Skip to main content
Erschienen in: Diabetologia 9/2016

06.04.2016 | Mini-Review

MST1: a promising therapeutic target to restore functional beta cell mass in diabetes

verfasst von: Amin Ardestani, Kathrin Maedler

Erschienen in: Diabetologia | Ausgabe 9/2016

Einloggen, um Zugang zu erhalten

Abstract

The loss of insulin-producing beta cells by apoptosis is a hallmark of all forms of diabetes mellitus. Strategies to prevent beta cell apoptosis and dysfunction are urgently needed to restore the insulin-producing cells and to prevent severe diabetes progression. We recently identified the serine/threonine kinase known as mammalian sterile 20-like kinase 1 (MST1) as a critical regulator of apoptotic beta cell death and dysfunction. MST1 activates several apoptotic signalling pathways, which further stimulate its own cleavage, leading to a vicious cycle of cell death. This led us to hypothesise that MST1 signalling is central to the initiation of beta cell death in diabetes. We found that MST1 is strongly activated in a diabetic beta cell and induces not only its death but also directly impairs insulin secretion through promoting proteasomal degradation of key beta cell transcription factor, pancreatic and duodenal homeobox 1 (PDX1), which is critical for insulin production.
Pre-clinical studies in various animal models of diabetes have reported that MST1 deficiency remarkably restores normoglycaemia and beta cell function and prevents the development of diabetes. Importantly, MST1 deficiency can revert fully diabetic beta cells to a non-diabetic state. MST1 may serve as a target for the development of novel therapies for diabetes that trigger the cause of the disease, namely, the destruction of the beta cells. The major current focus of our investigation is to identify and test the efficacy of potent inhibitors of this death signalling pathway to protect beta cells against the effects of autoimmune attack in type 1 diabetes and to preserve beta cell mass and function in type 2 diabetes. This review summarises a presentation given at the ‘Can we make a better beta cell?’ symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Heiko Lickert and colleagues, DOI: 10.​1007/​s00125-016-3949-9, and by Harry Heimberg and colleagues, DOI: 10.​1007/​s00125-016-3879-6) and a commentary by the Session Chair, Shanta Persaud (DOI: 10.​1007/​s00125-016-3870-2).
Literatur
1.
Zurück zum Zitat Ritzel RA, Butler AE, Rizza RA, Veldhuis JD, Butler PC (2006) Relationship between beta cell mass and fasting blood glucose concentration in humans. Diabetes Care 29:717–718CrossRefPubMed Ritzel RA, Butler AE, Rizza RA, Veldhuis JD, Butler PC (2006) Relationship between beta cell mass and fasting blood glucose concentration in humans. Diabetes Care 29:717–718CrossRefPubMed
2.
Zurück zum Zitat Herold KC, Usmani-Brown S, Ghazi T et al (2015) Beta cell death and dysfunction during type 1 diabetes development in at-risk individuals. J Clin Invest 125:1163–1173CrossRefPubMedPubMedCentral Herold KC, Usmani-Brown S, Ghazi T et al (2015) Beta cell death and dysfunction during type 1 diabetes development in at-risk individuals. J Clin Invest 125:1163–1173CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC (2003) Beta cell deficit and increased beta cell apoptosis in humans with type 2 diabetes. Diabetes 52:102–110CrossRefPubMed Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC (2003) Beta cell deficit and increased beta cell apoptosis in humans with type 2 diabetes. Diabetes 52:102–110CrossRefPubMed
4.
Zurück zum Zitat Mathis D, Vence L, Benoist C (2001) Beta cell death during progression to diabetes. Nature 414:792–798CrossRefPubMed Mathis D, Vence L, Benoist C (2001) Beta cell death during progression to diabetes. Nature 414:792–798CrossRefPubMed
5.
6.
Zurück zum Zitat Huang J, Wu S, Barrera J, Matthews K, Pan D (2005) The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila homolog of YAP. Cell 122:421–434CrossRefPubMed Huang J, Wu S, Barrera J, Matthews K, Pan D (2005) The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila homolog of YAP. Cell 122:421–434CrossRefPubMed
7.
8.
Zurück zum Zitat Harvey KF, Zhang X, Thomas DM (2013) The Hippo pathway and human cancer. Nat Rev Cancer 13:246–257CrossRefPubMed Harvey KF, Zhang X, Thomas DM (2013) The Hippo pathway and human cancer. Nat Rev Cancer 13:246–257CrossRefPubMed
9.
Zurück zum Zitat Tremblay AM, Missiaglia E, Galli GG et al (2014) The Hippo transducer YAP1 transforms activated satellite cells and is a potent effector of embryonal rhabdomyosarcoma formation. Cancer Cell 26:273–287CrossRefPubMed Tremblay AM, Missiaglia E, Galli GG et al (2014) The Hippo transducer YAP1 transforms activated satellite cells and is a potent effector of embryonal rhabdomyosarcoma formation. Cancer Cell 26:273–287CrossRefPubMed
10.
Zurück zum Zitat Lin Z, von Gise A, Zhou P et al (2014) Cardiac-specific YAP activation improves cardiac function and survival in an experimental murine MI model. Circ Res 115:354–363CrossRefPubMedPubMedCentral Lin Z, von Gise A, Zhou P et al (2014) Cardiac-specific YAP activation improves cardiac function and survival in an experimental murine MI model. Circ Res 115:354–363CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Barry ER, Camargo FD (2013) The Hippo superhighway: signaling crossroads converging on the Hippo/Yap pathway in stem cells and development. Curr Opin Cell Biol 25:247–253CrossRefPubMed Barry ER, Camargo FD (2013) The Hippo superhighway: signaling crossroads converging on the Hippo/Yap pathway in stem cells and development. Curr Opin Cell Biol 25:247–253CrossRefPubMed
12.
13.
Zurück zum Zitat Tumaneng K, Schlegelmilch K, Russell RC et al (2012) YAP mediates crosstalk between the Hippo and PI(3)K–TOR pathways by suppressing PTEN via miR-29. Nat Cell Biol 14:1322–1329CrossRefPubMedPubMedCentral Tumaneng K, Schlegelmilch K, Russell RC et al (2012) YAP mediates crosstalk between the Hippo and PI(3)K–TOR pathways by suppressing PTEN via miR-29. Nat Cell Biol 14:1322–1329CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Avruch J, Zhou D, Fitamant J, Bardeesy N, Mou F, Barrufet LR (2012) Protein kinases of the Hippo pathway: regulation and substrates. Semin Cell Dev Biol 23:770–784CrossRefPubMedPubMedCentral Avruch J, Zhou D, Fitamant J, Bardeesy N, Mou F, Barrufet LR (2012) Protein kinases of the Hippo pathway: regulation and substrates. Semin Cell Dev Biol 23:770–784CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Radu M, Chernoff J (2009) The DeMSTification of mammalian Ste20 kinases. Curr Biol 19:R421–425CrossRefPubMed Radu M, Chernoff J (2009) The DeMSTification of mammalian Ste20 kinases. Curr Biol 19:R421–425CrossRefPubMed
16.
Zurück zum Zitat Graves JD, Draves KE, Gotoh Y, Krebs EG, Clark EA (2001) Both phosphorylation and caspase-mediated cleavage contribute to regulation of the Ste20-like protein kinase Mst1 during CD95/Fas-induced apoptosis. J Biol Chem 276:14909–14915CrossRefPubMed Graves JD, Draves KE, Gotoh Y, Krebs EG, Clark EA (2001) Both phosphorylation and caspase-mediated cleavage contribute to regulation of the Ste20-like protein kinase Mst1 during CD95/Fas-induced apoptosis. J Biol Chem 276:14909–14915CrossRefPubMed
17.
Zurück zum Zitat Lee KK, Ohyama T, Yajima N, Tsubuki S, Yonehara S (2001) MST, a physiological caspase substrate, highly sensitizes apoptosis both upstream and downstream of caspase activation. J Biol Chem 276:19276–19285CrossRefPubMed Lee KK, Ohyama T, Yajima N, Tsubuki S, Yonehara S (2001) MST, a physiological caspase substrate, highly sensitizes apoptosis both upstream and downstream of caspase activation. J Biol Chem 276:19276–19285CrossRefPubMed
18.
Zurück zum Zitat Ura S, Masuyama N, Graves JD, Gotoh Y (2001) Caspase cleavage of MST1 promotes nuclear translocation and chromatin condensation. Proc Natl Acad Sci U S A 98:10148–10153CrossRefPubMedPubMedCentral Ura S, Masuyama N, Graves JD, Gotoh Y (2001) Caspase cleavage of MST1 promotes nuclear translocation and chromatin condensation. Proc Natl Acad Sci U S A 98:10148–10153CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Cinar B, Fang PK, Lutchman M et al (2007) The pro-apoptotic kinase Mst1 and its caspase cleavage products are direct inhibitors of Akt1. EMBO J 26:4523–4534CrossRefPubMedPubMedCentral Cinar B, Fang PK, Lutchman M et al (2007) The pro-apoptotic kinase Mst1 and its caspase cleavage products are direct inhibitors of Akt1. EMBO J 26:4523–4534CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Yuan T, Awal S, Kerr-Conte J, Ardestani A (2015) The hippo kinase LATS2 impairs pancreatic beta cell survival and function. Diabetologia 58: S46-S47 (Abstract) Yuan T, Awal S, Kerr-Conte J, Ardestani A (2015) The hippo kinase LATS2 impairs pancreatic beta cell survival and function. Diabetologia 58: S46-S47 (Abstract)
22.
Zurück zum Zitat Bonny C, Oberson A, Negri S, Sauser C, Schorderet DF (2001) Cell-permeable peptide inhibitors of JNK: novel blockers of beta cell death. Diabetes 50:77–82CrossRefPubMed Bonny C, Oberson A, Negri S, Sauser C, Schorderet DF (2001) Cell-permeable peptide inhibitors of JNK: novel blockers of beta cell death. Diabetes 50:77–82CrossRefPubMed
23.
Zurück zum Zitat Kaneto H, Nakatani Y, Miyatsuka T et al (2004) Possible novel therapy for diabetes with cell-permeable JNK-inhibitory peptide. Nat Med 10:1128–1132CrossRefPubMed Kaneto H, Nakatani Y, Miyatsuka T et al (2004) Possible novel therapy for diabetes with cell-permeable JNK-inhibitory peptide. Nat Med 10:1128–1132CrossRefPubMed
24.
Zurück zum Zitat Aikin R, Maysinger D, Rosenberg L (2004) Cross-talk between phosphatidylinositol 3-kinase/AKT and c-jun NH2-terminal kinase mediates survival of isolated human islets. Endocrinology 145:4522–4531CrossRefPubMed Aikin R, Maysinger D, Rosenberg L (2004) Cross-talk between phosphatidylinositol 3-kinase/AKT and c-jun NH2-terminal kinase mediates survival of isolated human islets. Endocrinology 145:4522–4531CrossRefPubMed
25.
Zurück zum Zitat Tuttle RL, Gill NS, Pugh W et al (2001) Regulation of pancreatic beta cell growth and survival by the serine/threonine protein kinase Akt1/PKBα. Nat Med 7:1133–1137CrossRefPubMed Tuttle RL, Gill NS, Pugh W et al (2001) Regulation of pancreatic beta cell growth and survival by the serine/threonine protein kinase Akt1/PKBα. Nat Med 7:1133–1137CrossRefPubMed
26.
Zurück zum Zitat Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB (2005) Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov 4:988–1004CrossRefPubMed Hennessy BT, Smith DL, Ram PT, Lu Y, Mills GB (2005) Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov 4:988–1004CrossRefPubMed
27.
Zurück zum Zitat Jang SW, Yang SJ, Srinivasan S, Ye K (2007) Akt phosphorylates MstI and prevents its proteolytic activation, blocking FOXO3 phosphorylation and nuclear translocation. J Biol Chem 282:30836–30844CrossRefPubMed Jang SW, Yang SJ, Srinivasan S, Ye K (2007) Akt phosphorylates MstI and prevents its proteolytic activation, blocking FOXO3 phosphorylation and nuclear translocation. J Biol Chem 282:30836–30844CrossRefPubMed
28.
Zurück zum Zitat Yuan Z, Kim D, Shu S et al (2010) Phosphoinositide 3-kinase/Akt inhibits MST1-mediated pro-apoptotic signaling through phosphorylation of threonine 120. J Biol Chem 285:3815–3824CrossRefPubMed Yuan Z, Kim D, Shu S et al (2010) Phosphoinositide 3-kinase/Akt inhibits MST1-mediated pro-apoptotic signaling through phosphorylation of threonine 120. J Biol Chem 285:3815–3824CrossRefPubMed
29.
Zurück zum Zitat McKinnon CM, Docherty K (2001) Pancreatic duodenal homeobox-1, PDX-1, a major regulator of beta cell identity and function. Diabetologia 44:1203–1214CrossRefPubMed McKinnon CM, Docherty K (2001) Pancreatic duodenal homeobox-1, PDX-1, a major regulator of beta cell identity and function. Diabetologia 44:1203–1214CrossRefPubMed
30.
Zurück zum Zitat Stoffers DA, Zinkin NT, Stanojevic V, Clarke WL, Habener JF (1997) Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat Genet 15:106–110CrossRefPubMed Stoffers DA, Zinkin NT, Stanojevic V, Clarke WL, Habener JF (1997) Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat Genet 15:106–110CrossRefPubMed
31.
Zurück zum Zitat Brissova M, Shiota M, Nicholson WE et al (2002) Reduction in pancreatic transcription factor PDX-1 impairs glucose-stimulated insulin secretion. J Biol Chem 277:11225–11232CrossRefPubMed Brissova M, Shiota M, Nicholson WE et al (2002) Reduction in pancreatic transcription factor PDX-1 impairs glucose-stimulated insulin secretion. J Biol Chem 277:11225–11232CrossRefPubMed
33.
Zurück zum Zitat Stoffers DA, Ferrer J, Clarke WL, Habener JF (1997) Early-onset type-II diabetes mellitus (MODY4) linked to IPF1. Nat Genet 17:138–139CrossRefPubMed Stoffers DA, Ferrer J, Clarke WL, Habener JF (1997) Early-onset type-II diabetes mellitus (MODY4) linked to IPF1. Nat Genet 17:138–139CrossRefPubMed
34.
Zurück zum Zitat Claiborn KC, Sachdeva MM, Cannon CE, Groff DN, Singer JD, Stoffers DA (2010) Pcif1 modulates Pdx1 protein stability and pancreatic beta cell function and survival in mice. J Clin Invest 120:3713–3721CrossRefPubMedPubMedCentral Claiborn KC, Sachdeva MM, Cannon CE, Groff DN, Singer JD, Stoffers DA (2010) Pcif1 modulates Pdx1 protein stability and pancreatic beta cell function and survival in mice. J Clin Invest 120:3713–3721CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Babu DA, Deering TG, Mirmira RG (2007) A feat of metabolic proportions: Pdx1 orchestrates islet development and function in the maintenance of glucose homeostasis. Mol Genet Metab 92:43–55CrossRefPubMedPubMedCentral Babu DA, Deering TG, Mirmira RG (2007) A feat of metabolic proportions: Pdx1 orchestrates islet development and function in the maintenance of glucose homeostasis. Mol Genet Metab 92:43–55CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Fujimoto K, Ford EL, Tran H et al (2010) Loss of Nix in Pdx1-deficient mice prevents apoptotic and necrotic beta cell death and diabetes. J Clin Invest 120:4031–4039CrossRefPubMedPubMedCentral Fujimoto K, Ford EL, Tran H et al (2010) Loss of Nix in Pdx1-deficient mice prevents apoptotic and necrotic beta cell death and diabetes. J Clin Invest 120:4031–4039CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Leibowitz G, Ferber S, Apelqvist A et al (2001) IPF1/PDX1 deficiency and beta cell dysfunction in Psammomys obesus, an animal with type 2 diabetes. Diabetes 50:1799–1806CrossRefPubMed Leibowitz G, Ferber S, Apelqvist A et al (2001) IPF1/PDX1 deficiency and beta cell dysfunction in Psammomys obesus, an animal with type 2 diabetes. Diabetes 50:1799–1806CrossRefPubMed
39.
Zurück zum Zitat Fujimoto K, Hanson PT, Tran H et al (2009) Autophagy regulates pancreatic beta cell death in response to Pdx1 deficiency and nutrient deprivation. J Biol Chem 284:27664–27673CrossRefPubMedPubMedCentral Fujimoto K, Hanson PT, Tran H et al (2009) Autophagy regulates pancreatic beta cell death in response to Pdx1 deficiency and nutrient deprivation. J Biol Chem 284:27664–27673CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Elrick LJ, Docherty K (2001) Phosphorylation-dependent nucleocytoplasmic shuttling of pancreatic duodenal homeobox-1. Diabetes 50:2244–2252CrossRefPubMed Elrick LJ, Docherty K (2001) Phosphorylation-dependent nucleocytoplasmic shuttling of pancreatic duodenal homeobox-1. Diabetes 50:2244–2252CrossRefPubMed
41.
Zurück zum Zitat Macfarlane WM, McKinnon CM, Felton-Edkins ZA, Cragg H, James RF, Docherty K (1999) Glucose stimulates translocation of the homeodomain transcription factor PDX1 from the cytoplasm to the nucleus in pancreatic beta-cells. J Biol Chem 274:1011–1016CrossRefPubMed Macfarlane WM, McKinnon CM, Felton-Edkins ZA, Cragg H, James RF, Docherty K (1999) Glucose stimulates translocation of the homeodomain transcription factor PDX1 from the cytoplasm to the nucleus in pancreatic beta-cells. J Biol Chem 274:1011–1016CrossRefPubMed
42.
Zurück zum Zitat Boucher MJ, Selander L, Carlsson L, Edlund H (2006) Phosphorylation marks IPF1/PDX1 protein for degradation by glycogen synthase kinase 3-dependent mechanisms. J Biol Chem 281:6395–6403CrossRefPubMed Boucher MJ, Selander L, Carlsson L, Edlund H (2006) Phosphorylation marks IPF1/PDX1 protein for degradation by glycogen synthase kinase 3-dependent mechanisms. J Biol Chem 281:6395–6403CrossRefPubMed
43.
Zurück zum Zitat Lebrun P, Montminy MR, van Obberghen E (2005) Regulation of the pancreatic duodenal homeobox-1 protein by DNA-dependent protein kinase. J Biol Chem 280:38203–38210CrossRefPubMed Lebrun P, Montminy MR, van Obberghen E (2005) Regulation of the pancreatic duodenal homeobox-1 protein by DNA-dependent protein kinase. J Biol Chem 280:38203–38210CrossRefPubMed
44.
Zurück zum Zitat Kishi A, Nakamura T, Nishio Y, Maegawa H, Kashiwagi A (2003) Sumoylation of Pdx1 is associated with its nuclear localization and insulin gene activation. Am J Physiol Endocrinol Metab 284:E830–E840CrossRefPubMed Kishi A, Nakamura T, Nishio Y, Maegawa H, Kashiwagi A (2003) Sumoylation of Pdx1 is associated with its nuclear localization and insulin gene activation. Am J Physiol Endocrinol Metab 284:E830–E840CrossRefPubMed
45.
Zurück zum Zitat Gao Y, Miyazaki J, Hart GW (2003) The transcription factor PDX-1 is post-translationally modified by O-linked N-acetylglucosamine and this modification is correlated with its DNA binding activity and insulin secretion in min6 beta-cells. Arch Biochem Biophys 415:155–163CrossRefPubMed Gao Y, Miyazaki J, Hart GW (2003) The transcription factor PDX-1 is post-translationally modified by O-linked N-acetylglucosamine and this modification is correlated with its DNA binding activity and insulin secretion in min6 beta-cells. Arch Biochem Biophys 415:155–163CrossRefPubMed
46.
Zurück zum Zitat An R, da Silva XG, Semplici F et al (2010) Pancreatic and duodenal homeobox 1 (PDX1) phosphorylation at serine-269 is HIPK2-dependent and affects PDX1 subnuclear localization. Biochem Biophys Res Commun 399:155–161CrossRefPubMedPubMedCentral An R, da Silva XG, Semplici F et al (2010) Pancreatic and duodenal homeobox 1 (PDX1) phosphorylation at serine-269 is HIPK2-dependent and affects PDX1 subnuclear localization. Biochem Biophys Res Commun 399:155–161CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Kawamori D, Kaneto H, Nakatani Y et al (2006) The forkhead transcription factor Foxo1 bridges the JNK pathway and the transcription factor PDX-1 through its intracellular translocation. J Biol Chem 281:1091–1098CrossRefPubMed Kawamori D, Kaneto H, Nakatani Y et al (2006) The forkhead transcription factor Foxo1 bridges the JNK pathway and the transcription factor PDX-1 through its intracellular translocation. J Biol Chem 281:1091–1098CrossRefPubMed
48.
Zurück zum Zitat Rafiq I, Kennedy HJ, Rutter GA (1998) Glucose-dependent translocation of insulin promoter factor-1 (IPF-1) between the nuclear periphery and the nucleoplasm of single MIN6 beta-cells. J Biol Chem 273:23241–23247CrossRefPubMed Rafiq I, Kennedy HJ, Rutter GA (1998) Glucose-dependent translocation of insulin promoter factor-1 (IPF-1) between the nuclear periphery and the nucleoplasm of single MIN6 beta-cells. J Biol Chem 273:23241–23247CrossRefPubMed
Metadaten
Titel
MST1: a promising therapeutic target to restore functional beta cell mass in diabetes
verfasst von
Amin Ardestani
Kathrin Maedler
Publikationsdatum
06.04.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 9/2016
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-016-3892-9

Weitere Artikel der Ausgabe 9/2016

Diabetologia 9/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Triglyzeridsenker schützt nicht nur Hochrisikopatienten

10.05.2024 Hypercholesterinämie Nachrichten

Patienten mit Arteriosklerose-bedingten kardiovaskulären Erkrankungen, die trotz Statineinnahme zu hohe Triglyzeridspiegel haben, profitieren von einer Behandlung mit Icosapent-Ethyl, und zwar unabhängig vom individuellen Risikoprofil.

Gibt es eine Wende bei den bioresorbierbaren Gefäßstützen?

In den USA ist erstmals eine bioresorbierbare Gefäßstütze – auch Scaffold genannt – zur Rekanalisation infrapoplitealer Arterien bei schwerer PAVK zugelassen worden. Das markiert einen Wendepunkt in der Geschichte dieser speziellen Gefäßstützen.

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Wie managen Sie die schmerzhafte diabetische Polyneuropathie?

10.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Mit Capsaicin-Pflastern steht eine neue innovative Therapie bei schmerzhafter diabetischer Polyneuropathie zur Verfügung. Bei therapierefraktären Schmerzen stellt die Hochfrequenz-Rückenmarkstimulation eine adäquate Option dar.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.