naqash: Hello and welcome to JCO Precision Oncology Conversations, where we bring you engaging conversations with authors of clinically relevant and highly significant JCO PO articles. I am your host, Dr. Rafeh Naqash, Social Media Editor for JCO Precision Oncology, and Assistant Professor at the OU Stephenson Cancer Center.  Today we are excited to be joined by Dr. Alexander Drilon, Chief of the Early Drug Development Service and Medical Oncologist on the Thoracic Oncology Service at the Memorial Sloan Kettering Cancer Center and lead author of the JCO Precision Oncology article “Efficacy and Safety of Larotrectinib in Patients With Tropomyosin Receptor Kinase Fusion–Positive Lung Cancers.” Our guests' disclosures will be linked in the transcript.  Dr. Drilon, welcome to the podcast and thank you for joining us today. We're really excited to be discussing this topic with you.  Dr. Alexander Drilon: It's my pleasure and thank you for the invitation. Dr. Rafeh Naqash: For the sake of this podcast, we will refer to each other using our first names. So, Alex, you've led the development for some of these agents targeting NTRK. So it's really timely that you're coming onto this podcast to not just discuss this very interesting paper that you published in JCO Precision Oncology, but also the general landscape of NTRK. So could you briefly tell us about the history of the drug development process behind NTRK fusions, when it started, how you got involved, and where it stands currently? Dr. Alexander Drilon: Sure. So, as you mentioned, my background is in lung cancer, where when I came on as a fellow, there was a lot of excitement around EGFR and ALK, but then subsequently other oncogene drivers were also discovered and many of them were fusion. So, as we know, ALK in the fuse state is a driver of many tumors, as is ROS1 and RET. And interestingly, NTRK fusions share many similarities with ALK, RET, and ROS1 in that you have an intact kinase domain that's in the three prime position, it's fused to a different gene in the five prime position and basically describes oncogenesis.  And the beautiful thing about NTRK fusions is that they are widely found across many different cancers. And I like to think of these cancers in two major buckets. So there is a bucket for cancers that are rare where we see these NTRK fusions with a very high frequency. And examples here are your secretory carcinomas of the salivary gland and the breast, for example, more congenital fibrosarcoma, where the frequency exceeds 90% in some series, and then there are much more common tumors where the frequency is much lower. So lung cancer is an example where you find it in less than 1% of cases. There are some other tumors like GI cancers also where the frequency is low. And beyond these two major groups, we also see these NTRK fusion-positive cancers occur not just in the adult population, but the pediatric population. All of that thrown together means that it was a really great setup for exploring the activity and safety of targeted therapy in what we call a ‘basket trial' paradigm, where you design a trial and instead of selecting patients based on cancer type, you ignore cancer type and, of course, you accrue by an enrolling alteration, which in this case is the NTRK fusion.  Dr. Rafeh Naqash: Excellent. Thank you for that summary. It's interesting that just yesterday in my phase I clinic, I had an individual who was supposed to go on a certain study, and liquid biopsy came back and showed an NTRK fusion for a very odd presentation of a prostate cancer, which, again, got me thinking about the paper that you published trying to read about NTRK and then this happened and I got thinking about a bunch of other questions. But, for starters, though, from a receptor standpoint and I know you published on this in different journals, could you briefly tell, for the sake of the audience, describe the pathway and the tyrosine kinase signaling and associated resistance pathways that are concurrently acting in a different direction, perhaps, and also discuss briefly from neural development? I know the pathway, the NTRK gene or TRK gene as such is involved in different neuronal signaling aspects. Could you briefly touch on that? Dr. Alexander Drilon: Sure. And thankfully there are a lot of parallels with other things that perhaps some of the listeners are more familiar with. We'll start with the fact that it is a receptor tyrosine kinase, NTRK. It's a gene that encodes a receptor tyrosine kinase just like other receptor tyrosine kinases that may be fused such as ALK, RET, and ROS1. But remember also that other RTKs are EGFR, FGFR, which are also well known. The important thing to remember for NTRK is that you have three different genes, NTRK 1, 2, and 3 that encode three different proteins which are called TRK A, B, and C. And as you intimated, in the non-oncogenic state, these are very important for the development and the maintenance of the nervous system. And in the fused state, of course, similar to other fusions that we spoke about, the chimeric oncoprotein will drive downstream signaling and tumor growth and metastases. And in general, these cancers can be very reliant on downstream signaling in the MAP Kinase pathway but may also on occasion activate other downstream pathways like the PI3 Kinase pathway. Dr. Rafeh Naqash: And I know some of that could potentially play into resistance mechanisms for some of these first or second-generation NTRK inhibitors. From a fusion partner standpoint, the data that I came across that you're very well aware of is different fusion partners, and maybe some have a slightly better prognosis than some other fusions. But, in your practice and in your experience, does it matter what the other fusion partner is if the kinase domain is intact, meaning the signaling for the NTRK gene is intact? Have you seen any differences there from the other fusion partner standpoint? Dr. Alexander Drilon: From a patient-matching perspective, as long as you think the fusion is real, and by that I mean that you look at the report and you're sure the kinase domain is there and you're sure it's in frame, meaning connected well to the five prime partner so that the DNA strand is read through, the five prime partner does not play a major role in my deciding to give a TRK inhibitor or not. I would give anyone with a functional NTRK 1, 2, or 3 fusion a TRK inhibitor. Now, the data on whether or not select fusions do better than others is, I would say, still a little immature and perhaps conditioned by a few things. There are some of the cancers in the first bucket that we talked about, like the secretory carcinomas that harbor a recurrent event such as ETV6 NTRK3. And those cancers, in my experience in clinic, patients with those tumors can be on a TRK inhibitor for a very long time. And it's unclear if that's because of the exact fusion event or if it's because of the cancer type that might be more, say, genomically naïve compared to a gastrointestinal tumor, like a colorectal cancer with an NTRK fusion. So I hesitate to say that there are very strong and convincing data that if you have a particular five prime partner, you'll absolutely do better or worse. So, in the interim, I think the most important piece is just making sure that the event is real and actionable, and if it is, then you can give a TRK inhibitor. Dr. Rafeh Naqash: Thank you so much. I totally agree. And I think, for the sake of our listeners, as we see more and more sequencing being done on patients with cancer in the advanced stage setting especially, it's important to keep in mind when you have something that you can act on that has an actionable target that is FDA approved, then it's important to give the patient that option, especially in rare fusion events such as NTRK or TRK.   Now, you've touched upon this in your paper, but before we go into the details of the paper, specifically, I wanted you to briefly talk about the testing mechanisms which are important for some of these fusions and play into, for example, ROS1 ALK fusions also. Could you tell us what are the most appropriate ways to test for these fusions in patients harboring cancers, both from a tissue standpoint and from a blood-based assay standpoint? Dr. Alexander Drilon: This is a great question because if you don't have a test that's optimally poised to pick up an NTRK fusion, then you can't act on it. And a patient who would have benefited very durably from a TRK inhibitor won't get access to it. So there are different ways of testing for NTRK fusions, and I like to think of the central dogma here where you have DNA becomes RNA becomes protein because that really helps anchor the different types of assays that you might use. We commonly use next-generation sequencing of DNA, but even if you have a very good next-generation sequencing assay, that does have its limitations because there are some fusions that are structurally just difficult to pick up even with a great DNA-based NGS assay.  And for that reason, we and others have found that in tumors that have an equivocal NTRK fusion, or perhaps where you didn't find something but you really suspect that you missed something, particularly in cases where, historically, like congenital fibrosarcoma where you know there's a very good likelihood of finding NTRK fusion, we then reach for an RNA-based assay because at the RNA level, you've removed things like the intra-DNA based capture challenging. And so I think that from a nucleic acid standpoint, leveraging a test that looks both at DNA and RNA, maximizes the likelihood of finding this fusion. And just remember that there are different NGS assays in terms of the approach to design and some might be more Amplicon-based and that's less optimal, but the hybrid-capture-based ones tend to be better. The DNA and RNA tests can be done on tumors, and in blood, you could do a liquid biopsy. It's very hard to fish out RNA in blood given the current technology so we're still limited to circulating tumor DNA which shares the liabilities of doing DNA testing on a tumor sample. But if you find it and it looks real, then it's certainly actionable even if you detect an NTRK fusion with a liquid biopsy.  Now going back to the central dogma there, the third piece which we haven't touched on is protein. And there have been many papers published now on the utility of immunohistochemistry, and this helps you confirm that the TRK A, B, and C proteins are actually expressed. And what tends to happen is in many fusions, the chimeric oncoproteins strongly express as TRK A, B, and C that helps provide a complementary test or assay that confirms that you're dealing with something that is actionable.  So that is a very contemporary approach and a very thorough approach to looking for these NTRK fusions where you do DNA and RNA if possible. And if you still have questions, ask your pathologist to see if they can do Pan-TRK IHC. But depending on the resource environment that you're in, there are older tests like FISH which we use for ALK that can also find these fusions. RT-PCR which only finds particular events, these can detect NTRK fusions but really don't have the breadth and comprehensiveness as the other assays that we discussed like NGS.  Dr. Rafeh Naqash: Thank you so much, Alex, for that amazing summary of all the methods that potentially could help detect this rare but important event. From a therapeutic standpoint, now, taking a deeper dive into your very interesting JCO Precision Oncology paper that looked at larotrectinib data from a pooled analysis of two trials, a phase II and a phase I. Could you tell us a little background about these two trials, the patient population and what kind of data were you trying to evaluate? And then we can discuss some of the interesting results that you showcase in the paper. Dr. Alexander Drilon: It really helps as a background to realize that the initial approach to this was really on a basket trial where the programs for larotrectinib, which is a selective TRK A, B, and C inhibitor, and the other drug entrectinib, which inhibits ROS1 in addition to TRK, really accrued pediatric and adult cancers with NTRK fusions. And this paper pulls out the lung cancer subset and we'll discuss that in detail. But before getting into that, it's important to know that in the tumor agnostic data set of all patients with an NTRK fusion of any type, larotrectinib achieved a response rate of approximately 80%, entrectinib of approaching 60%, and disease control was durable with a median PFS for larotrectinib of approximately 28 months, and with entrectinib numerically, the number was lower at 11 months.   So with that background, this paper in JCO PO, in the interest of featuring the activity for lung cancers with NTRK fusions, pulled out 20 patients with NTRK fusion-positive lung cancers. And the punchline is that the activity was pretty comparable to that seen with a bigger data set. So the objective response rate was 73% and many patients had a partial response, 67% of the cases, 7% had a complete response, and really only a minority had primary progressive disease, 1 patient out of the 15 evaluable patients. These responses and clinical benefit overall were durable and the median duration of response was almost 34 months, with a median progression-free survival of almost 35 and a half months and an overall survival median of 40.7 months.  And just to talk about how that stacks up compared to other targeted therapies, this certainly is in the ballpark of some of the best ALK inhibitors that we have for ALK fusion-positive lung cancer. It's also comparable to osimertinib for EGFR mutant lung cancer. So we can confidently view TRK inhibition in lung cancers with NTRK fusions as a highly-active therapy.  Dr. Rafeh Naqash: Absolutely. I think you touched upon this earlier where in your cohort at least 50% of patients had central nervous system involvement, and it looks like larotrectinib does have CNS activity and benefit. Could you speak to the differences between potential entrectinib and larotrectinib from a CNS efficacy standpoint? And the second part of that question was going to be when you identify this fusion in patients, for example, with lung cancer, now, since TRK does have a role in neuronal development, do you think there is a role for closer CNS monitoring in these patients if they have not had brain metastasis identified because of the fact that they have an NTRK fusion? Is there some predilection for CNS involvement from a metastasis standpoint? It's just something that I've been thinking of over the last couple of days after I saw my patient who does have CNS involvement but with prostate cancer, which I have not seen in the phase I setting in all the prostate patients that I've come across. So what are your thoughts on that?  Dr. Alexander Drilon: These are great questions. In lung cancers with NTRK fusions, there is a proclivity for metastasis to the CNS. And thankfully, both of these TKIs, larotrectinib and entrectinib, do have coverage of the CNS. Now, from a design perspective, the initial thought was perhaps entrechtinib was more CNS-penetrant. But if you look at the overall response rates in patients with brain metastases and the intracranial response rates where you have patients with target lesions in the brain that you're able to measure; if you look across the aisle, entrectinib and larotrectinib have comparable results, with the objective response rate being in the order of 50% to 60% and the intracranial response rate being also in the order of about 50% to 60%. So at the end of the day, it appears as if, despite the previous hypothesis that maybe one drug would work better in the CNS than the other, we're seeing equally good effects with both drugs.  For the second question you asked, it's also a very interesting question because, like you mentioned, the TRK receptors play a role in nervous system development. But we have not observed a much higher frequency of CNS metastases in NTRK fusion-positive lung cancers or cancers in general that I know of, compared to cancers that are wild type for an oncogene or have other oncogenes. So what's more important really to think about when you sort of chew on the fact that these TRK inhibitors are involved in nervous system development are the potential side effects that you may see in patients that you treat with these TRK inhibitors. Dr. Rafeh Naqash: Absolutely. Now, from the therapy standpoint that you discuss here, duration of responses, objective responses that you saw in your analysis were very impressive for these patients with lung cancer. In your clinical practice if you see a lung cancer patient with this fusion and you treat them with larotrectinib or entrectinib, and they have, let's say, de novo CNS metastases that are asymptomatic, do you generally try the targeted therapy first and hold off, perhaps, brain directed therapy in that setting? Similar to what one would do with osimertinib perhaps or alectinib?  Dr. Alexander Drilon: Absolutely. It's the same paradigm because we know that we are seeing in a larger population of patients, just generally good activity, both extracranially and intracranially. The goal is to try to spare patients the extra side effects of doing radiation by only giving the TKI. And in practice, even outside of the trial, in patients that I've treated with CNS metastases, the activity has been very good. Dr. Rafeh Naqash: Thank you so much. Now, all TKI therapies have, unfortunately, resistance mechanisms that come up eventually, in my experience at least. What is your experience as far as understanding resistance mechanisms on TRK-based therapies and potential second options after that, whether it's second-generation TRK inhibitors or subsequent targeted therapies in this space? Dr. Alexander Drilon: Thankfully, this has been looked at extensively and I like to categorize resistance into two major groups. So there's a type of resistance which we call on-target resistance and another type which we call off-target resistance. In simple terms, cancers that acquire on-target resistance are still dependent on the NTRK or TRK pathway. And often what happens is, like with other oncogene-targeted therapy pairs, you see the acquisition of a resistance mutation in the kinase domain of NTRK 1, 2, 3 that either changes the dynamics of the kinase or sort of kicks the drug off of the binding site due to steric hindrance.  And for those patients, companies have designed next-generation TRK inhibitors that abrogate resistance, meaning they were designed so that they would work despite the presence of these resistance mutations. And a few of them include repotrectinib, talatrectinib, and selitrectinib that are thought to have activity, but there are many other newer ones that are currently being explored. I will say that there's proof of concept that has been published as well showing that patients who progress on a first-generation TRK inhibitor like larotrectinib or entrectinib who develop acquired resistance that's on-target can respond very well to a next-generation NTRK inhibitor. And while these aren't approved just yet, these are of course available in clinical trials. Now, the second major group is more problematic. This is off-target resistance. And when I describe this to patients, what I usually say is that the cancer sort of ‘phones a friend' and activates a second gene perhaps that isn't NTRK. And examples of that include KRAS or MET or BRAF, very well-known oncogenes in other contexts, but it leads to a reliance outside of the NTRK or TRK pathway per se, which still effectively reactivates the MAP kinase pathway. What to do in that situation? Well, there are select cases and there have been case reports published of patients who get a combination. Say if it's acquired MET amplification, you give a MET inhibitor with a TRK inhibitor and that combination can work. But in many other cases where you don't have access to a combination on a clinical trial or on compassionate use, then you really default to the standard of care for that cancer type. So if it's lung cancer and they've never had chemotherapy before, then it would be platinum-based chemotherapy, say with pemetrexed and a third drug, perhaps if they have lung adenocarcinoma.  Dr. Rafeh Naqash: Thank you so much. This is definitely an exciting field and exciting time to be in this space of drug development, and especially when we have so many interesting tumor-agnostic approvals that have come along in the last few years and more to come. And you've led a lot of this development with several other leaders in this field. So it was very nice discussing this with you, and hopefully, our listeners find it equally interesting and educationally relevant to what we see day in and day out as we perform more and more sequencing for patients with cancer and try to identify some of these rare or not so rare events that are targetable and can definitely change the course of a patient's therapy and outcomes. So thank you once again, Alex, for the discussion on this paper.  But before we end, we'd like to spend a couple of minutes trying to know about the investigator. So could you tell us a little bit about your career trajectory, how you started your fellowship perhaps, how you ended up in drug development, and how you've successfully contributed so much in this field to date? Dr. Alexander Drilon: Sure. So I'm originally from the Philippines, was born there, finished med school, and really wanted to come to the United States to sort of broaden my education and my residency program in internal medicine, then called St. Luke's Roosevelt under Columbia, had a program that sent people to rotate through Memorial Sloan Kettering Cancer where I currently work. So that was my first exposure with oncology. I fell in love with it and eventually became a fellow, fortunately, at Memorial Sloan Kettering. And I mentioned earlier that during that time I had subspecialized in lung cancer and there was a lot of excitement around targeted therapy for oncogene-driven lung cancer. And that was my point of entry. I saw these drugs work very well and I said that if I were in a position to develop newer agents, perhaps for other oncogenes where there wasn't anything developed just yet, that would be really cool. And that was my entry into the phase I world where things later on expanded really the tumor agnostic interrogation using the same principles that were familiar to me in the lung cancer world. And I think I've been very fortunate with the environment and the ability, especially with good in-house sequencing, to match many patients to these trials. And it's been wonderful to see several of these drugs approved. Larotrectinib was the sort of seminal tumor-agnostic approval of a targeted therapy for the first time by any regulatory body. And like you said, the hope is that we see several more of these. Dr. Rafeh Naqash: Awesome. That sounds like a very interesting, phenomenal journey that you've had, and a lot of it is also probably related to the kind of people that you met, mentors, and other people who helped you along the way. And then, of course, you've done a lot for other fellows and trainees in this space of drug development. So thank you again, Alex, for joining us, and thank you for choosing JCO Precision Oncology as a destination for your work. I look forward to interacting with you further subsequently and hopefully seeing more development in this space of novel therapies for fusions and other interesting targets in the lung cancer space.  So thank you for listening to JCO Precision Oncology Conversations. Don't forget to give us a rating or review and be sure to subscribe so you never miss an episode. You can find all ASCO shows at asco.org/podcasts.  The purpose of this podcast is to educate and to inform. This is not a substitute for professional medical care and is not intended for use in the diagnosis or treatment of individual conditions.   Guests on this podcast express their own opinions, experience, and conclusions. Guest statements on the podcast do not express the opinions of ASCO. The mention of any product, service, organization, activity, or therapy should not be construed as an ASCO endorsement.  Bio: Alexander E. Drilon, MD, is a medical oncologist specializing in the treatment of lung cancer. He is chief of early drug development service at Memorial Sloan Kettering Cancer Center. He has clinical expertise in lung cancer and early-phase clinical trials.   COIs Alexander Drilon Honoraria: Medscape, OncLive, PeerVoice, Physicians' Education Resource, Targeted Oncology, MORE Health, Research to Practice, Foundation Medicine, PeerView Consulting or Advisory Role: Ignyta, Loxo, TP Therapeutics, AstraZeneca, Pfizer, Blueprint Medicines, Genentech/Roche, Helsinn Therapeutics, BeiGene, Hengrui Therapeutics, Exelixis, Bayer, Tyra Biosciences, Verastem, Takeda/Millennium, BerGenBio, MORE Health, Lilly, AbbVie, 14ner Oncology/Elevation Oncology, Remedica, Archer, Monopteros Therapeutics, Novartis, EMD Serono/Merck, Melendi, Repare Therapeutics Research Funding: Foundation Medicine Patents, Royalties, Other Intellectual Property: Wolters Kluwer (Royalties for Pocket Oncology) Other Relationship: Merck, GlaxoSmithKline, Teva, Taiho Pharmaceutical, Pfizer, PharmaMar, Puma Biotechnology

Blood Podcast
Sustained MRD negativity & outcomes in myeloma, therapeutic targeting of VWF in TTP, and improving CAR T-cells in CLL with PI3 kinase inhibition

Blood Podcast

Play Episode Listen Later Jan 28, 2022 19:32


On this week's podcast, we'll review a recent analysis of phase 3 daratumumab studies looking at the prognostic impact of sustained minimal residual disease, or MRD negativity in patients with multiple myeloma. Next, we'll hear more about a novel thrombolytic agent targeting von Willebrand factor that may represent a promising approach for the treatment of thrombotic thrombocytopenic purpura. We'll conclude with a study demonstrating that PI3 kinase inhibitor duvelisib can be used to enhance the in vivo efficacy of CAR T cells in CLL.

The Melanie Avalon Biohacking Podcast
#105 - Glen Matten: The Sirtfood Diet, Beneficial Plant Compounds Antioxidants & Polyphenols, Coffee, Chocolate, & Wine, Hormetic Stress & Xenohormesis, Sirtuin Activation, And More!

The Melanie Avalon Biohacking Podcast

Play Episode Listen Later Aug 6, 2021 97:59 Transcription Available


GET TRANSCRIPT AND FULL SHOWNOTES: melanieavalon.com/sirtuins 2:45 - IF Biohackers: Intermittent Fasting + Real Foods + Life: Join Melanie's Facebook Group At facebook.com/groups/paleoOMAD For A Weekly Episode GIVEAWAY, And To Discuss And Learn About All Things Biohacking! All Conversations Welcome! 3:05 - Follow Melanie On Instagram To See The Latest Moments, Products, And #AllTheThings! @MelanieAvalon 3:30 - FOOD SENSE GUIDE: Get Melanie's App At Melanieavalon.com/foodsenseguide To Tackle Your Food Sensitivities! Food Sense Includes A Searchable Catalogue Of 300+ Foods, Revealing Their Gluten, FODMAP, Lectin, Histamine, Amine, Glutamate, Oxalate, Salicylate, Sulfite, And Thiol Status. Food Sense Also Includes Compound Overviews, Reactions To Look For, Lists Of Foods High And Low In Them, The Ability To Create Your Own Personal Lists, And More! 4:00 - DRY FARM WINES: Low Sugar, Low Alcohol, Toxin-Free, Mold-Free, Pesticide-Free, Hang-Over Free Natural Wine! Use The Link dryfarmwines.com/melanieavalon To Get A Bottle For A Penny! 5:05 - BEAUTYCOUNTER: Non-Toxic Beauty Products Tested For Heavy Metals, Which Support Skin Health And Look Amazing! Shop At beautycounter.com/melanieavalon For Something Magical! For Exclusive Offers And Discounts, And More On The Science Of Skincare, Get On Melanie's Private Beautycounter Email List At melanieavalon.com/cleanbeauty! Find Your Perfect Beautycounter Products With Melanie's Quiz: melanieavalon.com/beautycounterquiz 10:25 - Glen Matten's Personal Story 12:00 - Antioxidants 14:40 - What Are The Beneficial Plant Compounds? 15:20 - Phytonutrients 20:45 - How We Benefit From Plant Foods 22:30 - Hormetic Stress 24:15 - Xenohormesis 25:35 - SUNLIGHTEN: Get $200 Off Any Sunlighten Cabin Model Or $100 Off The Solo Unit (That Melanie Has!) AND $99 Shipping (Regularly $598) With The Code MelanieAvalon At MelanieAvalon.Com/Sunlighten. Forward Your Proof Of Purchase To Podcast@MelanieAvalon.com, To Receive A Signed Copy Of What When Wine! 27:00 - The Carnivore Debate 29:45 - Olive Oil, Cocoa Flavonoids, Red Wine And Coffee 31:15 - Blue Zones 32:00 - Being A Dietary Agnostic 33:40 - Plants As Signaling Molecules 35:00 - What Are Sirtuins? 40:00 - What The 7 Sirtuins Are Responsible For 42:20 - The Connection Between Fasting And Sirtuin Activation 46:35 - Antioxidants While Fasting 48:45 - Sirtuin Activation 53:50 - The Difference Between Fasting And Sirtuin Activation 1:00:50 - The First 7 Days Of The Sirtfood Diet 1:05:10 - Gaining Muscle 1:07:50 - LUMEN: The Lumen Breath Analyzer That Tells Your Body If You're Burning Carbs Or Fat For Energy! Get $25 Off A Lumen Device At melanieavalon.com/lumen With The Code melanieavalon25! 1:10:15 - Leucine 1:12:45 -PI3 Kinase, AMPK, MTor 1:19:20 - Home Gardens 1:20:05 - Wine 1:24:25 - Dry Farming 1:25:05 - Chocolate & Cocoa Go To thegoodchocolatecompany.com And Use The Coupon Code MelanieAvalon For 10% Off! 1:32:00 - A Diet Of Inclusion

Better with Dr. Stephanie
All Calories Are Not Created Equal

Better with Dr. Stephanie

Play Episode Listen Later Apr 28, 2021 39:11


This week on Geeky Magic, we debunk the myth that a calorie is a calorie. In this podcast, we discuss how this is incorrect as it does not account for many other important variables, such as ghrelin, leptin, insulin, and fructose. We discuss how energy is made, mitochondria, cell metabolism, and recommended lab tests. Join The Better! Community On Facebook. Get the show notes here.

LowCarbUSA Podcast
Real Hope For Many Cancer Patients - Dr. Lewis Cantley: Ep 46

LowCarbUSA Podcast

Play Episode Listen Later Apr 28, 2020 25:47


Dr. Lewis Cantley has made significant advances in cancer research stemming from his discovery of the signalling pathway phosphoinositide 3-kinase (Pi3K) in 1984.  It is an enzyme that mediates pretty much everything that insulin does, in the liver, in the fat cells, and it mediates the lowering of serum glucose levels because it drives glucose into the muscle, AND into tumours. Since tumours thrive on glucose, they thought to develop a Pi3 Kinase inhibitor in order to try to treat tumours but the inhibitor resulted in startling rises in blood glucose levels.  In order to manage this they tried using many different drugs, including the use of Metformin with very limited success. Most people had to drop out of the treatments as their glucose levels rose dangerously high. Also, for those who could stay on the treatment only got, on average a life extension of just short of one year. His team then tried advocating a very strict ketogenic diet for a small control group and had great success for those who did stay in the diet (which was most of them).  Together with the results they have on combining the ketogenic diet with a Pi3K inhibitor in rat models which, in his words, produced miraculous results they now have formal human clinical trials underway which I am confident will produce miraculous results as well.

Science Signaling Podcast
Science Signaling Podcast, 25 March 2014

Science Signaling Podcast

Play Episode Listen Later Mar 24, 2014 11:10


Maurizio Scaltriti explains why a combination therapy that targets the PI3K-Akt pathway and signaling through the growth factor receptors EGFR and HER3 may be effective against triple-negative breast cancer.

IMPAKT 2013
The Cancer Genome Atlas (TCGA): comprehensive molecular portraits of breast cancer

IMPAKT 2013

Play Episode Listen Later May 28, 2013 13:59


Prof Perou talks to ecancer reporter Peter Goodwin about the Cancer Genome Atlas; working to target drugs and improve treatment by forming a comprehensive picture of breast cancer biology. They also discuss an overview of biomarkers, pathways and inhibitors, drugs in the pipeline for PI3 Kinase and looking at ER positive, HER2 negative cancers. Potential therapeutic advances in triple negative breast cancers are also covered.

AACR 2013 Annual Meeting
Next generation PI3 kinase inhibition: Dr Maurizio Scaltriti – Massachusetts General Hospital, Boston, MA, US

AACR 2013 Annual Meeting

Play Episode Listen Later Apr 22, 2013 13:08


Dr Maurizio Scaltriti talks with ecancer at the 2013 AACR Annual Meeting in Washington DC about hyper activation of the PI3 kinase pathway and its prevalence in breast and head and neck cancer subtypes. Many inhibitors currently inhibit cell activity on many different levels, but recent findings in preclinical and phase I studies reveal that, while they do inhibit this pathway, their affect is limited. Complications predominantly include toxicity and tumours cells overcoming inhibition. The new types of inhibitors have a large therapeutic window, but those that are close to clinical use, such as alpha inhibitors, will be used in combination with other agents rather than as a single agent.

Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 14/19
Etablierung genetischer Modellsysteme der kolorektalen Karzinogenese und Abhängigkeit onkogener Merkmale von B-Raf(V600E)

Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 14/19

Play Episode Listen Later Oct 29, 2012


Für ein genetisches Modell B-Raf(V600E)-mutierter Darmkrebszellen und korrespondierender Wildtyp-Zellen wurde erstmalig in Deutschland das Somatic Cell Gene Targeting eingesetzt. Dabei konnte demonstriert werden, dass RKO eine Oncogene Addiction bezüglich der BRAF-Mutation aufweist. Als weitere B-Raf(V600E)-abhängige Effekte wurden die Selbstversorgung mit Wachstumssignalen (Self-Sufficiency of Growth Signals) und die Resistenz gegen Apoptose in dem Modell festgestellt. Darüber hinaus war die proliferative Kontaktinhibition in V600E-mutierten Klonen durch eine verstärkte Akt-Phosphorylierung aufgehoben und wurde nach Knockout der mutierten Allele im Wildtyp-Zellklon RBW-1 wieder hergestellt. Somit konnten vier zentrale Merkmale der Onkogenität dem mutierten B-Raf(V600E) zugeordnet werden. Andere onkogene Mechanismen waren dagegen vermutlich aufgrund einer Mutation der PI3-Kinase auch in BRAF-Wildtyp-Zellen noch intakt. So waren das Wachstum unter guten Kulturbedingungen und eine verstärkte Expression des EGF-Rezeptors unter Mangelbedingungen nicht vom BRAF-Mutationsstatus abhängig. Außerdem behielten Wildtyp-Zellen ihre Immortalisierung bei und zeigten weiterhin kein relevantes Auftreten von Seneszenz. Es wurden neue Spleißvarianten des BRAF-Gens gefunden und basal charakterisiert. Die alternativen Transkripte zeigten keine Kinase-Aktivität und waren in einem Ausmaß nachweisbar, das eine physiologische Bedeutung vermuten lässt. Hinsichtlich der Herkunft-Allele alternativer Isoformen und den Ursachen für das Auftreten alternativen Spleißens wurden neue Erkenntnisse gewonnen, die zudem die Interpretation publizierter Daten erleichtern. Es wurde gezeigt, dass die durch E-Cadherin vermittelten Zellkontakte essentiell für die epitheliale Komponente der intestinalen Barriere sind. Darüber hinaus wurde der Einfluss von E-Cadherin auf die Ausreifung sekretierender Zellen im Darm ermittelt und damit ein weiterer entscheidender Mechanismus der Abwehr bakterieller Invasionen aufgeklärt.

Science Signaling Podcast
Science Signaling Podcast, 6 March 2012

Science Signaling Podcast

Play Episode Listen Later Mar 5, 2012 12:06


Signaling from two growth factors converges on the cell cycle inhibitor p57 to regulate proliferation and limit oncogenesis.

Fakultät für Physik - Digitale Hochschulschriften der LMU - Teil 04/05
CHEMOTACTIC GRADIENT GENERATOR - A microfluidic Approach on how D. discoideum change direction

Fakultät für Physik - Digitale Hochschulschriften der LMU - Teil 04/05

Play Episode Listen Later Feb 3, 2012


Chemotaxis, the ability of cells to detect and migrate directly towards a source of a chemically active agent, is the result of a sophisticated interplay of proteins within a complex regulatory network. However, partially redundant pathways that simultaneously mediate chemotaxis and dynamic protein distributions complicate the experimental identication of distinct signaling cascades and their inuence on chemotactic migration. Yet, increasingly precise generation and rapid modication of chemotactic stimuliin microuidic devices promise further insight into the basic principles of cellular feedback signaling. I developed a Chemotactic Gradient Generator (CGG) for the exposure of living cells to chemotactic gradient elds with alternating gradient direction based on a double T-junction microuidic chamber. A large extension of the concentration gradients enables the parallel exposure of several dozens of cells to identical chemotactic stimuli, allowing for a reliable quantitative analysis of the chemotactic migration behavior. Two pressure pumps and a syringe pump facilitate accurate control of the inow velocities at the individual ow chamber inlets, pivotal for precise manipulation of the chemotactic stimuli. The CGG combines homogeneous gradients over a width of up to 300 µm and rapid alterations of gradient direction with switching frequencies up to 0.7 Hz. Fast gradient switching in our experimental design facilitates cell stimulation at the intrinsic time scales of their chemotactic response as demonstrated by a gradual increase in the switching frequency of the gradient direction. We eventually observe a "chemotactically trapped" state of Dictyostelium discoideum (D. discoideum) cells at a switching rate of 0.01 Hz. Here, gradient switching proves too fast for the cells to respond to the altered gradient direction by migration. In contrast, we observe oscillatory runs at switching frequencies of less than 0.02 Hz. We distinguish between re-polymerizing cells that exhibit an internal re-organization of the actin cortex in response to chemotactic stimulation and stably polarized cells that gradually adjust their leading edge when the gradient is switched. To experimentally characterize both response types, we record cell shape and the intracellular distribution of actin polymerization activity. Cell shape is readily described by the eccentricity of the cell and to record F-actin polymerization dynamics we introduce a fluorescence distribution moment (FDM). Accurate description of the migratory response behavior facilitates a quantitative analysis of the inuence of both the experimental boundary conditions such as gradient shape, ongoing starvation of the cells, and in particular the inuence of distinct signaling cascades on chemotactic migration. Here, we demonstrate this ability of the GCC by inhibition of PI3-Kinase with LY 294002. PI3-Kinase initiates the formation of fresh pseudopods in the direction of the chemotactic gradient and therefore is one of the key signaling pathways mediating the chemotactic response. In shallow gradients and with ongoing starvation of the cells, we find a decreased ratio of re-polymerizing cells, pointing towards a diminished influence of PI3-Kinase. After inhibition of PI3-Kinase, cell re-polymerization in response to a switch in gradient direction is hindered at 5h of starvation, whereas at 7h of starvation evidence is found that chemotactic migration is more efficient. We observe the astonishing result that in dependency of the boundary conditions of the experiment inhibition of PI3-Kinase promotes an effective chemotactic response. Thus, the CGG for the rst time facilitates a quantitative analysis of the starvation time dependent effect of PI3-Kinase inhibition on D. discoideum chemotaxis.

accurate generator hz gcc ly gradient fdm change direction microfluidics ddc:500 cgg chemotaxis dictyostelium pi3 kinase ddc:530
Science Signaling Podcast
Science Signaling Podcast, 26 October 2010

Science Signaling Podcast

Play Episode Listen Later Oct 25, 2010 9:51


Disrupting activity of the guanosine triphosphatase-activating protein ARAP3 is a potential strategy for anti-angiogenic therapy.

Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 10/19
Zur Bedeutung von Proteinkinasen und Phosphatidylinositol- Kinasen für (anti-) cholestatische und (anti-) apoptotische Effekte von Gallensäuren in der Leberzelle

Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 10/19

Play Episode Listen Later Nov 12, 2009


Gallensäuren stellen potente Signalmoleküle dar, die schon in geringen mikromolaren Konzentrationen, wie sie beim Menschen im Serum beobachtet werden, zentrale Leberzellfunktionen auf transkriptioneller und posttranskriptioneller Ebene beeinflussen. Die hydrophoben und potentiell toxischen Gallensäuren Lithocholsäure (LCA) und Chenodeoxycholsäure (CDCA) induzieren Apoptose und Cholestase, während hydrophile Gallensäuren hepatoprotektiv wirken können. Unter ihnen ist die antiapoptotisch und anticholestatisch wirksame Ursodeoxycholsäure (UDCA) von besonderer Bedeutung. UDCA stellt derzeit das einzige wirksame Therapeutikum bei chronischen cholestatischen Leberkrankheiten dar. Die vorliegende Arbeit untersuchte die Bedeutung intrazellulärer Signaltransduktionswege für die choleretischen, (anti-)cholestatischen und (anti-)apoptotischen Wirkungen physiologischer Gallensäuren in verschiedenen experimentellen Modellen. Hauptziel der Arbeit war die genauere Charakterisierung (i) der für den klinisch bedeutenden anticholestatischen Effekt des Taurinkonjugats der Ursodeoxycholsäure (TUDCA) verantwortlichen Signaltransduktionswege, und (ii) der zentralen Stellung von PI3-Kinasen in der intrazellulären Signalvermittlung der biologischen Effekte hydrophiler und hydrophober Gallensäuren. Im Modell der isoliert perfundierten Rattenleber untersuchten wir die anticholestatische und hepatoprotektive Wirkung der TUDCA in der intakten Leber unter Einsatz pharmakologischer Enzyminhibitoren. Als Leberfunktionsparameter dienten quantitativer Gallenfluß, Sekretion des Modellsubstrats der Konjugatexportpumpe Mrp2, GS-DNP, in die Galle und als Marker der Leberzellschädigung die hepatovenöse LDH-Freisetzung. Simultane Hemmung der cPKCa und der PKA, nicht aber Hemmung von cPKCa oder PKA allein antagonisierte bei Taurolithocholsäure (TLCA)-induzierter Cholestase die protektive Wirkung der TUDCA. Gallenfluß und GS-DNP-Sekretion waren unter gleichzeitiger Hemmung beider Signalwege signifikant reduziert, wohingegen die LDH-Freisetzung deutlich erhöht war. Die Ergebnisse zeigen, dass der posttranskriptionell vermittelte anticholestatische Effekt der TUDCA im etablierten Modell TLCA-induzierter Cholestase durch einen kooperativen cPKC- und PKA-abhängigen Signalweg vermittelt wird. Mitogenaktivierte Proteinkinasen Erk1/2- und p38-abhängige Signalwege hingegen, die als Vermittler von TUDCA-induzierter Cholerese unter nicht-cholestatischen Bedingungen beschrieben wurden, waren im untersuchten Modell ohne Bedeutung für die anticholestatische Wirkung der TUDCA. Mit Hilfe der neu etablierten Biotinylierung von Membranproteinen konnten wir in Ntcp-transfizierten humanen Hepatomzellen (HepG2-Ntcp) zeigen, dass TUDCA unter Cholestase die Insertion von MRP2 in die Hepatozytenmembran anregt. Dieser für die klinische Wirksamkeit der (T)UDCA potentiell bedeutende und im Tiermodell von uns vorbeschriebene Wirkmechanismus konnte damit erstmals in einem humanen Modell nachvollzogen werden. Ein weiterer in vitro Ansatz untersuchte die Phosphorylierung von aus HepG2-Ntcp immunopräzipitiertem MRP2 durch die als Gallensäureneffektoren diskutierten Proteinkinasen cPKCa, nPKCe und PKA. Alle drei Proteinkinasen phosphorylierten, durch den PKC/PKA-Inhibitor Staurosporin hemmbar, MRP2. Diese Phosphorylierung könnte, wie für die Gallensäurentransporter BSEP und NTCP bereits gezeigt, Einfluss auf Aktivität und Membraninsertion von MRP2 haben. Der funktionellen Bedeutung der PI3-Kinasen, welchen in den bisher entschlüsselten Signalwegen sowohl hydrophober/toxischer wie auch hydrophiler/protektiver Gallensäuren eine zentrale Rolle zugesprochen worden war („PI3-Kinasen-Paradoxon“), galten unsere in vitro Untersuchungen zur Aktivität der Isoformen der Klasse I PI3-Kinasen p110a, p110b und p110g nach Stimulation von primären Rattenhepatozyten mit TLCA, GCDCA, TCA und TUDCA in einem neu etablierten isoformspezifischen Kinaseassay. Dabei zeigte sich für jede Gallensäure ein für sie spezifisches Aktivierungsmuster unterschiedlicher PI3-Kinase-Isoformen. PI3-Kinase p110g wurde dabei spezifisch durch die cholestatisch und apoptotisch wirkenden Gallensäuren TLCA und GCDCA aktiviert. In HepG2-Ntcp-Zellen untersuchten wir daher die Bedeutung von p110g für Gallensäuren-induzierte Apoptose nach deren pharmakologischer Hemmung bzw. nach Transfektion mit siRNA gegen p110g. Die apoptotische Wirkung u.a. der Gallensäuren TLCA und GCDCA war unter beiden Methoden der p110g-Antagonisierung deutlich reduziert, wie sowohl in einem Caspase3/7-Assay als auch morphologisch evaluiert. Gallensäuren-unabhängige Apoptose, durch Etoposid bzw. TNFa ausgelöst, war p110g-unabhänig. Die Bedeutung der Aktivierung der PI3-Kinase-Isoform p110a durch TUDCA ist durch weitere experimentelle Untersuchungen zu klären. Die Erkenntnisse der vorliegenden Arbeit tragen zum Verständnis der komplexen Signalgebung im Rahmen cholestatischer Leberschädigung und der therapeutischen Wirkung der (T)UDCA bei und sind damit für die Entwicklung neuer Therapiestrategien bei cholestatischen Leberkrankheiten potentiell von Bedeutung.

arbeit dabei rolle unter entwicklung bedeutung einfluss verst rahmen einsatz dieser wirkung methoden ebene ansatz aktivit modell effekt stellung mit hilfe effekte marker stimulation die bedeutung serum leber untersuchungen wirksamkeit die ergebnisse modellen tca wirkungen aktivierung lca insertion vermittler galle uren assay die erkenntnisse hemmung sirna pka hauptziel charakterisierung konzentrationen zur bedeutung apoptose tiermodell wirkmechanismus sekretion signalwege tudca phosphorylierung cdca ddc:600 gallens im modell tnfa udca isoformen therapeutikum signalweg therapiestrategien signalmolek transfektion lebersch pi3 kinase membranproteinen signalwegen antagonisierung rattenleber proteinkinasen aktivierungsmuster signalvermittlung gcdca
Science Signaling Podcast
Science Signaling Podcast, 09 June 2009

Science Signaling Podcast

Play Episode Listen Later Jun 8, 2009 24:14


Adaptor subunits of phosphoinositide 3-kinase gamma specify distinct cellular responses.

science allergy signaling mast cell adaptor pip2 pi3 kinase degranulation
Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 08/19
Die Bedeutung der Tyrosinkinase Syk für die Aktivierung der PI3-Kinase bei der β2-Integrin-abhängigen Leukozytenmigration

Medizinische Fakultät - Digitale Hochschulschriften der LMU - Teil 08/19

Play Episode Listen Later May 29, 2008


Thu, 29 May 2008 12:00:00 +0100 https://edoc.ub.uni-muenchen.de/8607/ https://edoc.ub.uni-muenchen.de/8607/1/Then_Cornelia.pdf Then, Cornelia

die bedeutung integrin die aktivierung ddc:600 pi3 kinase
Fakultät für Biologie - Digitale Hochschulschriften der LMU - Teil 02/06
Charakterisierung podosomaler Adhäsionsstrukturen in primären humanen Nabelschnurendothelzellen

Fakultät für Biologie - Digitale Hochschulschriften der LMU - Teil 02/06

Play Episode Listen Later Oct 17, 2007


Der Hauptregulator der vaskulären Homöostase ist das Endothel, welches eine Vielzahl an vasoprotektiven Effekten ausübt. Die Integrität und Regulation des endothelialen Zellayers der Blutgefäße ist von großer Bedeutung bei physiologischen und pathologischen Prozessen. Die Basis dieser Phänomene ist in der dynamischen und exakt kontrollierten Regulation des Zytoskeletts begründet. Die wichtigsten Regulatoren des Zytoskeletts stellen die GTPasen der Rho-Familie und deren Effektoren dar. Im Rahmen dieser Doktorarbeit untersuchten wir in primären humanen Nabelschnurendothelzellen, eine neu in Erscheinung tretende Zytoskelettstruktur, der wir in Anlehnung an ähnliche Proteingruppierungen in monozytären Zellen den Namen HUVEC-Podosomen gaben. HUVEC-Podosomen sind aufgrund ihrer Komponenten mit klassischen Podosomen vergleichbar. Allerdings gibt es zwischen beiden Strukturen auch Unterschiede, denn während klassische Podosomen aus Ring und Kern bestehen, zeigen HUVEC-Podosomen eine zweischichtige Architektur. Wie wir weiterhin nachweisen konnten liegen Podosomen der Endothelzellen an der ventralen Plasmamembran und haben engen Kontakt mit der extrazellulären Matrix.. Somit fungieren sie, wie auch die klassischen Podosomen, als Adhäsionsstrukturen. Sie dienen aber nicht nur der Adhäsion, denn wie bei FITC-markierten Monolayer-Versuchen gezeigt werden konnte, haben sie auch eine proteolytische Aktivität, die insbesondere beim Matrixverdau und der daran anschließenden Migration von Bedeutung ist. Ferner können wir zeigen, daß HUVEC-Podosomen in ruhenden, konfluenten Zellayern nicht beobachtet werden können. Sie lassen sich aber in migratorischen (subkonfluent oder nach Verwundung) HUVEC, in hoher Anzahl und diversen ringförmigen Formationen, vorwiegend in Bereichen nahe dem Leitsaum nachweisen. Wie wir mit Hilfe von live cell imaging-Experimenten zeigen konnten, sind diese Strukturen hochdynamisch und breiten sich wellenartig mit einem weiten Radius innerhalb einer Zelle aus. Scheinbar dispergieren diese Formationen oder fusionieren mit der Zellplasma, wodurch sie die enthaltenen Proteine für viele andere zytoplasmatische oder membranöse Prozesse freigeben könnten. Durch Experimente, in denen Zytokine wie VEGF, bzw. Zytokin-produzierende Zellen wie Monozyten den HUVEC-Kulturen zugegeben wurden, konnten wir zeigen, daß diese die Bildung von Podosomen induzieren und sogar erheblich steigern. Unsere Arbeiten mit konstitutiv aktiven und dominant negativen GTPase-Mutanten zeigten weiterhin, daß diese bei der Organisation und Entstehung der HUVEC-Podosomen von entscheidender Bedeutung sind. Ferner konnte mit Hilfe von Mikroinjektionsversuchen von einer Teildomäne (A) des N-WASP-Proteins verifiziert werden, daß der Mechanismus zur Bildung der HUVEC-Podosomen eine Arp2/3-abhängige Aktinnukleation beinhaltet. Weiterhin ist die Bildung dieser Adhäsionsstrukturen auch von Src Tyrosinkinasen und PI3-Kinase abhängig. Eine der Komponenten von HUVEC-Podosomen ist das Markerprotein Drebrin. Drebrin kann nur in diesen Strukturen und an Zell-Zell-Kontakten in HUVEC detektiert werden. Mikroinjektionsversuche von diversen Konstrukten der unterschiedlichen Regionen von Drebrin zeigen, daß dieses Protein von großer Bedeutung für die Bildung und Struktur der HUVEC-Podosomen ist. Die einzelnen Protein-Protein-Interaktionen von podosomalen Komponenten untereinander und mit Drebrin wurden mit Hilfe von Immunpräzipitation getestet. Es ist uns jedoch nicht gelungen einen Drebrin-Interaktionspartner zu finden. Eine Interaktion von Drebrin konnten wir nur mit Drebrin selbst in Form einer Dimerisierung bzw. mit F-Aktin nachgeweisen. Es ist sehr wahrscheinlich, daß es sich bei HUVEC-Podosomen um ein multifunktionelles Organell handelt. Wie wir in dieser Arbeit darstellen, sind HUVEC-Podosomen Adhäsionsstrukturen. Sie können am häufigsten am Leitsaum detektiert werden, wobei ihre Generierung nur in Zellen mit migratorischen Phänotyp (in Zellen am Wundrand oder in subkonfluenten layern) detektiert werden kann. Beide Tatsachen sprechen dafür, daß HUVEC-Podosomen den Prozeß der Migration unterstützen. Zudem können diese Adhäsionsstrukturen die Matrix degradieren, wodurch sie so wiederum zur Migration aber auch invasiven Prozessen beitragen könnten. HUVEC-Podosomen könnten auch eine Funktion als Speicherform ihrer Komponenten ausüben. Sie fusionieren mit der Zellmembran und liefern so möglicherweise notwendige Proteine und Signale, die die Induktion von Protrusionen ermöglichen und so migratorische Prozesse unterstützen könnten. Durch die Involvierung u. a. von Drebrin, das an Zell-Grenzen detektiert werden kann, können HUVEC-Podosomen möglicherweise einen Einfluß auf Zell-Zell-Kontakte und Vorgänge wie Angiogenese ausüben. Dies bestätigt auch die Tatsache, daß Zytokine die Anzahl an Zellen erhöhen, die HUVEC-Podosomen generieren können und Vorgänge wie Wundheilung beschleunigt ablaufen lassen und so u. U. eine klinische Relevanz haben könnten.

Fakultät für Chemie und Pharmazie - Digitale Hochschulschriften der LMU - Teil 02/06
The role of Src-homology 2 domain containing tyrosine phosphatase 2 in growth factor dependent endothelial signalling and angiogenesis

Fakultät für Chemie und Pharmazie - Digitale Hochschulschriften der LMU - Teil 02/06

Play Episode Listen Later May 24, 2007


Endothelial cell survival is indispensable to maintain endothelial integrity and initiate new vessel formation. We investigated the role of SHP-2 in proliferation, survival and sprouting of human microvascular- and umbilical vein endothelial cells (HMEC, HUVEC) using antisense oligonucleotides (AS-ODN) and a pharmacological SHP-2 inhibitor (PtpI IV). Knock-down of SHP-2 decreased bFGF and PDGF dependent endothelial cell proliferation (p

knock map domain dependent akt signalling bf gf growth factor endothelial angiogenesis shp tyrosine pi3k homology ddc:500 phosphatase erk1 pdgf huvec pi3 kinase ddc:540 hmec annexin v
Fakultät für Chemie und Pharmazie - Digitale Hochschulschriften der LMU - Teil 01/06
Charakterisierung der funktionalen Rolle von Cytohesin-1 in der LFA-1-vermittelten T-Zell-Adhäsion

Fakultät für Chemie und Pharmazie - Digitale Hochschulschriften der LMU - Teil 01/06

Play Episode Listen Later Feb 1, 2001


Das Leukocyten-spezifische Integrin LFA-1 spielt eine wichtige Rolle bei der Immunantwort, durch die Vermittlung dynamischer Zell-Zell- bzw. Zell-Matrix-Interaktionen. Die kontrollierte Adhäsion bzw. Deadhäsion von Leukocyten bedarf einer spezifischen Regulation des LFA-1-Integrins und die Aufklärung der molekularen Grundlagen dieser Vorgänge ist von großem Interesse. Cytohesin-1 war unmittelbar vor Beginn dieser Arbeit als cytoplasmatischer Regulationsfaktor der durch LFA-1 vermittelten Zelladhäsion identifiziert worden und seine spezifische Interaktion mit der cytoplasmatischen Domäne von CD18 konnte in vitro dokumentiert werden. Im Rahmen dieser Arbeit gelang es zunächst, die Assoziation von Cytohesin-1 und LFA-1 auch endogen, im intakten Zellverband, mittels Kolokalisationsstudien in der lymphoblastoiden B-Zellinie LCL-721, zu demonstrieren. Ferner konnte mit Hilfe von Mutationsanalysen die, für die Interaktion kritische Region in der cytoplasmatischen Domäne von CD18 lokalisiert werden. Sie befindet sich im aminoterminalen Bereich und umfaßt die Aminosäuren WKA(723 - 725). Die Mutation dieser Aminosäurereste nach TRG resultierte in einem vollständigen Interaktionsverlust mit Cytohesin-1. Die Inhibition der Cytohesin-1/CD18-Bindung konnte dabei sowohl durch Protein-Protein-Interaktionsanalysen in Hefe als auch durch biochemische Bindungsstudien in vitro dokumentiert werden, wobei jeweils Fusionsproteine der cytoplasmatischen Domäne von CD18 charakterisiert wurden. Funktionale Analysen der WKA(723-725)-Region von CD18 ergaben, daß die Mutation von WKA(723-725) nach TRG im intakten LFA-1-Molekül eine signifikante Reduktion der Integrin- Aktivität zur Folge hatte. Sowohl T-Zellklone als auch nicht hämatopoetische Zellen, wie HeLa, wiesen nach Expression von LFA-1(TRG), mit Hilfe rekombinanter Vaccinia- Viren, eine stark reduzierte Adhäsionsfähigkeit an immobilisiertes ICAM-1 auf. Ferner ergaben funktionale Studien mit HeLa-Zellen, die LFA-1 stabil exprimierten, daß Cytohesin-1 nur dann eine gesteigerte Adhäsion dieser Zellen an ICAM-1 induzierte, wenn sie Wildtyp-LFA-1 exprimierten. HeLa-Zellen, die LFA-1(TRG) exprimierten, ließen sich durch Cytohesin-1 zu keiner verstärkten Adhäsion aktivieren. Diese Ergebnisse demonstrierten die Bedeutsamkeit der Cytohesin-1/CD18-Interaktion für eine effiziente, durch LFA-1 vermittelte Zelladhäsion. Unklar war jedoch der Mechanismus, durch den Cytohesin-1 die Integrin/Liganden-Bindung regulierte. Studien mit dem Reporterantikörper 24 ließen darauf schließen, daß Cytohesin-1 durch die Bindung an CD18 eine Konformationsänderung in der extrazellulären Domäne des LFA-1-Integrins induzieren konnte, die möglicherweise die Affinität des Rezeptors modulierte. Diese Modulation der LFA-1-Konformation schien jedoch nicht hinreichend für eine stabile Bindung an ICAM-1 zu sein, wie eingehendere Analysen von Dr. W. Kolanus zeigten. Vielmehr erforderte eine effiziente Zelladhäsion zusätzlich die Guaninnukleotid-Austauschfunktion (GEF-Funktion) von Cytohesin-1, da die GEF-defekte Punktmutante, Cytohesin-1(E157K), nicht mehr in der Lage war, die Adhäsion von Jurkat E6-Zellen an ICAM-1 stabil zu induzieren. Biochemische Interaktionsstudien konnten dabei zeigen, daß die Mutante weiterhin fähig war, die cytoplasmatische Domäne von CD18 zu binden. Diese und weitere Ergebnisse von Dr. W. Nagel, die einen Zusammenhang zwischen der GEF-Funktion von Cytohesin-1 und dem „Spreading“ von adhärenten Jurkat E6-Zellen aufzeigten, legen die Vermutung nahe, daß Cytohesin-1 durch einen dualen Mechanismus in die LFA-1-Regulation involviert ist. Sowohl die direkte Interaktion von Cytohesin-1 und dem Integrin als auch seine GEF-Funktion stellen essentielle Faktoren für eine stabile Zelladhäsion, die durch LFA-1 vermittelt wird, dar. Welche funktionalen Mechanismen dabei durch den Guaninnukleotid-Austausch und der damit verbundenen Aktivierung einer GTPase induziert werden, ist noch unklar. Primär wäre eine Modulation des Aktin-Cytoskelettes und eine damit verbundene erhöhte laterale Mobilität der Integrine denkbar, die eine verstärkte Rezeptormultimerisierung und dadurch eine Aviditätsänderung des Integrins ermöglicht. Weitere Studien dieser Arbeit analysierten die Regulation von Cytohesin-1 selbst. Es konnte gezeigt werden, daß PI3-Kinase in die Kontrolle der Cytohesin-1-Funktion involviert war. Die Überexpression einer konstitutiv aktiven Form dieser Kinase (P110*) führte zu einer gesteigerten Adhäsion von Jurkat E6-Zellen an ICAM-1. Eine Inkubation dieser Zellen mit dem PI3-Kinase-spezifischen Inhibitor Wortmannin resultierte dagegen in einer signifikanten Reduktion der Zelladhäsion. Weitere funktionale Analysen, die die Zelladhäsion von Jurkat E6-Zellen nach Koexpression von P110* und der PH-Domäne von Cytohesin-1 untersuchten, sowie eingehendere Studien von Dr. W. Nagel, ermöglichten die Entwicklung eines Modells zur Regulation von Cytohesin- 1. Demzufolge führt die Aktivierung der PI3-Kinase zu einer verstärkten Rekrutierung von Cytohesin-1 an die Plasmamembran. Als Rekrutierungsmodul fungiert dabei die PHDomäne, die durch Bindung von PtdIns(3,4,5)P3, einem Produkt der PI3-Kinase, die Assoziation mit der Membran gewährleistet. Die Rekrutierung von Cytohesin-1 an die Plasmamembran führt zur Aktivierung von LFA-1 und der damit verbundenen stabilen Zelladhäsion an ICAM-1.