Etiketter

måndag 14 oktober 2019

Terminologiaa: hampaiden vieruskudoksen sairaudet

KÄYTETTÄVÄ TERMI
periodontaalinen atrofia  
KÄYTTÖHUOMAUTUS
MUUNKIELISET TERMIT
Periodontal Atrophy
englanti
Atrophy of Periodontium
Periodontium Atrophies
Periodontium Atrophy
Parodontal atrofi

Hampaattomuus (edentata) , proteesit, hammasinfektiot ja röntgenlöydöt . Suomal. tutkimus 2014.

Suomalainen tutkimus http://jultika.oulu.fi/files/isbn9789526206141.pdf
Haikola Bb (2014) Oral health among Finns  aged 60 years and  older,
Edentulousness, fixed prostheses, dental infections detected from radiographs and their association factors.
Vanhusväestön suun terveydentila on useassa tutkimuksessa todettu huonommaksi kuin nuoremmilla. Tämän tutkimuksen tarkoituksena oli selvittää suomalaisen ikääntyvän väestön suun ter-veydentilaa ja siihen liittyviä tekijöitä. Epidemiologinen poikkileikkaustutkimus tehtiin Kirkkonummella Etelä-Suomessa ja Lakeuden terveyskeskuksen alueella Pohjois-Suomessa. Tutkimukseen ja haastatteluun osallistuiyhteensä 1191 iältään 60–78-vuotiasta henkilöä, 625 K:lta ja 566 L:ltä. Kliinisessä tutkimuksessa selvitettiin hampaattomuus ja proteesien purentatasapaino. Alaleuan leuka-kulma, nousevan haaran ja nivellisäkkeen korkeus, hammasharjanteen korkeus ja kiinteiden proteesien esiintyminen arvioitiin panoraamaröntgenkuvista (pantomografia). Tulehdusriskiä kuvaava indeksi arvioitiin röntgenkuvien tulehdusmuutoksista, joina rekisteröitiin syvä karies, hampaan juurenpääntulehdus, tulehdus takahampaiden juurten haarautumassa, syventyneet luutaskut ja kiinnityskudoskato usean hampaan alueella. Hampaattomuuden vallitsevuus oli 37 %. Se oli tilastollisesti merkitsevästi korkeampi naisilla kuin miehillä ja pohjoisella alueella merkitsevästi korkeampi kuin eteläisellä. Hampaattomuus oli yleisempää vähiten koulutusta saaneilla kuin korkeamman koulutuksen saaneilla. Hampaallisilla oli pienempi alaleuan leukakulma kuin hampaattomilla, ja heillä oli myös korkeampi nouseva haara ja nivellisäke kuin hampaattomilla. Naisilla oli merkitsevästi useammin vakavaa hammasharjanteen madaltumista kuin miehillä. Molemmilla sukupuolilla tyytymättömyys proteeseihin liittyi merkitsevästi huonoon purentatasapainoon. Yli kolmella neljäsosalla tutkituistaoli vakava tulehdusriski, eteläisellä alueella enemmän kuin pohjoisella. Hampaallisessa aineis-tossa naisilla oli enemmän kiinteitä proteeseja kuin miehillä. K:n alueella tutkituilla oli enemmän kiinteitä proteeseja kuin L:n alueella.Hampaattomuus oli hyvin yleistä 60–78-vuotiailla suomalaisilla. Alaleuanluun muoto muuttui hampaiden menetyksen seurauksena. Kiinteiden proteesien määrä oli vähäinen ja ikääntyvillä suomalaisilla oli röntgenkuvissa runsaasti hammasperäisten tulehdusten löydöksiä. Suun terveyden alueelliset erot tulisi ottaa huomioon suu- ja hammasterveyden edistämistyössä, jossa erityisesti tulisi huomioida alhaisen sosioekonomisen aseman ja huonon terveyden omaava vanhempi väestö. 
Asiasanat: 
 alveolaarinen luukato
hampaan pesäkeinfektio,
 hampaaton suu, (Edentata:  tandloshet )
 ikääntyvä,
 kiinteä proteesi, 
luun uudelleen muotoutuminen
 panoraamaröntgenkuvaus, suunterveys

Hammasta kiinnittävien kudosten kunto, Foolihappo ja Folaattireseptori-1 (Oct 2019) ,

https://www.ncbi.nlm.nih.gov/pubmed/31604439


2019 Oct 11;19(1):218. doi: 10.1186/s12903-019-0909-z.
Folate-receptor 1 level in periodontal disease: a pilot study.
Alkan D1, Guven B2, Turer CC3, Balli U3, Can M2. BACKGROUND:
The purpose of this study was to investigate gingival crevicular fluid (GCF) and serum folate-receptor 1 (FOLR1) levels in subjects with different periodontal status.
METHODS: The study consists of three groups: Healthy group (n = 15), gingivitis group (n = 15) and chronic periodontitis group (n = 15). Clinical periodontal parameters including probing pocket depth (PPD), clinical attachment level (CAL), gingival index (GI) and bleeding on probing (BOP) were assessed. GCF and serum samples were collected from each patient and were analyzed FOLR1 levels by enzyme-linked immunosorbent assay. RESULTS: The values of FOLR1 in GCF were higher in gingivitis and periodontitis groups than among patient in control group (p < 0.016). Serum FOLR1 levels showed no significant difference between the groups. A significant correlation was observed between FOLR1 levels of GCF and BOP (p < 0.05).
CONCLUSIONS:
Our preliminary data suggest that FOLR1 is not useful in monitoring the periodontal disease. Further studies are necessary to clarify the role, regulation and function of folate and it's receptors in the pathogenesis of periodontal disease. KEYWORDS:
Folate-receptor 1; Gingivitis; Periodontitis PMID: 31604439 DOI:

Duodecim: ientulehduksesta ja hammastakiinnittävien kudosten taudeista

Dentistikandin perusohjekirjoja: Kariesoppi

https://www.utupub.fi/bitstream/handle/10024/104369/Kandin%20selviytymisopas%20klinikkaan%20Uusitalo_Havukainen.pdf?sequence=2&isAllowed=y

fredag 12 juli 2019

Metastaattissta luustosairaudesta. Gas6 avainsanojen joukossa

https://www.ncbi.nlm.nih.gov/pubmed/30937279

2018 Nov 6;15:004-4. doi: 10.1016/j.jbo.2018.10.004. eCollection 2019 Apr.
Metastatic bone disease: Pathogenesis and therapeutic options: Up-date on bone metastasis management.

Abstract

Bone metastases (BM) are a common complication of cancer, whose management often requires a multidisciplinary approach. Despite the recent therapeutic advances, patients with BM may still experience skeletal-related events and symptomatic skeletal events, with detrimental impact on quality of life and survival. A deeper knowledge of the mechanisms underlying the onset of lytic and sclerotic BM has been acquired in the last decades, leading to the development of bone-targeting agents (BTA), mainly represented by anti-resorptive drugs and bone-seeking radiopharmaceuticals. Recent pre-clinical and clinical studies have showed promising effects of novel agents, whose safety and efficacy need to be confirmed by prospective clinical trials. Among BTA, adjuvant bisphosphonates have also been shown to reduce the risk of BM in selected breast cancer patients, but failed to reduce the incidence of BM from lung and prostate cancer. Moreover, adjuvant denosumab did not improve BM free survival in patients with breast cancer, suggesting the need for further investigation to clarify BTA role in early-stage malignancies. The aim of this review is to describe BM pathogenesis and current treatment options in different clinical settings, as well as to explore the mechanism of action of novel potential therapeutic agents for which further investigation is needed.

KEYWORDS:

ActRIIA, activin-A type IIA receptor;
BC, breast cancer;
 BM, bone metastases;
BMD, bone mineral density;
BMPs, bone morphogenetic proteins;
BMSC, bone marrow stromal cells;
BPs, bisphosphonates;
BTA, bone targeting agents;
BTM, bone turnover markers;
Bone metastases;
 Bone targeting agents;
CCR, chemokine-receptor;
CRPC, castration-resistant PC;
CXCL-12,
C–X–C motif chemokine-ligand-12;
CXCR-4, chemokine-receptor-4;
DFS, disease-free survival;
DKK1, dickkopf1;
EBC, early BC;
ECM, extracellular matrix;
 ET-1, endothelin-1;
 FDA, food and drug administration;
 FGF, fibroblast growth factor;
GAS6, growth-arrest specific-6;
GFs, growth factors;
GnRH, gonadotropin-releasing hormone;
HER-2, human epidermal growth factor receptor 2;
 HR, hormone receptor;
IL, interleukin;
 LC, lung cancer;
MAPK, mitogen-activated protein kinase;
 MCSF, macrophage colony-stimulating factor;
MCSFR, MCSF receptor;
MIP-1α, macrophage inflammatory protein-1 alpha;
MM, multiple myeloma;
MPC, malignant plasma cells;
N-BPs, nitrogen-containing BPs;
NF-κB, nuclear factor-κB;
ONJ, osteonecrosis of the jaw;
 OS, overall survival;
Osteotropic tumors;
 PC, prostate cancer;
 PDGF, platelet-derived growth factor;
 PFS, progression-free survival;
PIs, proteasome inhibitors;
PSA, prostate specific antigen;
 PTH, parathyroid hormone;
 PTH-rP, PTH related protein;
 QoL, quality of life;
RANK-L, receptor activator of NF-κB ligand;
 RT, radiation therapy;
SREs, skeletal-related events;
SSEs, symptomatic skeletal events;
Skeletal related events;
TGF-β, transforming growth factor β;
TK, tyrosine kinase;
TKIs, TK inhibitors;
 TNF, tumornecrosis factor;
VEGF, vascular endothelial growth factor;
 VEGFR, VEGF receptor;
 mTOR, mammalian target of rapamycin;
 non-N-BPs, non-nitrogen containing BPs;
v-ATPase, vacuolar-type H+ ATPase
PMID:
30937279
PMCID:
PMC6429006
DOI:
10.1016/j.jbo.2018.10.004

onsdag 3 juli 2019

Deubikitinoivat enzyymit ja luun uudelleenmuovautuminen ( A) Artikkelin alkuosa UPS ja DUBs

Stem Cells International
Volume 2018, Article ID 3712083, 9 pages
https://doi.org/10.1155/2018/3712083

Review Article
Deubiquitinating Enzymes and Bone Remodeling
Yu-chen Guo
, Shi-wen Zhang, and Quan Yuan

State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China

Correspondence should be addressed to Quan Yuan; yuanquan@scu.edu.cn

Received 29 March 2018; Accepted 29 May 2018; Published 8 July 2018

Academic Editor: Bo Yu

Copyright © 2018 Yu-chen Guo et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

1. Introduction
The human skeleton undergoes continuous bone remodeling throughout a lifetime [1]. This process initiates with the destruction of mineralized bone, followed by the formation and mineralization of a new bone matrix [1, 2]. This critical process adapts bone architecture and strength to mechanical needs as well as growth. Meanwhile, it repairs microdamage of bone structure and maintains calcium homeostasis [1, 2]. Thus, bone remodeling is pretty important to general health.

To maintain bone homeostasis, bone remodeling is carried out by three main cell lineages: osteoclasts, multinucleated cells differentiate from macrophages and monocytes in the human hematopoietic lineage, resorb mineralized bone, and initiate the bone remodeling cycle [3]; osteoblasts, differentiate from mesenchymal stem cells (MSCs), deposit, and mineralize a new bone matrix [4]; osteocytes, which are the most common cells divided from osteoblasts, serve as a sensing and information transfer system [2]. These cells constitute the basic multicellular unit (BMU) that carries out the bone remodeling cycle. Based on current knowledge, bone remodeling mainly involves the following phases: formation of osteoclasts and resorption of bone, which initiates the cycle; completion of bone resorption followed by recruitment and differentiation of MSCs into osteoblasts; and bone formation mediated by osteoblasts [2]. Thus, the differentiation, function, and interaction of these BMU cells are critical to regulate bone remodeling and maintain bone homeostasis.

Osteoclasts that trigger the bone remodeling cycle are formed by the fusion of mononuclear progenitors in osteoclastogenesis [2]. They exist in a motile state during which they migrate from the bone marrow to the resorption site or a resorptive state performing their bone resorption function [5]. Osteoclasts are derived from the hematopoietic lineage and regulated by several factors [6]. Among these factors, M-SCF and RANKL produced by marrow stromal cells and osteoblasts are essential to promote osteoclastogenesis [2]. Osteoblasts play a key role in bone formation. They arise from MSCs and their differentiation is mainly regulated by transcription factor RUNX2 at the early time. They begin to express osteoblast phenotypic genes and synthesize the bone matrix at a later stage [7, 8]. Then osteoblasts are embedded into the bone matrix as osteocytes or die at the end of their destiny [9]. Several mechanisms including transcription factors, growth factors, hormones, and the extracellular matrix regulate these stages [7, 10]. In the last few years, significant findings have unveiled the mysterious role of the ubiquitin-dependent proteolysis system (UPS) in regulating differentiation and function of osteoclasts as well as osteoblasts [11–13].
 

2. Ubiquitin-Dependent Proteolysis System (UPS)


Ubiquitin
is a highly conserved protein which is made up of 76 amino acids. It is linked to the lysine side chains of target proteins, which results in monoubiquitination or polyubiquitination of the protein. Polyubiquitylated proteins are degraded within a cylindrical multiprotein complex that is named proteasome [14, 15], while monoubiquitination has a variety of ends except proteasomal degradation [14, 15]. For example, the adapter protein TRAF6 contains the RING finger domain which could generate nondegradative K63-linked ubiquitin and contribute to form signaling complexes [16]. This is important to mediate RANK/TRAF6 signaling [17]. To successfully add ubiquitin to target protein, three enzymes involved in this process are essential. The E1 enzyme that recruits ubiquitin is named ubiquitin-activating enzyme. The E2 enzyme, called ubiquitin-conjugating enzyme, transfers the ubiquitin to protein. The E3 enzyme, also known as ubiquitin ligase, acts as a scaffold protein which interacts with the ubiquitin-conjugating enzyme and transfers ubiquitin to protein [18]. Consequently, the UPS affects multiple processes such as protein degradation, cell death, vesicular trafficking, signal transduction, DNA repair, and stress responses [11, 14, 15, 19–23].

The ubiquitin-dependent proteolysis system
plays an important role in mediating bone remodeling. Initially, by inhibiting the proteasomal function through proteasome inhibitor I (PSI), study demonstrated that the UPS is an important regulator of bone turnover and chondrogenesis [24]. And administration of proteasome inhibitor Bortezomib induced MSCs to undergo osteoblastic differentiation partially by modulation of RUNX2 in mice [25]. As a clinically available proteasome inhibitor used in myeloma, Bortezomib is also reported to promote osteoblastogenesis as well as inhibit bone resorption in clinical studies [26, 27]. Following studies demonstrated that these effects are mainly mediated by inhibiting the proteasomal degradation of important proteins, which regulate osteoblast function such as β-catenin [28] and Dkk1 [26]. Another protein stabilized by proteasome inhibitor is Gli2, which promotes bone formation through upregulating bone morphogenetic protein-2 (BMP2) [29, 30].

To date, studies investigating ubiquitin ligase and bone remodeling have demonstrated that several E3 ubiquitin ligases take part in regulation of bone metabolism. For example, the first known ubiquitin ligase affecting bone formation is Smuf1. Smurf1 has been proved to mediate RUNX2 degradation, resulting in downregulated osteoblast differentiation and bone formation [31–35]. Smurf1 also regulates the degradation of Smad1 and downregulates BMP-induced osteogenic differentiation of MSCs [35–37]. Moreover, Smuf1 mediates JunB, MEKK2, and other molecule proteasomal degradation, which causes the inhibition of osteoblast differentiation [32, 38, 39]. Another important ubiquitin ligase which regulates osteoblastogenesis is Cbl. It controls osteoblastogenesis by controlling the ubiquitination and degradation of receptor tyrosine kinases (RTKs), including IGFR, FGFR, and PDGFR [40–43]. Cbl also interacts with Pl3K to regulate bone formation [44–47]. Besides, Itch and Wwp1 are demonstrated to regulate osteogenesis by promoting RUNX2 degradation [48, 49]. On the other hand, E3 ligases also influence osteoclastogenesis and bone resorption. The E3 ligase LNX2 promotes osteoclastogenesis through M-SCF/RANKL signaling as well as the Notch pathway [13]. Another ubiquitin E3 ligase RNF146 inhibits osteoclastogenesis and cytokine production via RANK signaling [50]. As there are over 600 E3 ligases expressed in the human genome, lots of E3 ligases are found to regulate bone remodeling by governing BMU cell differentiation and function.

3. Deubiquitinases (DUBs)


Like other posttranslational modifications, the process of ubiquitination is reversible by the function of deubiquitinases (DUBs) which remove monoubiquitin or polyubiquitin chains from such ubiquitin-modified proteins [51]. Ubiquitin itself is a long-lived protein [52, 53]; thus, it is necessary to remove ubiquitin from proteins for maintaining a sufficient pool of free ubiquitin in the cell to sustain a normal rate of proteolysis. As key hydrolytic emzymes, DUBs hydrolyze the peptide bond that links target protein and ubiquitin [54]. Deubiquitinases are modular proteins which contain catalytic domains, ubiquitin binding domains, and protein-protein interaction domains. Such modules make positive contribution to the recognition of and binding to various chain linkages [55]. To date, about 100 DUBs have been reported to be encoded by the human genome [56, 57] (Table 1). According to their catalytic domains, these DUBs can be classified into five families including 4 thiol protease DUBS (USP, UCH, OUT, and Josephin) and 1 ubiquitin specific metalloproteases (JAMM) [54].
Table 1: Members of deubiquitinases.


Family    Members

USP  

  USPL1, CYLD, USP1, USP2, USP3, USP4, USP5, USP6, USP7, YSP8, USP9x, USP10, USP11, USP12, USP13, USP14, USP15, USP16, USP17L2, USP18, USP19, USP20, USP21, USP22, USP23, USP24, USP25, USP26, USP27, USP28, USP29, USP30, USP31, USP32, USP33, USP34, USP35, USP36, USP37, USP38, USP39, USP40, USP41, USP42, USP43, USP44, USP45, USP46, USP47, USP48, USP49, USP50, USP51, USP52, USP53, USP54

OTU   

 OTUB1, OTUB2, OTUD1, OTUD3, OTUD4, OTUD5, OTUD6A, OTUD6B, OTU1, HIN1L, A20, Cezanne, Cezanne2, TRABID, VCPIP1

UCH

    UCH-L1, UCH-L3, UCH37/UCH-L5, BAP1

Josephin  

  ATXN3, ATXN3L, JOSD1, JOSD2

JAMM/MPN+ 

   BRCC36, CSNS, POH1, AMSH, AMSH-LP, MPND, MYSM1, PRPF8


Deubiquitination has also been reported to be involved in many cellular functions, including DNA repair, protein degradation, cell cycle regulation, stem cell differentiation, and cell signaling [58–69]. Besides, a number of articles demonstrated that DUBs are essential for bone remodeling through regulating related BMU cell differentiation and function [69–78].
..

Deubikitinaasit ja luun uudelleenmuovautuminen (Bone remodellng ) (B) Artikkelin abstrakti ja jatko-osa)

https://www.hindawi.com/journals/sci/2018/3712083/
 Stem Cells International
Volume 2018, Article ID 3712083, 9 pages
https://doi.org/10.1155/2018/3712083

Review Article
Deubiquitinating Enzymes and Bone Remodeling
Yu-chen Guo
, Shi-wen Zhang, and Quan Yuan

State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China

Correspondence should be addressed to Quan Yuan; yuanquan@scu.edu.cn

Received 29 March 2018; Accepted 29 May 2018; Published 8 July 2018

Academic Editor: Bo Yu

Copyright © 2018 Yu-chen Guo et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.



4. Perspective

The ubiquitin-dependent proteolysis system is crucial to cellular functions including skeleton cell functions. The roles of ubiquitin ligases in regulating osteoblast and osteoclast differentiation are well studied, while studies about deubiquitinating enzymes and skeleton cell differentiation are still lacking. In order to delineate the ubiquitin-dependent proteolysis system to regulate bone remodeling, it is important to establish our knowledge about DUBs and bone remodeling. To date, several DUBs are found to regulate osteoblast function (USP4, USP7, USP9x, USP15, UCH-l3, and MYSM1) and osteoclast function (CYLD, USP15, USP18, A20, and POH1) (Table 2). But the mechanisms by which these DUBs regulate skeleton cell functions are not exhaustively described. Future studies should find more DUBs that are involved in BMU cell function and bone remodeling. Importantly, the major challenge is to well describe the actual mechanisms behind these phenotypes. With these novel findings, drugs targeting these DUBs will be designed to treat related skeleton diseases.

Table 2: Deubiquitinases and bone remodeling.
Family    Name    Function    Mechanism    Ref.
USP   
  •   USP4    Inhibits osteoblast differentiation and mineralization    Regulates Wnt signaling by deubiquitinating Dvl, Nik, TGF4, and β-catenin; may regulate TGF/BMP signal    [67–79, 81–85]
  • USP7    Enhances osteogenic differentiation of hASCs    Stabilizes PHF8 that triggers osteogenic differentiation of BMMSCs    [69, 86–88]
  • USP15    Enhances osteoblast-mediated bone formation    Regulates Wnt signaling via deubiquitinating β-catenin    [67, 89–92]
  • CYLD    Inhibits osteoclastogenesis    Regulates RANK signaling through deubiquitinating TRAF6    [97–99]
  • USP18    Inhibits osteoclastogenesis    Regulates IFN signaling by deconjugating ISGlation    [75, 102–104]
  • USP15    Inhibits osteoclastogenesis    Stabilizes IκBα, leading to decreased RANKL-mediated NF-κB activation    [74]
  • USP2    Necessary for PTH (1-34) to induce osteoblast proliferation    Upregulated by the PKA/cAMP pathway and stabilizes PTHR    [105–108]
UCH  
  •  UCH-L3    Increases osteoblast differentiation    Interacts with Smad1 and stabilizes it by deubiquitylating its polyubiquitin    [71]
OTU  
  •   A20 (OTUD7C)    Inhibits osteoclastogenesis    Regulates RANK signaling by controlling TRAF6 and NF-κB degradation    [76, 119–123]
JAMM
  •     POH1    Enhances osteoclast differentiation    Regulates Mitf    [126]
  • MYSM1 (2A DUB)  Enhances osteogenic differentiation    —    [73, 127, 128]

 

3. Deubiquitinases

Like other posttranslational modifications, the process of ubiquitination is reversible by the function of deubiquitinases (DUBs) which remove monoubiquitin or polyubiquitin chains from such ubiquitin-modified proteins [51]. Ubiquitin itself is a long-lived protein [52, 53]; thus, it is necessary to remove ubiquitin from proteins for maintaining a sufficient pool of free ubiquitin in the cell to sustain a normal rate of proteolysis. As key hydrolytic emzymes, DUBs hydrolyze the peptide bond that links target protein and ubiquitin [54]. Deubiquitinases are modular proteins which contain catalytic domains, ubiquitin binding domains, and protein-protein interaction domains. Such modules make positive contribution to the recognition of and binding to various chain linkages [55]. To date, about 100 DUBs have been reported to be encoded by the human genome [56, 57] (Table 1). According to their catalytic domains, these DUBs can be classified into five families including 4 thiol protease DUBS (USP, UCH, OUT, and Josephin) and 1 ubiquitin specific metalloproteases (JAMM) [54].
Table 1: Members of deubiquitinases.


Deubiquitination has also been reported to be involved in many cellular functions, including DNA repair, protein degradation, cell cycle regulation, stem cell differentiation, and cell signaling [58–69]. Besides, a number of articles demonstrated that DUBs are essential for bone remodeling through regulating related BMU cell differentiation and function [69–78].

3.1. Ubiquitin-Specific Protease (USP) and the Bone

The ubiquitin-specific protease family, which contains 56 members in human, is the largest and most diverse family of the DUB families. Consisting of 6 conserved motifs, these USP catalytic domains vary between 295 and 850 residues [57]. Within these 6 motifs, there are two well-conserved motifs that are named Cys-box and His-box. They contain all the necessary catalytic residues [55, 57]. The structure of USP7 is the first well described with three subdomains resembling like a right hand [79]. The thumb and the palm contain Cys-box and His-box, respectively. The cleft between them is the catalytic center. The finger domains can interact with ubiquitin to transfer its C-terminal to the cleft [79]. Then USP5 showed us how UBL domains inserted into a single USP domain to provide additional ubiquitin binding sites which make it possible for the enzyme to bind and disassemble poly-Ub chains [80].

USP is reported to be involved in many cell functions. Most importantly, as the largest family of DUB, USPs are found to regulate bone remodeling by controlling the function of osteoblast, osteoclast, and even PTH.

 3.1.1. USP and Osteoblast

USP4 is found to regulate osteoblast differentiation through the Wnt/β-catenin signaling pathway [70]. The canonical Wnt signaling pathway is essential for osteoblast differentiation and bone formation. A study demonstrates that USP4 inhibits this pathway by deubiquitinating the polyubiquitin chain from Dvl, resulting in inhibiting of Wnt signal and decreased osteoblast differentiation and mineralization [70]. USP4 also deubiquitinates other Wnt signaling components such as Nik and TCF4 [81]. There are also findings indicating that USP4 positively controls β-catenin stability by deubiquitinating, leading to the activation of Wnt signaling [82, 83]. Thus, further researches focusing on USP4 and the Wnt signaling pathway are strongly needed. Besides, USP4 is an important TGF/BMP signaling pathway regulator [69]. After phosphorylation by AKT, USP4 associates with and deubiquitinates ALK5, leading to upregulation of TGFβ signal [84]. In accordance with this finding, USP4 is also reported to interact with Smurf2 and Smad7 [85]. Furthermore, USP4 stabilizes Smad4 through inhibiting its monoubiquitination and enhances activin as well as BMP signaling [86]. Because TGF/BMP signaling plays a pivotal role in osteogenic differentiation of MSCs and bone formation [87], future studies may reveal the essential role of USP4 in control osteoblast differentiation and function through regulating this signaling.


Recently, a study has revealed that USP7 is related to osteogenic differentiation of human adipose-derived stem cells (hASCs) [71]. Like MSCs, hASC is also a stem cell with multilineage differentiation ability, including osteogenic differentiation. USP7 depletion leads to impaired osteogenic differentiation of hASCs. Overexpression of USP7 upregulates hASC osteogenesis. Moreover, knockdown of USP7 results in impaired bone formation in vivo [71]. USP7 acts to ubiquitinate and stabilize PHF8, an epigenetic factor which is essential for stem cell fate determination [88, 89]. Importantly, PHF8 triggers osteogenic differentiation of BMSCs [90]. Thus, the possible mechanism by which USP7 upregulates osteogenic differentiation of hASCs might be that USP7 stabilizes PHF8. A further study is still needed to uncover the actual mechanisms.

USP15, which is highly similar with USP4 [69], also is involved in Wnt signaling and bone formation [91]. USP15 stabilizes β-catenin and enhances Wnt signaling. These processes are initiated by FGF2, which activates MEKK2, causing recruitment of USP15 [91]. USP15 is involved in the TGF/BMP signaling pathway through connecting with ALK3, ALK5, and monoubiquitylated R-SMADs [92–94]. Future studies might reveal the relationship among USP15, TGF/BMP signaling, and osteoblast function.

Interestingly, USP9x, also known as fat facets in mouse (FAM), is closely associated with the TGF/BMP cell signaling pathway, a key signal pathway related to osteogenesis and bone formation. USP9x hydrolyzes Smad4 monoubiquitination [95–97], enhancing TGF-β signal. Moreover, USP9x interacts with the WW domain of Smurf1 and stabilizes it [72]. As told above, Smurf1 plays a pivotal role in osteogenic differentiation and bone formation [31–37]. Likely, USP11 is also involved in the TGF/BMP signaling pathway by deubiquitylating ALK5 [98]. These data suggest the potential direction of future studies.

  3.1.2. USP and Osteoclast

USPs not only control osteogenic differentiation and bone formation but also regulate osteoclast differentiation and function. For example, CYLD inhibits osteoclastogenesis via downregulating RANK signaling [99]. CYLD deubiquitylates TRAF6, which transduces the RANK-mediated signal [99]. By this mechanism, CYLD inhibits osteoclast differentiation, leading to severe osteoporosis in vivo [99]. Using proteasome inhibitors, another study also emphasizes the key role of CYLD in osteoclast formation and function [100]. Furthermore, SCF-TRCP controls the degradation of CYLD itself, which pinpoints SCF-TRCP/CYLD as a pivotal modulator of osteoclastogenesis [101].

USP18 inhibits osteoclastogenesis in mice [77]. IFN signaling negatively influences osteoclastogenesis [102]. Type I IFN stimulates ISG, a ubiquitin-like protein, to express and conjugate to its target ISGylation [103]. Research data demonstrates that USP18 is a negative regulator of IFN signaling via deconjugating ISGylation [104–106]. USP18 deficiency leads to increased RANKL-mediated osteoclastogenesis, resulting in osteopenia phenotype in vivo and in vitro [77].

USP15, which regulates osteoblast function and bone formation, is connected to osteoclast function too [76]. USP15 is the key DUB which cooperates with CHMP5 to stabilize IκBα, leading to decreased RANKL-mediated NF-κB activation and osteoclast differentiation [76]. Taken together, USP15 might be an essential regulator of bone remodeling.

3.1.3. USPs and PTH

In addition to some USPs that regulate osteoblast and/or osteoclast function, there are also some other USPs which collaborate with PTH to influence bone turnover. USP2 was found to be stimulated by PTH in the bone. These osteotropic agents, including PTH, PTHrP, and PGE2, can stimulate USP2 expression selectively in the bone through the PKA/cAMP pathway [107]. A further study revealed that PTH (1-34) could upregulate the expression of USP2 and promote PTHR deubiquitination as well as stabilization [108]. Recently, research data have demonstrated that USP2 is necessary for PTH (1-34) to induce osteoblast proliferation [109]. These findings emphasize the importance of USP2 in PTH mediating anabolic action of bone formation. Another study focusing on the relationship between miRNAs and the PTH level in end-stage renal disease patients demonstrates the close connection between miR-3680-5p and the PTH level. Interestingly, the target genes of miR-3680-5p are USP2, USP6, USP46, and DLT, all of which are members of the UPS [110]. Taken together, USPs may regulate bone turnover via the influence of PTH-associated bone formation. In the future, studies about the details of this interesting mechanism will be the focus.

 3.2. Ubiquitin C-Terminal Hydrolase (UCH) and Bone Formation

The members of the UCH family are several thiol proteases which contain a 230-residue domain as a catalytic core, an N-terminal, and followed by C-terminal extensions which mediate protein to protein interactions sometimes [54]. In human, four UCHs are grouped into smaller UCHs (UCH-L1 and UCH-L3) that prefer to cleave small leaving groups from the C-terminal of Ub and larger UCHs (UCH37 and BAP1) that hydrolyze polyubiquitin chains [54].

Like USPs, UCHs are also reported to have multiple functions [111–113]. Importantly, UCH-L3 deubiquitylates Smad1 and enhances osteoblast differentiation [73]. UCH-L3 physically interacts with Smad1 and stabilizes it by deubiquitylating its polyubiquitin. UCH-L3 promotes the differentiation of osteoblast from C2C12 cells, while knockdown of Uch-l3 delays osteoblast differentiation [73]. Likely, UCH37 is found to connect to Smad7 and reverse Smurf-mediated ubiquitination [114]. Moreover, UCH37 affects TGF-β signaling by connecting to ALK5 [115]. In all, UCH37 influences TGF-β signaling that suggests the role of UCH37 in regulating osteoblast differentiation and function.

 

 

3.3. Ovarian Tumor (OTU) and the Bone

The OTU family was identified based on their homology to the ovarian tumor gene [54]. In human, there are 15 OTUs that are usually grouped into three subclasses: the otubains or OTUBs, the OTUs, and the A20-like OTUs [54].

Among numerous functions of OTUs [116–120], A20 demonstrates the ability to regulate osteoclastogenesis [78, 121, 122]. Bacterial lipopolysaccharides and RANKL induce human peripheral blood mononuclear cells to express A20, which is associated with TRAF6 and NF-κB degradation. Knockdown of A20 results in increased bone resorption [121]. A20 has anti-inflammatory effects as well as antiosteoclastogenic effects [78, 122], which is mainly governed by its attenuation of NF-κB signaling through regulating IKKs [123]. Moreover, A20, which is recruited by Smad6 to TRAF6, plays an important role in inhibition of noncanonical TGF-β signaling [124], indicating its possible regulation of osteoblastogenesis via this main pathway. Besides, like A20, OTUB1 is also involved in TGF-β signaling through deubiquitination of the p-SMAD2/3 complex [125]. Studies focusing on the function of OTUs in osteoclast differentiation and function will reveal more details about the second largest DUB family.

 3.4. JAB1/MPN+/MOV34 (JAMM) and the Bone

There are eight JAMM domain proteins in human, including PRPF8 without protease activity [51, 54]. All of JAMM DUBs are found with subunit complexes of proteasome, such as the proteasome 19S lid complex (POH1/hRpn11) and the COP9 signalosome (CSN5/Jab1) [54]. As an endopeptidase, RPN11 functions to cleave polyubiquitin chains from substrates [126] While CSN5/Jab1 hydrolyzes the ubiquitin-like modifier Nedd8 [127], POH1 enhances osteoclast differentiation and RANKL signaling via regulating Mitf, an important regulator of osteoclast differentiation which required gene expression [128]. MYSM1, a member of the JAMM family, is a histone DUB which specifically deubiquitinates histone 2A [129]. MYSM1 deficiency leads to decreased bone mass. MYSM1 deficiency results in impaired osteogenic differentiation of both mouse MSCs and MC3T3-E1 cell [75]. Recently, study demonstrates that MYSM1 deficiency impairs the potential for primary osteoblasts to differentiate into mature osteoblasts. Meanwhile, MYSM1 knockout reduces the proliferation of osteoclast progenitor and the osteoclast resorption activity [130]. With further studies that might uncover the detailed mechanisms of MYSM1 regulating osteoblast and osteoclast differentiation, this DUB may be a potential therapeutic target for related bone diseases.

The last member of DUBs is Josephin.
There are four proteins belonging to this family, including Ataxin-3, Ataxin-3L, Josephin-1, and Josephin-2 [54]. Unfortunately, current studies have not reported the relationship between Josephin DUBs and skeleton cell differentiation and function. Further studies about the members of Josephin may find novel mechanisms by which these DUBs regulate osteoblast and osteoclast functions.

söndag 30 juni 2019

MZ-xTC scaffold , luuta parantavaa biomateriaalia kehitteillä , Magnesium-sinkki-tetrasykliini-perusteinen ehdokas

https://www.ncbi.nlm.nih.gov/pubmed/31147024

2019 Sep;102:53-65. doi: 10.1016/j.msec.2019.04.010. Epub 2019 Apr 5.

Magnesium-zinc scaffold loaded with tetracycline for tissue engineering application: In vitro cell biology and antibacterial activity assessment.

Abstract

Recently, porous magnesium and its alloys are receiving great consideration as biocompatible and biodegradable scaffolds for bone tissue engineering application. However, they presented poor antibacterial performance and corrosion resistance which limited their clinical applications. In this study, Mg-Zn (MZ) scaffold containing different concentrations of tetracycline (MZ-xTC, x = 1, 5 and 10%) were fabricated by space holder technique to meet the desirable antibacterial activity and corrosion resistance properties. The MZ-TC contains total porosity of 63-65% with pore sizes in the range of 600-800 μm in order to accommodate bone cells. The MZ scaffold presented higher compressive strength and corrosion resistance compared to pure Mg scaffold. However, tetracycline incorporation has less significant effect on the mechanical and corrosion properties of the scaffolds. Moreover, MZ-xTC scaffolds drug release profiles show an initial immediate release which is followed by more stable release patterns. The bioactivity test reveals that the MZ-xTC scaffolds are capable of developing the formation of HA layers in simulated body fluid (SBF). Next, Staphylococcus aureus and Escherichia coli bacteria were utilized to assess the antimicrobial activity of the MZ-xTC scaffolds. The findings indicate that those scaffolds that incorporate a high level concentration of tetracycline are tougher against bacterial organization than MZ scaffolds. However, the MTT assay demonstrates that the MZ scaffolds containing 1 to 5% tetracycline are more effective to sustain cell viability, whereas MZ-10TC shows some toxicity. The alkaline phosphatase (ALP) activity of the MZ-(1-5)TC was considerably higher than that of MZ-10TC on the 3 and 7 days, implying higher osteoblastic differentiation. All the findings suggest that the MZ-xTC scaffolds containing 1 to 5% tetracycline is a promising candidate for bone tissue healing due to excellent antibacterial activity and biocompatibility.

KEYWORDS:

Antibacterial activity; Bioactivity; Biocompatibility; Drug delivery; Mg composite scaffold

onsdag 19 juni 2019

Luuytimen glykaatiosta (haku )

Search results
Items: 1 to 20 of 134
1.
Davis HM, Valdez S, Gomez L, Malicky P, White FA, Subler MA, Windle JJ, Bidwell JP, Bruzzaniti A, Plotkin LI.
J Cell Biochem. 2019 May 20. doi: 10.1002/jcb.28932. [Epub ahead of print]
2.
Stamatopoulos A, Stamatopoulos T, Gamie Z, Kenanidis E, Ribeiro RDC, Rankin KS, Gerrand C, Dalgarno K, Tsiridis E.
J Bone Oncol. 2019 Mar 19;16:100231. doi: 10.1016/j.jbo.2019.100231. eCollection 2019 Jun. Review.
3.
Alamri BN, Bahabri A, Aldereihim AA, Alabduljabbar M, Alsubaie MM, Alnaqeb D, Almogbel E, Metias NS, Alotaibi OA, Al-Rubeaan K.
Eur Rev Med Pharmacol Sci. 2019 Mar;23(5):2139-2150. doi: 10.26355/eurrev_201903_17259.
4.
Tamada K, Nakajima S, Ogawa N, Inada M, Shibasaki H, Sato A, Takasawa R, Yoshimori A, Suzuki Y, Watanabe N, Oyama T, Abe H, Inoue S, Abe T, Yokomizo T, Tanuma S.
Biochem Biophys Res Commun. 2019 Apr 9;511(3):665-670. doi: 10.1016/j.bbrc.2019.01.136. Epub 2019 Feb 27.
PMID:
30826057
5.
Jin H, Zhang Z, Wang C, Tang Q, Wang J, Bai X, Wang Q, Nisar M, Tian N, Wang Q, Mao C, Zhang X, Wang X.
Exp Mol Med. 2018 Nov 21;50(11):154. doi: 10.1038/s12276-018-0177-z.
6.
Xiu G, Xiong W, Yin Y, Chen X, Liu P, Sun J, Ling B.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2018 Sep;30(9):830-835. doi: 10.3760/cma.j.issn.2095-4352.2018.09.003. Chinese.
7.
Wang Y, Ma WQ, Zhu Y, Han XQ, Liu N.
Front Endocrinol (Lausanne). 2018 Sep 21;9:524. doi: 10.3389/fendo.2018.00524. eCollection 2018.
8.
Grosjean F, Yubero-Serrano EM, Zheng F, Esposito V, Swamy S, Elliot SJ, Cai W, Vlassara H, Salem F, Striker GE.
PLoS One. 2018 Sep 25;13(9):e0204366. doi: 10.1371/journal.pone.0204366. eCollection 2018.
9.
Kubota K, Nakano M, Kobayashi E, Mizue Y, Chikenji T, Otani M, Nagaishi K, Fujimiya M.
PLoS One. 2018 Sep 21;13(9):e0204252. doi: 10.1371/journal.pone.0204252. eCollection 2018.
10.
Cortet B, Lucas S, Legroux-Gerot I, Penel G, Chauveau C, Paccou J.
Joint Bone Spine. 2019 May;86(3):315-320. doi: 10.1016/j.jbspin.2018.08.002. Epub 2018 Aug 8.
Both type 1 and type 2 diabetes mellitus are associated with bone disorders, albeit via different mechanisms. Early studies in patients with type 1 diabetes suggested a 10-fold increase in the hip fracture risk compared to non-diabetic controls. Meta-analyses published more recently indicate a somewhat smaller risk increase, with odds ratios of 6 to 7. Diminished bone mineral density is among the contributors to the increased fracture risk. Both types of diabetes are associated with decreased bone strength related to low bone turnover. The multiple and interconnected pathophysiological mechanisms underlying the bone disorders seen in type 1 diabetes include insulin deficiency, accumulation of advanced glycation end (AGE) products, bone microarchitecture alterations, changes in bone marrow fat content, low-grade inflammation, and osteocyte dysfunction. The bone alterations are less severe in type 2 diabetes. Odds ratios for hip fractures have ranged across studies from 1.2 to 1.7, and bone mineral density is higher than in non-diabetic controls. The odds ratio is about 1.2 for all bone fragility fractures combined. The pathophysiological mechanisms are complex, particularly as obesity is very common in patients with type 2 diabetes and is itself associated with an increased risk of fractures at specific sites (humerus, tibia, and ankle). The main mechanisms underlying the bone fragility are an increase in the risk of falls, sarcopenia, disorders of carbohydrate metabolism, vitamin D deficiency, and alterations in cortical bone microarchitecture and bone matrix. The medications used to treat both types of diabetes do not seem to play a major role. Nevertheless, thiazolidinediones and, to a lesser extent, sodium-glucose cotransporter inhibitors may have adverse effects on bone, whereas metformin may have beneficial effects. For the most part, the standard management of bone fragility applies to patients with diabetes. However, emphasis should be placed on preventing falls, which are particularly common in this population. Finally, there is some evidence to suggest that anti-fracture treatments are similarly effective in patients with and without diabetes.

11.
Kwiatkowski J, Halupczok-Żyła J, Bolanowski M, Kuliszkiewicz-Janus M.
Adv Clin Exp Med. 2018 Oct;27(10):1447-1452. doi: 10.17219/acem/71054. Review.
Diabetes mellitus (DM), a growing health problem itself, is accompanied by an increased risk of cardiovascular and thrombotic complications. The imbalance between coagulation and fibrinolysis processes observed in patients with diabetes may be defined as diabetic thrombophilia. Several mechanisms are involved in the hypercoagulability state in diabetics, including endothelial cell damage, altered platelet structure and function, increased microparticle formation, different structure of fibrin clots, disturbances in the activity of coagulation factors, fluctuations in the concentrations of fibrinolysis activators and inhibitors, and qualitative changes of proteins due to glycation and oxidation processes. These all are the reasons why DM is the most common cause of acquired thrombophilia. Moreover, diabetes changes the efficacy of certain medications. Results of various trials seem to suggest that thrombolytic drugs are less effective in patients suffering from this disease. The impact of DM on the effectiveness of treatment with acetylsalicylic acid (ASA) remains unclear. Awareness of thrombotic complications in diabetic patients may enable earlier diagnosis and proper therapy.Free Article
12.
Perusko M, van Roest M, Stanic-Vucinic D, Simons PJ, Pieters RHH, Cirkovic Velickovic T, Smit JJ.
Mol Nutr Food Res. 2018 Sep;62(17):e1800341. doi: 10.1002/mnfr.201800341. Epub 2018 Jul 29.
13.
Kovačić M, Mitrović-Ajtić O, Beleslin-Čokić B, Djikić D, Subotički T, Diklić M, Leković D, Gotić M, Mossuz P, Čokić VP.
Cell Oncol (Dordr). 2018 Oct;41(5):541-553. doi: 10.1007/s13402-018-0392-6. Epub 2018 Jun 26.Abstract PURPOSE:
Previously, the family of S100A proteins has been found to be associated with inflammation and myelopoiesis and to be able to induce or support myeloproliferation during chronic inflammation. Here, we studied the inflammatory myeloid-related proteins S100A4, S100A8, S100A9 and S100A12 in myeloproliferative neoplasms (MPNs) in order to assess the involvement of chronic inflammation in the pathogenesis of MPN. METHODS: We analyzed the S100A4, S100A8, S100A9 and S100A12 mRNA and protein levels in the bone marrow and circulation of 140 patients with MPN and 15 healthy controls using Western blotting, microarray-based mRNA expression profiling and ELISA assays, respectively. In addition we performed functional studies on the proliferation-related AKT and ERK1/2 signaling pathways in MPN-derived granulocytes using Western blotting and proteomic analyses. RESULTS: We found that the S100A mRNA levels were increased in MPN patient-derived circulatory CD34+ cells, and that their protein expression levels were also augmented in their granulocytes and bone marrow stroma cells, depending on the JAK2V617F mutation allele burden. We also found that calreticulin (CALR) mutations were related to reduced S100A8 plasma levels in primary myelofibrosis (PMF). The S100A8 plasma levels were found to be increased in MPN, the S100A9 plasma levels in PMF and essential thrombocythemia (ET), and the S100A12 plasma levels in polycythemia vera (PV). These S100A plasma levels showed a positive correlation with the systemic inflammation marker IL-8, as well as with the numbers of leukocytes and thrombocytes, depending on the JAK2V617F mutation status. Additionally, we found that heterodimeric S100A8/9 can inhibit the AKT pathway in MPN-derived granulocytes mediated by the Toll-like receptor 4 (TLR4), depending on the CALR mutation status. Conversely, we found that blocking of the receptor for advanced glycation end products (RAGE) increased the S100A8/9-mediated inhibition of AKT signaling in the MPN-derived granulocytes. Moreover, we found that heterodimeric S100A8/9 generally induced TLR4-mediated ERK1/2 dephosphorylation proportionally to the JAK2V617F mutation allele burden. TLR4/RAGE blocking prevented the S100A8/9-mediated inhibition of ERK1/2 phosphorylation in PV. CONCLUSIONS:
From our data we conclude that the S100A8 and S100A9 granulocyte and plasma levels are increased in MPN patients, along with inflammation markers, depending on their JAK2V617F mutation allele burden. We also found that S100A8/9-mediated inhibition of the proliferation-related AKT and ERK1/2 signaling pathways can be decreased by CALR mutation-dependent TLR4 blocking and increased by RAGE inhibition in MPN.
14.
Jin X, Liu L, Zhang Y, Xiang Y, Yin G, Lu Y, Shi L, Dong J, Shen C.
J Diabetes Res. 2018 Mar 22;2018:2527406. doi: 10.1155/2018/2527406. eCollection 2018.
15.
Nagareddy PR, Noothi SK, Flynn MC, Murphy AJ.
J Endocrinol. 2018 Jul;238(1):R1-R11. doi: 10.1530/JOE-18-0082. Epub 2018 May 2. Review.

16.
Najar M, Fayyad-Kazan M, Raicevic G, Fayyad-Kazan H, Meuleman N, Bron D, Lagneaux L.
Cell J. 2018 Jul;20(2):250-258. doi: 10.22074/cellj.2018.5104. Epub 2018 Mar 18.
17.
Li R, Wang J, Zhu F, Li R, Liu B, Xu W, He G, Cao H, Wang Y, Yang J.
Mol Immunol. 2018 May;97:45-55. doi: 10.1016/j.molimm.2018.02.014. Epub 2018 Mar 19.

18.
Kim JH, Kim KA, Shin YJ, Kim H, Majid A, Bae ON.
J Toxicol Environ Health A. 2018;81(9):266-277. doi: 10.1080/15287394.2018.1440185. Epub 2018 Feb 23.Abstract
Endothelial cells (ECs) maintain the structure and function of blood vessels and are readily exposed to exogenous and endogenous toxic substances in the circulatory system. Bone marrow-derived endothelial progenitor cells (EPCs) circulate in the blood and differentiate to EC, which are known to participate in angiogenesis and regeneration of injured vessels. Dysfunction in EPC contributes to cardiovascular complications in patients with diabetes, but the precise molecular mechanisms underlying diabetic EPC abnormalities are not completely understood.
 The aim of this study was to investigate the mechanisms underlying diabetic EPC dysfunction using methylglyoxal (MG), an endogenous toxic diabetic metabolite. Data demonstrated that MG decreased cell viability and protein expression of vascular endothelial growth factor receptor (VEGFR)-2 associated with functional impairment of tube formation in EPC. The generation of advanced glycation end (AGE) products was increased in EPC following exposure to MG.
Blockage of receptor for AGE (RAGE) by FPS-ZM1, a specific antagonist for RAGE, significantly reversed the decrease of VEGFR-2 protein expression and angiogenic dysfunction in MG-incubated EPC. Taken together, data demonstrated that MG induced angiogenic impairment in EPC via alterations in the AGE/RAGE-VEGFR-2 pathway which may be utilized in the development of potential therapeutic and preventive targets for diabetic vascular complications.
19.
Wang B, Yu J, Wang T, Shen Y, Lin D, Xu X, Wang Y.
Acta Diabetol. 2018 May;55(5):419-427. doi: 10.1007/s00592-018-1109-z. Epub 2018 Feb 8.

20.
Tachibana M.
Yakugaku Zasshi. 2018;138(2):143-148. doi: 10.1248/yakushi.17-00158. Review. Japanese.