已入深夜,您辛苦了!由于当前在线用户较少,发布求助请尽量完整的填写文献信息,科研通机器人24小时在线,伴您度过漫漫科研夜!祝你早点完成任务,早点休息,好梦!

Sweating chloride bullets: understanding the role of calcium in eccrine sweat glands and possible implications for hyperhidrosis

汗水 多汗症 汗腺 出汗 汗腺 皮肤病科 汗腺 医学 化学 内科学 精神科
作者
Thad E. Wilson,Kristen Metzler‐Wilson
出处
期刊:Experimental Dermatology [Wiley]
卷期号:24 (3): 177-178 被引量:17
标识
DOI:10.1111/exd.12595
摘要

Eccrine (atrichial) sweat glands are large, active cutaneous end-organs vital to human thermal and fluid homeostasis, but regional overactivity of these glands can lead to focal hyperhidrosis. Eccrine sweat glands can expulse over 10 nl/min in a single isolated gland and as much as 3.7 l/h systemically in response to thermal and non-thermal stimuli (1, S1, S2). How do these glands produce such copious secretions? Sweating involves a two-stage process of first producing an isotonic primary solution within the bulbous coiled region of the gland and then modifying (primarily via reabsorbing NaCl) this fluid within the ductal portion as it travels to the skin surface. Thus, production of this primary fluid requires careful scrutiny to understand hyperhidrosis. Epithelial transport in these glands appears to be primarily mediated via clear (agranular) cells, which are also the proposed cell type associated with hyperhidrosis 2. In the current understanding of clear cell function, Cl− is transported through the clear cell, Na+ is transported through tight junctions via a transepithelial voltage gradient, and finally water is pulled through aquaporin channels via osmotic forces. Thus, Cl− is a principal player in the formation of the isotonic primary solution. It is, however, another ion, Ca2+, that appears to be the primary regulator within clear cells. The majority of sudorific agonists (e.g. acetylcholine, norepinephrine via α1-adrenergic receptors and ATP) increase cytosolic Ca2+ via efflux from cellular stores and influx from extracellular fluid. Ca2+ release from intracellular stores is derived via IP3-activated Ca2+ and Ca2+-induced Ca2+ release channels. The influx into the clear cells is a bit more complex, likely involving TRPV1, store-operated Ca2+ entry (Orai1 and TRPC1) and L-type voltage-gated Ca2+ channels (3-5, S3). Highlighting the importance of Ca2+ influx, secretions are abated when isolated glands are placed into Ca2+-free bath (S4, S5), and recent in vivo experiments identified right shifts in the cholinergic agonist to sweating relation with both EDTA (used as an interstitial Ca2+ chelator) and verapamil (L-type channel blocker) 4. The regulatory role of cytosolic Ca2+ is to activate many of the ion channels involved in epithelial transport. This is where the study by Ertongur-Fauth and colleagues 6 enters the picture. Anoctamin 1 (ANO1, also termed TMEM16A), is a Ca2+-activated Cl− channel, which is an anion-selective membrane protein that allows passive Cl− flow and is activated by intracellular Ca2+ as well as being voltage-activated at low Ca2+ concentrations (S6). Ertongur-Fauth and colleagues 6 identified 3 TMEM16A splice variants in the NCL-SG3 sweat gland epithelial cell line as well as in hyperhidrotic and control skin. Investigating the function of a novel splice variant, TMEM16A(acΔe3), authors found it lacks the dimerization domain but still forms a functional channel. Previous studies indicated that TMEM16A subunits appeared to dimerize before reaching the plasma membrane and that dimerization was required for functionality 7. Overexpression of TMEM16A(acΔe3) increased basal Cl− transport even without the dimerization domain 6. This may indicate that the novel TMEM16A splice variant is more constitutively active and thus could possibly produce primary solution during non-stimulated conditions. This, in theory, could lead to increases in production of primary fluid. Alternately, it is possible that sudorific modulators such as galanin and CGRP 8-10, which appear to accentuate sweat formation once transport is initiated, may now be more functionally important due to this increased basal Cl− secretion. Overexpression of the novel TMEM16A splice variant had another interesting result: unlike in basal conditions, expressing either the novel TMEM16A(acΔe3) or the canonical TMEM16A(ac) splice variant enhanced Ca2+-induced Cl− secretion during activated conditions, as compared to the parental cells containing only endogenously expressed TMEM16A splice variants 6. Overexpression of TMEM16A(acΔe3), however, decreased Ca2+-dependent Cl− transport compared to the canonical variant 6. This seems to suggest that a certain mix of TMEM16A splice variants may enhance Ca2+-regulated transport and that the novel variant may be less regulated and less responsive to activators than other splice variants. Thus, overexpression of either splice variant in disease states, such as hyperhidrosis, could yield greater sweating per agonist activation of Ca2+. TMEM16A Cl− channels are not the only channels involved in the formation of isotonic primary solution. Another Ca2+-activated Cl− channel, bestrophin 2 (Best2), is expressed in basolateral and apical membranes of sweat gland secretory cells (S7) and among other functions regulates L-type voltage-gated Ca2+ channels (S8). Clear cells are arranged with a basolateral membrane that contains Na+-K+ ATPase, Na+-Cl−-K+ cotransporters (NCCK1), Ca2+-activated K+ channels, Best2, aquaporin-5 channels (AQP5) and various channels that allow for the Ca2+ influx discussed above. The apical membrane contains TMEM16A and Best2 Ca2+-activated Cl− channels, Ca2+-activated K+ channels, vacuolar H+-ATPase and AQP5. A conceptual model of the role of Ca2+ in activating basolateral Cl− transport, primarily via NCCK1, and then apical Cl− transport, via TMEM16A and Best2, is depicted in Fig. 1. Both known and proposed transporters in clear cells are included in the model to show not only ion transport but also the potential interactions, regulatory points and redundancies in the system. The NCL-SG3 cell line (S9) has dramatically increased our understanding of sweating, but there are some inherent limitations in the use of the cell line. In addition to not knowing precisely which eccrine sweat gland cells are included in the line, cultured cells cannot interact with surrounding cells of other types in cell–cell interactions as would occur in intact human skin. Cultured cells also do not have the neural influences that drive responses in vivo. This specifically relates to agonist-induced sweating in which the traditional neural agonists such as acetylcholine do not cause secretion (S9, S10). Thus, the normal IP3 response to release Ca2+ from intracellular stores cannot occur. While in vitro work such as that described by Ertongur-Fauth and colleagues 6 tells an important story, there is also a need for in vivo studies to determine whether agonist-induced Cl− secretion is similar in intact human skin. It would have been nice for the purpose of telling this story if the novel TMEM16A splice variant would have only been expressed in hyperhidrotic skin and if overexpression would have yielded extremely high agonist-induced Cl− secretion, but, alas, it appears more complex. It is interesting that a combination of isoforms may cause greater function (more Cl− transport and therefore more sweating) than any one isoform. Speculations on the data from Ertongur-Fauth and colleagues 6 could include TMEM16A splice variant differences leading to different rates of sweating and perhaps even differences between hyperhidrotic and normal skin. One of the most effective hyperhidrosis treatments consists of blocking neural activation of sweat glands through cholinergic presynaptic release inhibitors like botulinum toxin, but perhaps the root cause of focal hyperhidrosis is not only at the neural activation level but also includes downstream components such as at the Ca2+-activated Cl− or other Ca2+-activated ion channel level. Both authors contributed to all aspects of background research, interpretation and manuscript preparation. The authors have declared no conflict of interests. Data S1-S10: Supplemental References. Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
4秒前
humengxiao完成签到 ,获得积分10
6秒前
Jasper应助无限桐采纳,获得10
6秒前
zzy发布了新的文献求助10
7秒前
脑洞疼应助橘涂采纳,获得10
8秒前
10秒前
weiyi完成签到,获得积分10
12秒前
清新的音响完成签到 ,获得积分10
14秒前
简墨发布了新的文献求助10
16秒前
17秒前
橘涂发布了新的文献求助10
23秒前
nadia完成签到,获得积分10
26秒前
34秒前
oleskarabach发布了新的文献求助10
37秒前
打打应助杨111采纳,获得10
37秒前
海洋完成签到,获得积分10
37秒前
小二郎应助独特的尔风采纳,获得10
38秒前
39秒前
蟹蟹发布了新的文献求助10
40秒前
41秒前
大个应助zzy采纳,获得10
42秒前
43秒前
43秒前
科研通AI2S应助科研通管家采纳,获得10
43秒前
深情安青应助科研通管家采纳,获得20
43秒前
英姑应助科研通管家采纳,获得10
44秒前
44秒前
qaw完成签到,获得积分10
44秒前
45秒前
默认用户名完成签到 ,获得积分10
46秒前
无花果应助欧皇采纳,获得30
47秒前
48秒前
49秒前
53秒前
hhmm00发布了新的文献求助10
54秒前
科研通AI2S应助123采纳,获得10
54秒前
55秒前
Darren发布了新的文献求助10
1分钟前
蟹蟹完成签到,获得积分20
1分钟前
1分钟前
高分求助中
Exploring Mitochondrial Autophagy Dysregulation in Osteosarcoma: Its Implications for Prognosis and Targeted Therapy 4000
Impact of Mitophagy-Related Genes on the Diagnosis and Development of Esophageal Squamous Cell Carcinoma via Single-Cell RNA-seq Analysis and Machine Learning Algorithms 2000
Migration and Wellbeing: Towards a More Inclusive World 1200
How to Create Beauty: De Lairesse on the Theory and Practice of Making Art 1000
Evolution 1000
Gerard de Lairesse : an artist between stage and studio 670
On the Refined Urban Stormwater Modeling 500
热门求助领域 (近24小时)
化学 医学 材料科学 生物 工程类 有机化学 生物化学 物理 内科学 纳米技术 计算机科学 化学工程 复合材料 基因 遗传学 物理化学 催化作用 免疫学 细胞生物学 电极
热门帖子
关注 科研通微信公众号,转发送积分 2970052
求助须知:如何正确求助?哪些是违规求助? 2632487
关于积分的说明 7091602
捐赠科研通 2265747
什么是DOI,文献DOI怎么找? 1201471
版权声明 591470
科研通“疑难数据库(出版商)”最低求助积分说明 587594