what term best describes a failure of the body’s cells to respond to secretion of insulin?

  • Loading metrics

Oscillations, Intercellular Coupling, and Insulin Secretion in Pancreatic β Cells

  • Patrik Rorsman

Oscillations, Intercellular Coupling, and Insulin Secretion in Pancreatic β Cells

  • Patrick Eastward MacDonald,
  • Patrik Rorsman

PLOS

x

  • Published: Feb 14, 2006
  • https://doi.org/x.1371/periodical.pbio.0040049

Information technology's piece of cake to say nosotros are what we eat, just this unproblematic statement belies the complexity of metabolic signalling that goes into balancing food intake with energy expenditure. One hormone in particular—insulin—is a critically important regulator of whole body energy metabolism. Information technology is secreted from the pancreas when blood glucose levels are high, and it acts to maintain glucose homeostasis by promoting glucose uptake and storage in muscle, fat, and liver. When insulin secretion is absent-minded or reduced, or when peripheral tissues fail to respond to insulin, the result is hyperglycaemia leading ultimately to diabetes. Diabetes affects more than than 170 million people worldwide and is associated with several long-term complications including nerve damage, kidney failure, microcirculatory damage, and a greater hazard for heart disease and stroke.

There are two types of secretion: exocrine and endocrine. In endocrine secretion, the secreted molecules terminate up in the blood and they reach their target cells throughout the body via the circulation. By dissimilarity, exocrine secretion does not involve the circulation and the products are released direct into the exterior world. Almost of the pancreas serves the exocrine function of secreting digestive enzymes into the gut. Less than 1% of the pancreatic tissue is devoted to an endocrine role. The endocrine tissue of the pancreas is organized as cell clusters, called the islets of Langerhans, which are dispersed throughout the pancreatic exocrine tissue and receive a rich vascular (blood vessel) supply (Figure ane). A pancreatic islet comprises three master cell types. Pancreatic α cells (fifteen%) occupy the islet periphery and secrete glucagon in response to low blood glucose. Glucagon opposes the deportment of insulin, thereby increasing circulating glucose levels. Pancreatic δ cells, the to the lowest degree abundant prison cell type (5%), are dispersed throughout the islet and secrete somatostatin, which has important paracrine effects that suppress insulin and glucagon secretion. The insulin-secreting β cells are the most abundant cell type (80%) and comprise the islet core.

thumbnail

Effigy one. Pancreatic Endocrine Tissue Comprises 1%, or Less, of the Pancreas and Is Organized every bit Clusters of Cells Dispersed throughout the Exocrine Pancreas

These cell clusters, the islets of Langerhans, are heterogeneous and composed of 3 main cell types that secrete distinct hormones. The majority of islet cells comprise insulin secreting β cells and act as glucose sensors, releasing insulin in response to increased circulating glucose. The machinery controlling regulated insulin secretion from β cells is shown in the right panel.

https://doi.org/10.1371/journal.pbio.0040049.g001

During development, the pancreas arises as an off-branching of early gut tissues, and develops every bit a prepare of branching tubules which give rise to clusters of endocrine and exocrine cells. Studies have shown that the cytokine TGF-β plays a major role in the development of pancreatic β cells during development of the organ [1, 2], and a newspaper past Smart et al. in this issue of PLoS Biology [3] demonstrates that TGF-β signalling is also critical in the maintenance of β cell functional identity in the adult. Smart and her colleagues were able to show that loss of TGF-β signalling in these cells causes reversion of these cells to an immature differentiated state and resulted in diabetes. Therefore, TGF-β is important for maintaining the functional characteristics of β cells.

In type i diabetes, the less common but more than astringent form of the disease, pancreatic β cells are destroyed by an autoimmune reaction. Type 2 diabetes accounts for greater than 85% of the cases of diabetes. In this course of the disease, the β cells persist, simply for reasons that remain to be established they fail to secrete insulin in sufficient quantities to maintain claret glucose within the normal range. Disrupted insulin secretion is observed prior to onset of type two diabetes [four], and when combined with the development of insulin resistance in peripheral tissues, results in chronic hyperglycaemia. Further deterioration of β cell function contributes to the progression of blazon 2 diabetes [five]. Type two diabetes is believed to effect from an unfortunate combination of variants (polymorphisms) in several diabetes susceptibility genes [half dozen]. Rarer monogenic forms of the disease result from mutations in genes encoding proteins that are critical to glucose-sensing in the β jail cell [7]. Thus, an appreciation of the mechanisms regulating β cell function and insulin secretion is crucial towards understanding the pathogenesis of type 2 diabetes.

The Stimulus-Secretion Coupling Mechanism

Glucose-dependent insulin secretion from β cells, by illustration to excitation–contraction coupling in musculus, is referred to equally stimulus-secretion coupling. Indeed, like musculus activation, the secretion of insulin is dependent on electrical activity and calcium, Catwo+, entry. β cells have channels in their membranes that let for the flow of ions (mainly calcium, Ca2+, and potassium, K+) into and out of the cell. Considering ions are electrically charged, their flux across the membrane may give rising to sharp changes in voltage (action potentials). Glucose stimulation elicits depolarisation of the cell membrane and electrical activeness in β cells [viii–10]. This serves to open Catwo+ channels in the membrane that answer to changes in voltage—voltage-dependent calcium channels (VDCCs)—and allow Ca2+ entry and activity potential firing. Ca2+ acts on the exocytotic machinery to stimulate fusion of insulin-containing vesicles with the plasma membrane for secretion into the bloodstream [xi]. Removal of extracellular Ca2+ prevents activeness potential firing [12] and insulin secretion [13, 14]. Numerous subsequent studies accept confirmed the essential roles of glucose-stimulated membrane depolarisation, activity potential firing, and entry of Caii+ in the regulation of insulin secretion.

Metabolism of glucose is essential for insulin secretion, and inhibition of mitochondrial metabolism blocks insulin secretion [xv]. Mechanisms of β cell glucose metabolism and metabolic signal generation accept been recently reviewed [16]. The breakdown of glucose results in the generation of ATP, one of the key molecules fueling cellular reactions. An increased ATP:ADP ratio represents the critical link betwixt mitochondrial metabolism and insulin secretion through its ability to close ATP-dependent K+ (KATP) channels and depolarise the cell [17] (Figure ane). KATP channels are equanimous of four pore-forming subunits (Kir6.2 in β cells) and four accessory sulfonylurea receptor subunits (SUR1 in β cells). The latter are the target of the anti-diabetic sulphonylurea drugs which stimulate insulin secretion by mimicking the outcome of glucose to close KATP channels. Polymorphism in 1000ATP subunits contribute to diabetes susceptibility by altering the biophysical properties of the channels [6].

Under low glucose conditions, KATP channels are open up, assuasive the outward flux of K+ and holding the cell membrane potential at about −70 mV. Closure of GATP channels, by glucose-induced increases in ATP, drives the membrane voltage to more positive potentials, and somewhen triggers the firing of action potentials resulting from activation of VDCCs (Figure ane). The major VDCC subtype expressed in β cells and that regulates insulin secretion is the L-type Ca2+ channel (Cavone.2). The essential role of this channel has been demonstrated both past pharmacological [eighteen] and genetic [19] inhibition of the channel. Both of these approaches event in a severe reduction in glucose stimulated insulin secretion. Although the L-type Caii+ aqueduct certainly plays a chief role in the regulation of insulin secretion, information technology is non the but VDCC expressed in β cells, and recent work suggests an important role for the R-type Ca2+ in insulin secretion during prolonged stimulation [twenty].

(United nations)Coupling Glucose Metabolism and ATP Production in β Cells

Because the membrane voltage is sensitive to changes in ATP levels within the jail cell, perturbations of the metabolic pathways that generate ATP can have a potent upshot on insulin secretion. ATP is generated in mitochondria through the electron transport chain, and is dependent upon the presence of a proton gradient (H+) beyond the mitochondrial membrane. In β cells, expression of uncoupling protein-2 (UCP2) can disrupt the generation of ATP in mitochondria by permitting protons to leak across the mitochondrial membrane. When UCP2 is overexpressed, the generation of ATP is bypassed [21], while loss of UCP2 expression results in increased ATP levels and as well enhanced insulin release past islets [22]. Accordingly, there may exist a correlation between expression levels of UCP2 and diabetes or obesity.

Although UCP2 clearly plays a role in regulating ATP product, the molecular pathways controlling its expression are not well understood. Bordone et al. (in a paper published in this issue of PLoS Biological science [23]) uncovered one potential regulator of UCP2 expression in their studies of Sirt1 expression and office in murine islets. The authors institute that Sirt1, a homologue of Sir2 (which itself is known to play diverse and important roles in regulating metabolism in organisms from yeast to mammals) is expressed in β cells, and that it downregulates UCP2 expression in these cells. This identifies Sirt1 every bit a positive regulator of insulin secretion from β cells.

Oscillatory Responses and Cell-to-Jail cell Coupling in β cells

Over the physiological range of glucose concentrations, β cell electric activity consists of oscillations in membrane potential between depolarised plateaux, on which bursts of activity potentials are superimposed, separated by repolarized electrically silent intervals. These oscillations in electrical activity are accompanied by changes in the cytoplasmic Ca2+ concentration [24], every bit demonstrated in Figure 2, which in plow give ascent to brief pulses (∼10 s) of insulin secretion [25–27].

thumbnail

Figure 2. The Responses of β Cells within Intact Islets Are Oscillatory and Synchronised

Here, the intracellular Ca2+ responses were measured using ratiometric methods and confocal microscopy. In islet β cells, marked R1-R6 in (A), glucose-stimulation results in increases in intracellular Ca2+ equally shown in (B). Oscillations in intracellular Ca2+, with a flow of ∼10 s, are observed. Furthermore, every bit seen in the expanded time scale in (C), these oscillations are synchronized within separate β cells throughout the islet.

https://doi.org/ten.1371/journal.pbio.0040049.g002

These oscillations reflect a rest betwixt activation of VDCCs (depolarization) and 1000+ channel activity (repolarization) [ten]. The depolarizing component predominates at the beginning of the outburst, merely the resultant influx of Catwo+ during the plateau leads to a progressive Ca2+-induced increment in K+ channel activity. This occurs both via a directly effect on small-scale conductance Caii+-activated Chiliad+ (SK) channels [28], and via an indirect effect on ThouATP channels by lowering of the cytoplasmic ATP:ADP ratio due to increased Catwo+ ATPase action [29]. The increase in Thou+ channel activity somewhen becomes big enough to repolarize the β cell, ending the burst. In this scenario, the wearisome pacemaker depolarization between 2 successive bursts results from the gradual restoration of [Ca2+]i and the ATP:ADP ratio until SK and GrandATP channels are again closed and the background depolarizing conductance becomes sufficiently big to trigger a new outburst of action potentials.

Glucose produces a concentration-dependent increase in the duration of the bursts at the expense of the silent intervals until eventually, at glucose concentrations across twenty mM, uninterrupted action potential firing is observed. This may result from the higher rate of glucose metabolism at high concentrations of the carbohydrate and then that Ca2+ influx is unable to lower ATP sufficiently to produce an increment in 1000+ conductance large enough to trigger membrane repolarization. This model is supported by the ability of tolbutamide, a blocker of the MATP channel that has been used for more than than 50 years to treat diabetes, to suppress β cell membrane potential oscillations that results in continuous firing [29, thirty]. Thus, the role of 1000ATP channels in the β cell extends across just serving as the glucose-regulated resting conductance. They besides contribute to the progressive stimulation of electric activity and insulin release by supra-threshold glucose levels.

In that location is an interesting dependence of oscillatory electric activity on islet integrity and the 10–xv s catamenia typically observed in intact pancreatic islets is for the most part lost in isolated cells maintained in brusque-term tissue civilisation [30]. This has been attributed to changes in channel expression [30], loss of paracrine signalling [31], and requirement of cell coupling [32]. Indeed, β cells within the aforementioned pancreatic islet are electrically coupled [33, 34], such that the [Ca2+ ]i oscillations within dissimilar parts of the islet occur in stage (Figure ii). This synchronization presumably accounts for the observation of pulsatile insulin secretion from individual pancreatic islets [26]. Pancreatic β cells contain the gap junction protein connexin-36, ablation of which leads to loss of oscillatory insulin secretion, whereas [Ca2+]i oscillations in the private cells is maintained [35].

Whereas β cells are electrically coupled to each other, electrical coupling [36] and synchronization of the [Catwo+ ]i oscillations [37] between β cells and non β cells and between non β cells appears much weaker if it exists at all. This it at variance with some of the early data looking at the period of an injected dye between cells which demonstrated the existence of both homotypic (i.e., β to β cell) and heterotypic (e.g., β to a cell) cell coupling [38, 39]. Nonetheless, more than contempo observations using noninvasive techniques suggest that dye coupling may be less all-encompassing than previously idea [xl].

In this issue of PLoS Biology, Rocheleau et al. [41] have studied the functional significance of electrical coupling between β cells using a novel and ingenious arroyo. They accept used genetically engineered mice in which the ThousandATP channel is rendered not-functional—by replacement of specific amino acids—in only some of the pancreatic β cells. This mosaic expression of the transgene (Kir6.2[AAA]) results in functional KATP channel knockout in ∼lxx% of the β cells. Somewhat surprisingly, intact islets from mice expressing the transgene exhibited an essentially normal glucose-dependent insulin secretion, when tested in vitro. Importantly, this required the integrity of the pancreatic islet since normal glucose regulation was lost upon dispersion of the islet into single cells. Insulin secretion from individual Kir6.2[AAA] islet cells occurred already at 1 mM glucose, which in normal cells is a not-stimulatory concentration. Moreover, insulin release was not further stimulated with increasing glucose concentrations. The observation that application of the gap junction inhibitor 18a-glycyrrhetinic acid to intact islets mimicked the effect of islet dispersion makes it probable that this difference results from electrical coupling that can only operate within the intact islet.

These data are consistent with the view that the islet functions as a syncytium (that is, an organ that in electrical terms behaves similar i cell) where KATP channel activeness in the private cells determines the excitability of the entire organ. This is reminiscent of the channel sharing concept originally proposed by Sherman et al. [42] to explain the membrane potential oscillations in islets. Piece of work on isolated cells, even when taken from the aforementioned animal, indicate a significant heterogeneity in the fourth dimension courses and magnitude of their responses to glucose stimulation. It seems possible that this reflects a metabolic heterogeneity and that some cells metabolise glucose better than others. This metabolic heterogeneity will result in variable ThouATP aqueduct activity in the individual cells. The written report by Rochelau et al. is significant besides in this context. They show that all cells inside an intact islet respond to glucose in the aforementioned fashion although the GrandATP channel activity in the individual cells ranged between zero and 100% of the normal. The only divergence from normality was a slight shift (∼2 mM) towards lower concentration in the glucose dose-response curve. Thus, a lowered KATP aqueduct activity in the Kir6.2[AAA] expressing cells will increase excitability in their normal neighbours and vice versa.

Can cell coupling exist of pathophysiological significance? Given that well-nigh of the ATP required for β cell function is of mitochondrial origin, processes that interfere with oxidative phosphorylation are likely to be important in the aetiology of type 2 diabetes. Heteroplasmy of mitochondrial cistron mutations leading to lowered ATP production (reviewed by [43]) and increased KATP channel activeness in a minority of the β cells within the cell may thus, via cell coupling, compromise electric activity and secretion in the entire islet, perhaps enough to result in clinical diabetes.

Acknowledgments

Supported by the Wellcome Trust. PEM is the European Foundation for the Written report of Diabetes/AstraZeneca Swain in Islet Biological science, and PR is a Imperial Society Wolfson Fellow.

References

  1. i. Sanvito F, Herrera PL, Huarte J, Nichols A, Montesano R, et al. (1994) TGF-beta 1 influences the relative development of the exocrine and endocrine pancreas in vitro. Development 120: 3451–3462.
  2. 2. Miralles F, Battelino T, Czernichow P, Scharfmann R (1998) TGF-beta plays a key role in morphogenesis of the pancreatic islets of Langerhans past controlling the activity of the matrix metalloproteinase MMP-2. J Cell Biol 143: 827–836.
  3. 3. Smart NG, Gilthorpe AA, Gu X, Harmon EB, Topper JN, et al. (2006) Conditional expression of smad7 in pancreatic β-cells disrupts TGF-β signaling and induces reversible diabetes mellitus. PLoS Biol 4(2): e39.
  4. 4. LeRoith D (2002) Beta-jail cell dysfunction and insulin resistance in blazon ii diabetes: Role of metabolic and genetic abnormalities. Am J Med 113: Suppl 6A3S–11S.
  5. v. Kahn SE (2000) The importance of the beta-cell in the pathogenesis of blazon two diabetes Mellitus. Am J Med 108: Suppl 6a2S–8S.
  6. half-dozen. Ashcroft FM, Rorsman P (2004) Molecular defects in insulin secretion in type-2 diabetes. Rev Endocr Metab Disord 5: 135–142.
  7. 7. Bell GI, Polonsky KS (2001) Diabetes mellitus and genetically programmed defects in beta-cell function. Nature 414: 788–791.
  8. 8. Dean PM, Matthews EK (1968) Electrical activity in pancreatic islet cells. Nature 219: 389–390.
  9. 9. Henquin JC, Meissner HP (1984) Significance of ionic fluxes and changes in membrane potential for stimulus-secretion coupling in pancreatic B-cells. Experientia 40: 1043–1052.
  10. 10. Ashcroft FM, Rorsman P (1989) Electrophysiology of the pancreatic beta-prison cell. Prog Biophys Mol Biol 54: 87–143.
  11. 11. Rorsman P, Renstrom Eastward (2003) Insulin granule dynamics in pancreatic beta cells. Diabetologia 46: 1029–1045.
  12. 12. Matthews EK, Sakamoto Y (1975) Electrical characteristics of pancreatic islet cells. J Physiol 246: 421–437.
  13. xiii. Curry DL, Bennett LL, Grodsky GM (1968) Requirement for calcium ion in insulin secretion by the perfused rat pancreas. Am J Physiol 214: 174–178.
  14. 14. Hales CN, Milner RD (1968) Cations and the secretion of insulin from rabbit pancreas in vitro. J Physiol 199: 177–187.
  15. 15. Ashcroft SJ, Sugden MC, Williams IH (1980) Carbohydrate metabolism and the glucoreceptor machinery. Horm Metab Res (Suppl 10)1–7.
  16. sixteen. MacDonald PE, Joseph JW, Rorsman P (2005) Glucose-sensing mechanisms in pancreatic beta-cells. Philos Trans R Soc Lond B Biol Sci 360: 2211–2225.
  17. 17. Rorsman P, Trube One thousand (1985) Glucose dependent 1000+-channels in pancreatic beta-cells are regulated by intracellular ATP. Pflugers Arch 405: 305–309.
  18. xviii. Malaisse WJ, Boschero Air conditioning (1977) Calcium antagonists and islet function. Eleven. Consequence of nifedipine. Horm Res 8: 203–209.
  19. 19. Schulla V, Renstrom E, Feil R, Feil S, Franklin I, et al. (2003) Impaired insulin secretion and glucose tolerance in β cell-selective Cav1.2 Ca2+ aqueduct zero mice. EMBO J 22: 3844–3854.
  20. 20. Jing X, Li DQ, Olofsson CS, Salehi A, Surve VV, et al. (2005) Cav2.3 calcium channels command second-phase insulin release. J Clin Invest 115: 146–154.
  21. 21. Chan CB, De Leo D, Joseph JW, McQuaid TS, Ha XF, et al. (2001) Increased uncoupling protein-2 levels in beta-cells are associated with impaired glucosestimulated insulin secretion: Mechanism of activity. Diabetes 50: 1302–1310.
  22. 22. Zhang C, Baffy One thousand, Perret P, Krauss S, Peroni O, et al. (2001) Uncoupling poly peptide-2 negatively regulates insulin secretion and is a major link between obesity, beta cell dysfunction and blazon two diabetes. Cell 105: 745–755.
  23. 23. Bordone L, Motta MC, Picard F, Robinson A, Jhala United states, et al. (2006) Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic β cells. PLoS Biol iv(ii): e31.
  24. 24. Santos RM, Rosario LM, Nadal A, Garcia-Sancho J, Soria B, et al. (1991) Widespread synchronous [Ca2+]i oscillations due to bursting electrical activity in single pancreatic islets. Pflugers Arch 1991 418: 417–422.
  25. 25. Barbosa RM, Silva AM, Tome AR, Stamford JA, Santos RM, et al. (1998) Control of pulsatile 5-HT/insulin secretion from unmarried mouse pancreatic islets past intracellular calcium dynamics. J Physiol 510: 135–143. Pt 1.
  26. 26. Bergsten P (1995) Tedious and fast oscillations of cytoplasmic Ca2+ in pancreatic islets stand for to pulsatile insulin release. Am J Physiol 268: E282–E287.
  27. 27. Gilon P, Henquin JC (1992) Influence of membrane potential changes on cytoplasmic Ca2+ concentration in an electrically excitable cell, the insulin-secreting pancreatic B-jail cell. J Biol Chem 267: 20713–20720.
  28. 28. Zhang M, Houamed K, Kupershmidt Southward, Roden D, Satin LS (2005) Pharmacological backdrop and functional role of Kslow current in mouse pancreatic beta-cells: SK channels contribute to Kslow tail current and modulate insulin secretion. J Gen Physiol 126: 353–363.
  29. 29. Kanno T, Rorsman P, Gopel SO (2002) Glucose-dependent regulation of rhythmic action potential firing in pancreatic β cells by KATP-channel modulation. J Physiol 545: 501–507.
  30. 30. Gopel And then, Kanno T, Barg Southward, Eliasson L, Galvanovskis J, et al. (1999) Activation of Ca2+-dependent K+ channels contributes to rhythmic firing of activeness potentials in mouse pancreatic beta cells. J Gen Physiol 114: 759–770.
  31. 31. Grapengiesser East, Gylfe E, Hellman B (1991) Cyclic AMP equally a determinant for glucose induction of fast Ca2+ oscillations in isolated pancreatic beta-cells. J Biol Chem 266: 12207–12210.
  32. 32. Smolen P, Rinzel J, Sherman A (1993) Why pancreatic islets burst but unmarried beta cells do non. The heterogeneity hypothesis. Biophys J 64: 1668–1680.
  33. 33. Eddlestone GT, Goncalves A, Bangham JA, Rojas E (1984) Electric coupling between cells in islets of Langerhans from mouse. J Membr Biol 77: i–14.
  34. 34. Meissner HP (1976) Electrophysiological bear witness for coupling between beta cells of pancreatic islets. Nature 262: 502–504.
  35. 35. Ravier MA, Guldenagel M, Charollais A, Gjinovci A, Caille D, et al. (2005) Loss of connexin36 channels alters beta-jail cell coupling, islet synchronization of glucose-induced Ca2+ and insulin oscillations, and basal insulin release. Diabetes 54: 1798–1807.
  36. 36. Gopel Due south, Kanno T, Barg Southward, Galvanovskis J, Rorsman P (1999) Voltage-gated and resting membrane currents recorded from â-cells in intact mouse pancreatic islets. J Physiol 521: 717–728.
  37. 37. Nadal A, Quesada I, Soria B (1999) Homologous and heterologous asynchronicity between identified blastoff-, beta- and delta-cells within intact islets of Langerhans in the mouse. J Physiol 517: 85–93. Pt one.
  38. 38. Meda P, Santos RM, Atwater I (1986) Directly identification of electrophysiologically monitored cells inside intact mouse islets of Langerhans. Diabetes 35: 232–236.
  39. 39. Michaels RL, Sheridan JD (1981) Islets of Langerhans: Dye coupling among immunocytochemically distinct cell types. Science 214: 801–803.
  40. 40. Quesada I, Fuentes Eastward, Andreu E, Meda P, Nadal A, et al. (2003) On-line analysis of gap junctions reveals more efficient electric than dye coupling between islet cells. Am J Physiol Endocrinol Metab 284: E980–E987.
  41. 41. Rocheleau JV, Remedi MS, Granada B, Head WS, Koster JC, et al. (2005) Critical role of coupled KATP channel activity for regulated insulin secretion. PLoS Biol 4(ii): e26.
  42. 42. Sherman A, Rinzel J, Keizer J (1988) Emergence of organized bursting in clusters of pancreatic beta-cells by channel sharing. Biophys J 54: 411–425.
  43. 43. Maechler P, Wollheim CB (2001) Mitochondrial function in normal and diabetic betacells. Nature 414: 807–812.

collinssperwit.blogspot.com

Source: https://journals.plos.org/plosbiology/article?id=10.1371%2Fjournal.pbio.0040049

0 Response to "what term best describes a failure of the body’s cells to respond to secretion of insulin?"

إرسال تعليق

Iklan Atas Artikel

Iklan Tengah Artikel 1

Iklan Tengah Artikel 2

Iklan Bawah Artikel