リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Metalation and activation of Zn²⁺enzymes via early secretory pathway-resident ZNT proteins」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Metalation and activation of Zn²⁺enzymes via early secretory pathway-resident ZNT proteins

Kambe, Taiho Wagatsuma, Takumi 京都大学 DOI:10.1063/5.0176048

2023.12

概要

Zinc (Zn²⁺), an essential trace element, binds to various proteins, including enzymes, transcription factors, channels, and signaling molecules and their receptors, to regulate their activities in a wide range of physiological functions. Zn²⁺ proteome analyses have indicated that approximately 10% of the proteins encoded by the human genome have potential Zn²⁺ binding sites. Zn²⁺binding to the functional site of a protein (for enzymes, the active site) is termed Zn²⁺metalation. In eukaryotic cells, approximately one-third of proteins are targeted to the endoplasmic reticulum; therefore, a considerable number of proteins mature by Zn²⁺metalation in the early secretory pathway compartments. Failure to capture Zn²⁺ in these compartments results in not only the inactivation of enzymes (apo-Zn²⁺ enzymes), but also their elimination via degradation. This process deserves attention because many Zn²⁺ enzymes that mature during the secretory process are associated with disease pathogenesis. However, how Zn²⁺is mobilized via Zn²⁺ transporters, particularly ZNTs, and incorporated in enzymes has not been fully elucidated from the cellular perspective and much less from the biophysical perspective. This review focuses on Zn²⁺ enzymes that are activated by Zn²⁺ metalation via Zn²⁺ transporters during the secretory process. Further, we describe the importance of Zn²⁺ metalation from the physiopathological perspective, helping to reveal the importance of understanding Zn²⁺ enzymes from a biophysical perspective.

この論文で使われている画像

参考文献

Fukada, T. & Kambe, T. Molecular and genetic features of zinc transporters in

physiology and pathogenesis. Metallomics 3, 662-674 (2011).

10

11

12

Kambe, T. Metalation and maturation of zinc ectoenzymes: A perspective.

Biochemistry 59, 74-79 (2020).

Andreini, C., Banci, L., Bertini, I. & Rosato, A. Counting the zinc-proteins encoded in

the human genome. J Proteome Res 5, 196-201 (2006).

Kambe, T., Tsuji, T., Hashimoto, A. & Itsumura, N. The physiological, biochemical,

and molecular roles of zinc transporters in zinc homeostasis and metabolism. Physiol

Rev 95, 749-784 (2015).

Kambe, T., Taylor, K. M. & Fu, D. Zinc transporters and their functional integration

in mammalian cells. J Biol Chem 296, 100320 (2021).

Kambe, T., Hashimoto, A. & Fujimoto, S. Current understanding of ZIP and ZnT zinc

transporters in human health and diseases. Cell Mol Life Sci 71, 3281-3295 (2014).

Hara, T., Yoshigai, E., Ohashi, T. & Fukada, T. Zinc transporters as potential

therapeutic targets: An updated review. J Pharmacol Sci 148, 221-228 (2022).

Gupta, S. et al. Visualizing the kinetic power stroke that drives proton-coupled

zinc(II) transport. Nature 512, 101-104 (2014).

Lopez-Redondo, M. L., Coudray, N., Zhang, Z., Alexopoulos, J. & Stokes, D. L.

Structural basis for the alternating access mechanism of the cation diffusion facilitator

YiiP. Proc Natl Acad Sci U S A 115, 3042-3047 (2018).

Golan, Y., Alhadeff, R., Warshel, A. & Assaraf, Y. G. ZnT2 is an electroneutral

proton-coupled vesicular antiporter displaying an apparent stoichiometry of two

protons per zinc ion. PLoS Comput Biol 15, e1006882 (2019).

Ohana, E. et al. Identification of the Zn2+ binding site and mode of operation of a

mammalian Zn2+ transporter. J Biol Chem 284, 17677-17686 (2009).

Coudray, N. et al. Inward-facing conformation of the zinc transporter YiiP revealed

by cryoelectron microscopy. Proc Natl Acad Sci U S A 110, 2140-2145 (2013).

13

14

15

16

17

Xue, J., Xie, T., Zeng, W., Jiang, Y. & Bai, X.-C. Cryo-EM structures of human ZnT8

in both outward- and inward-facing conformations. eLife 9, e58823 (2020).

Zhang, S. et al. Cryo-EM structure of a eukaryotic zinc transporter at a low pH

suggests its Zn(2+)-releasing mechanism. J Struct Biol 215, 107926 (2023).

Bui, H. B. et al. Cryo-EM structures of human zinc transporter ZnT7 reveal the

mechanism of Zn(2+) uptake into the Golgi apparatus. Nat Commun 14, 4770 (2023).

Lu, M. & Fu, D. Structure of the zinc transporter YiiP. Science 317, 1746-1748

(2007).

Lu, Y. J., Liu, Y. C., Lin, M. C., Chen, Y. T. & Lin, L. Y. Coordinative modulation of

human zinc transporter 2 gene expression through active and suppressive regulators. J

10

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

18

19

20

21

22

23

24

25

26

27.

28.

Nutr Biochem 26, 351-359 (2015).

Wiuf, A. et al. The two-domain elevator-type mechanism of zinc-transporting ZIP

proteins. Sci Adv 8, eabn4331 (2022).

Zhang, Y. et al. Structural insights into the elevator-type transport mechanism of a

bacterial ZIP metal transporter. Nat Commun 14, 385 (2023).

Pang, C., Chai, J., Zhu, P., Shanklin, J. & Liu, Q. Structural mechanism of

intracellular autoregulation of zinc uptake in ZIP transporters. Nat Commun 14, 3404

(2023).

Zhang, T. et al. Crystal structures of a ZIP zinc transporter reveal a binuclear metal

center in the transport pathway. Sci Adv 3, e1700344 (2017).

Zhang, T., Sui, D., Zhang, C., Cole, L. & Hu, J. Asymmetric functions of a binuclear

metal center within the transport pathway of a human zinc transporter ZIP4. FASEB J

34, 237-247 (2020).

Gaither, L. A. & Eide, D. J. Functional expression of the human hZIP2 zinc

transporter. J Biol Chem 275, 5560-5564 (2000).

Girijashanker, K. et al. Slc39a14 gene encodes ZIP14, a metal/bicarbonate symporter:

similarities to the ZIP8 transporter. Mol Pharmacol 73, 1413-1423 (2008).

Hoch, E., Levy, M., Hershfinkel, M. & Sekler, I. Elucidating the H(+) coupled Zn(2+)

transport mechanism of ZIP4; implications in acrodermatitis enteropathica. Int J Mol

Sci 21, 734 (2020).

Gupta, S., Merriman, C., Petzold, C. J., Ralston, C. Y. & Fu, D. Water molecules

mediate zinc mobility in the bacterial zinc diffusion channel ZIPB. J Biol Chem 294,

13327-13335 (2019).

Wang, Y. et al. The cellular economy of the Saccharomyces cerevisiae zinc proteome.

Metallomics 10, 1755-1776 (2018).

Bird, A. J. & Wilson, S. Zinc homeostasis in the secretory pathway in yeast. Curr

Opin Chem Biol 55, 145-150 (2020).

29

Suzuki, T. et al. Two different zinc transport complexes of cation diffusion facilitator

30

proteins localized in the secretory pathway operate to activate alkaline phosphatases

in vertebrate cells. J Biol Chem 280, 30956-30962 (2005).

Fukunaka, A. et al. Demonstration and characterization of the heterodimerization of

31

32

ZnT5 and ZnT6 in the early secretory pathway. J Biol Chem 284, 30798-30806

(2009).

Ellis, C. D. et al. Zinc and the Msc2 zinc transporter protein are required for

endoplasmic reticulum function. J Cell Biol 166, 325-335 (2004).

Ellis, C. D., Macdiarmid, C. W. & Eide, D. J. Heteromeric protein complexes mediate

zinc transport into the secretory pathway of eukaryotic cells. J Biol Chem 280, 2881128818 (2005).

11

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

33

Fang, Y. et al. Cation diffusion facilitator Cis4 is implicated in Golgi membrane

trafficking via regulating zinc homeostasis in fission yeast. Mol Biol Cell 19, 12951303 (2008).

34

Choi, S., Hu, Y. M., Corkins, M. E., Palmer, A. E. & Bird, A. J. Zinc transporters

belonging to the Cation Diffusion Facilitator (CDF) family have complementary roles

in transporting zinc out of the cytosol. PLoS Genet 14, e1007262 (2018).

Kowada, T. et al. Quantitative imaging of Labile Zn(2+) in the Golgi apparatus using

a localizable small-molecule fluorescent probe. Cell Chem Biol 27, 1521-1531 e1528

35

36

37

38

39

40

41

42

43

(2020).

Amagai, Y. et al. Zinc homeostasis governed by Golgi-resident ZnT family members

regulates ERp44-mediated proteostasis at the ER-Golgi interface. Nat Commun 14,

2683 (2023).

Lieberwirth, J. K. et al. Bi-allelic loss of function variants in SLC30A5 as cause of

perinatal lethal cardiomyopathy. Eur J Hum Genet 29, 808-815 (2021).

Penon-Portmann, M. et al. De novo heterozygous variants in SLC30A7 are a

candidate cause for Joubert syndrome. Am J Med Genet A 188, 2360-2366 (2022).

Huang, L. et al. Identification of novel compound heterozygous variants in the

SLC30A7 (ZNT7) gene in two French brothers with stunted growth, testicular

hypoplasia, and bone marrow failure. Hum Mol Genet 2016-2031 (2023).

Watanabe, S. et al. Zinc regulates ERp44-dependent protein quality control in the

early secretory pathway. Nat Commun 10, 603 (2019).

Saito, Y., Ihara, Y., Leach, M. R., Cohen-Doyle, M. F. & Williams, D. B. Calreticulin

functions in vitro as a molecular chaperone for both glycosylated and nonglycosylated proteins. Embo J 18, 6718-6729 (1999).

Ohashi, W. et al. Zinc transporter SLC39A7/ZIP7 promotes intestinal epithelial selfrenewal by resolving ER stress. PLoS Genet 12, e1006349 (2016).

Bin, B. H. et al. Requirement of Zinc transporter SLC39A7/ZIP7 for dermal

development to fine-tune endoplasmic reticulum function by regulating protein

44

45

46

47

disulfide isomerase. J Invest Dermatol 137, 1682-1691 (2017).

Woodruff, G. et al. The zinc transporter SLC39A7 (ZIP7) is essential for regulation of

cytosolic zinc levels. Mol Pharmacol 94, 1092-1100 (2018).

Anzilotti, C. et al. An essential role for the Zn(2+) transporter ZIP7 in B cell

development. Nat Immunol 20, 350-361 (2019).

Ishihara, K. et al. Zinc transport complexes contribute to the homeostatic maintenance

of secretory pathway function in vertebrate cells. J Biol Chem 281, 17743-17750

(2006).

Kinoshita, T. Biosynthesis and biology of mammalian GPI-anchored proteins. Open

Biol 10, 190290 (2020).

12

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

48

Wagatsuma, T. et al. Zinc transport via ZNT5-6 and ZNT7 is critical for cell surface

glycosylphosphatidylinositol-anchored protein expression. J Biol Chem 298, 102011

(2022).

49

Galperin, M. Y. & Jedrzejas, M. J. Conserved core structure and active site residues in

alkaline phosphatase superfamily enzymes. Proteins 45, 318-324 (2001).

Mann, K. J. & Sevlever, D. 1,10-Phenanthroline inhibits glycosylphosphatidylinositol

anchoring by preventing phosphoethanolamine addition to

glycosylphosphatidylinositol anchor precursors. Biochemistry 40, 1205-1213 (2001).

50

51

52

53

54

55

56

57

58

Nakano, Y. et al. Biogenesis of GPI-anchored proteins is essential for surface

expression of sodium channels in zebrafish Rohon-Beard neurons to respond to

mechanosensory stimulation. Development 137, 1689-1698 (2010).

Schuchman, E. H. Acid sphingomyelinase, cell membranes and human disease:

lessons from Niemann-Pick disease. FEBS Lett 584, 1895-1900 (2010).

Ueda, S. et al. Early secretory pathway-resident Zn transporter proteins contribute to

cellular sphingolipid metabolism through activation of sphingomyelin

phosphodiesterase 1. Am J Physiol Cell Physiol 322, C948-C959 (2022).

Yamaguchi, Y. & Hearing, V. J. Melanocytes and their diseases. Cold Spring Harb

Perspect Med 4, a017046 (2014).

Solano, F. On the metal cofactor in the tyrosinase family. Int J Mol Sci 19, 633

(2018).

Lai, X., Wichers, H. J., Soler-Lopez, M. & Dijkstra, B. W. Structure and function of

human tyrosinase and tyrosinase-related proteins. Chemistry 24, 47-55 (2018).

Aguilera, F., McDougall, C. & Degnan, B. M. Origin, evolution and classification of

type-3 copper proteins: lineage-specific gene expansions and losses across the

Metazoa. BMC Evol Biol 13, 96 (2013).

Decker, H. & Tuczek, F. The recent crystal structure of human tyrosinase related

protein 1 (HsTYRP1) solves an old problem and poses a new one. Angew Chem Int

Ed Engl 56, 14352-14354 (2017).

59

60

Petris, M. J., Strausak, D. & Mercer, J. F. The Menkes copper transporter is required

for the activation of tyrosinase. Hum Mol Genet 9, 2845-2851 (2000).

Wagatsuma, T. et al. Pigmentation and TYRP1 expression are mediated by zinc

62

through the early secretory pathway-resident ZNT proteins. Commun Biol 6, 403

(2023).

Murisier, F. & Beermann, F. Genetics of pigment cells: lessons from the tyrosinase

gene family. Histol Histopathol 21, 567-578 (2006).

Puckett, E. E. et al. Genetic architecture and evolution of color variation in American

63

black bears. Curr Biol 33, 86-97 e10 (2023).

Li, J. et al. A missense mutation in TYRP1 causes the chocolate plumage color in

61

13

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

64

65

66

67

68

69

70

71

72

73

74

chicken and alters melanosome structure. Pigment Cell Melanoma Res 32, 381-390

(2019).

Braasch, I., Liedtke, D., Volff, J. N. & Schartl, M. Pigmentary function and evolution

of tyrp1 gene duplicates in fish. Pigment Cell Melanoma Res 22, 839-850 (2009).

Brown, A. R. et al. A community-science approach identifies genetic variants

associated with three color morphs in ball pythons (Python regius). PLoS One 17,

e0276376 (2022).

Lai, X., Wichers, H. J., Soler-Lopez, M. & Dijkstra, B. W. Structure of human

tyrosinase related protein 1 reveals a binuclear zinc active site important for

melanogenesis. Angew Chem Int Ed Engl 56, 9812-9815 (2017).

Lerner, A. B., Fitzpatrick, T. B., Calkins, E. & Summerson, W. H. Mammalian

tyrosinase; the relationship of copper to enzymatic activity. J Biol Chem 187, 793-802

(1950).

Allen, T. H. & Bodine, J. H. Enzymes in ontogenesis (Orthoptera). Xvii. The

importance of copper for protyrosinase. Science 94, 443-444 (1941).

Supuran, C. T. Carbonic anhydrases: novel therapeutic applications for inhibitors and

activators. Nat Rev Drug Discov 7, 168-181 (2008).

Keilin, D. & Mann, T. Carbonic anhydrase. Nature 144, 442-443 (1939).

Swietach, P., Hulikova, A., Vaughan-Jones, R. D. & Harris, A. L. New insights into

the physiological role of carbonic anhydrase IX in tumour pH regulation. Oncogene

29, 6509-6521 (2010).

Neri, D. & Supuran, C. T. Interfering with pH regulation in tumours as a therapeutic

strategy. Nat Rev Drug Discov 10, 767-777 (2011).

Tsuji, T. et al. Dissecting the process of activation of cancer-promoting zinc-requiring

ectoenzymes by zinc metalation mediated by ZNT transporters. J Biol Chem 292,

2159-2173 (2017).

Fukunaka, A. et al. Tissue nonspecific alkaline phosphatase is activated via a two-step

mechanism by zinc transport complexes in the early secretory pathway. J Biol Chem

75

76

77

78

79

286, 16363-16373 (2011).

Suzuki, E. et al. Detailed analyses of the crucial functions of Zn transporter proteins

in alkaline phosphatase activation. J Biol Chem 295, 5669-5684 (2020).

Takeda, T. A. et al. Zinc deficiency causes delayed ATP clearance and adenosine

generation in rats and cell culture models. Commun Biol 1, 113 (2018).

Haase, H. & Rink, L. Zinc signals and immune function. Biofactors 40, 27-40 (2014).

Kawamura, T. et al. Severe dermatitis with loss of epidermal Langerhans cells in

human and mouse zinc deficiency. J Clin Invest 122, 722-732 (2012).

Antonioli, L., Blandizzi, C., Pacher, P. & Hasko, G. Immunity, inflammation and

cancer: a leading role for adenosine. Nat Rev Cancer 13, 842-857 (2013).

14

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

80

81

82

83.

84.

85.

Cekic, C. & Linden, J. Purinergic regulation of the immune system. Nat Rev Immunol

16, 177-192 (2016).

Nolin, E. et al. Discovery of a ZIP7 inhibitor from a Notch pathway screen. Nat Chem

Biol 15, 179-188 (2019).

Pujol-Gimenez, J. et al. Inhibitors of human divalent metal transporters DMT1

(SLC11A2) and ZIP8 (SLC39A8) from a GDB-17 fragment library. ChemMedChem

16, 3306-3314 (2021).

Hessels, A. M. et al. eZinCh-2: A Versatile, Genetically Encoded FRET Sensor for

Cytosolic and Intraorganelle Zn(2+) Imaging. ACS Chem Biol 10, 2126-2134 (2015).

Hessels, A. M., Taylor, K. M. & Merkx, M. Monitoring cytosolic and ER Zn(2+) in

stimulated breast cancer cells using genetically encoded FRET sensors. Metallomics

8, 211-217 (2016).

Liu, R. et al. Organelle-Level Labile Zn(2+) Mapping Based on Targetable

Fluorescent Sensors. ACS Sens 7, 748-757 (2022).

Figure legends

Figure 1. Subcellular localization of ZNT and ZIP proteins. A. Most ZIPs function to

transport extracellular Zn2+ into the cytosol, whereas most ZNTs function to mobilize cytosolic

Zn2+ into the lumen of intracellular compartments, including the endoplasmic reticulum (ER),

Golgi apparatus, trans-Golgi network (TGN), and endosomes, as well as synaptic vesicles,

secretory vesicles, and insulin granules in specialized cells. ZNT10 is not shown because it

functions as a Mn2+ transporter. B. ZNTs transport Zn2+ in a rocker-switch manner, whereas

ZIPs use an elevator-type mechanism.

Figure 2. ZNT5-6 and ZNT7 contribute to the stabilization and metalation of Zn2+

enzymes in the early secretory pathway. ZNT5-6 and ZNT7 function to transport Zn2+ into

the lumen of the early secretory pathway compartments (ER and Golgi) to supply Zn2+ to apoenzymes. Apo-enzymes are thus converted into holo-enzymes. This conversion is required for

the activation and stabilization of a number of enzymes (listed in Table 1).

Figure 3. Proposed labile Zn2+ concentrations in subcellular compartments in cells. The

labile Zn2+ concentration is maintained at a very low level within cells, although it may vary

among different cell types and fluctuate in response to various stimuli. The measured

concentrations, determined using a FRET-based sensor (eZinCh-2, highlighted in light

green)83,84 and a fluorescent type probe (ZnDA, highlighted in light yellow)36,85, are indicated

15

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

as representative examples. Notably, vesicular Zn2+ concentrations, such as those found in

synaptic vesicles and insulin granules (refer to Figure 1), known to accumulate high amounts

of labile Zn2+, are not shown. The omission is due to the ongoing need for further investigation

to accurately determine their concentrations.

Figure 4. Phenotypes of medaka fish (Oryzias latipes) with disrupted Znt5 and/or Znt7. A.

Abnormal tactile phenotype in medaka fish. A mechanosensory stimulation induced swimming

away in WT, Znt5+/−;Znt7+/−, and Znt5+/−;Znt7−/− (three upper rows), but not in Znt5−/−;Znt7+/−

medaka. Znt5−/−;Znt7+/− medaka did not respond to touch (for 0–3 s). The figure shown in this

panel was taken from reference (48). B. Znt5-6 and Znt7 are required for melanogenesis in

medaka fish. Lateral views of Znt5−/−;Znt7−/− embryos at 8–9 days post-fertilization (left) and

of Znt5+/−;Znt7−/− (right) embryos before hatching (upper panels). Melanin content was

decreased in Znt5−/−;Znt7−/−medaka compared to that in Znt5+/−;Znt7−/− littermates (lower

panel). The images in these panels were taken from reference (60).

Figure 5. Zn2+ enzymes involved in extracellular adenine nucleotide metabolism. ATP

released extracellularly is hydrolyzed to ADP, AMP, and adenosine by several enzymes. Among

them, TNAP, CD73/NT5E, and ENPPs (ENPP1 and ENPP3) are Zn2+ enzymes. Extracellular

ATP and ADP bind to ionotropic P2X and metabotropic P2Y receptors and adenosine binds to

P1 receptors to transmit signals that have opposing effects (inflammation vs. antiinflammation).

16

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

ZNT1

ZIP1-6, ZIP8, ZIP10, ZIP12, ZIP14

ZNT4, ZNT5-ZNT6, ZNT7

Golgi

ZIP9, ZIP13

ZNT3

Synaptic vesicle

ZIP7

Insulin granule

ZNT

Extracellular/Lumen

Zn

ZNT2

Secretory vesicle

ZNT8

ER

ZNT4

Endosome

Zn

Cytosol

ZIP

Extracellular/Lumen

Zn

ZNT9

ZIP11

Nucleus

Mitochondria

Zn

Cytosol

Fig. 1

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

Stabilization

Activation

Zn

Zn

Holo

ZNT6

Metalation

Zn

Golgi

ZNT5

ZNT7

Zn

Apo

ER

Fig. 2

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

Cytosol

0.83 nM

0.87 nM

0.22 nM

Nucleus

0.13 nM

0.11 nM

0.2 nM

0.8 nM

Golgi

Mitochondria

0.17 nM

ER

3.3 pM

trans:

14 pM

∼80 nM

∼60 nM

cis: ∼80 nM

pre-cis: ∼100 nM

60 pM

medial:

Fig. 3

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

Znt5+/-;Znt7-/-

Znt5-/-;Znt7+/Melanin (µg) / Protein (µg)

Znt5-/-;Znt7-/- Znt5+/-;Znt7-/-

WT

Znt5+/-;Znt7+/1.0 mm

0s

1s

2s

3s

0.025

0.020

**

0.020

0.010

0.005

Znt5+/-;Znt7-/- Znt5-/-;Znt7-/-

Fig. 4

This is the author’s peer reviewed, accepted manuscript. However, the online version of record will be different from this version once it has been copyedited and typeset.

PLEASE CITE THIS ARTICLE AS DOI: 10.1063/5.0176048

Zn

ENPP1, ENPP3

ATP

Zn

TNAP

P2X

Zn

Zn

ADP

TNAP

Inflammation

Zn

AMP

CD73

TNAP

Zn

Adenosine

P2Y

P1

Cytosol

anti-Inflammation

Fig. 5

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る