+ All Categories
Home > Documents > New insights in parathyroid hormone secretion

New insights in parathyroid hormone secretion

Date post: 14-Apr-2022
Category:
Upload: others
View: 7 times
Download: 0 times
Share this document with a friend
63
1 Department of Molecular Medicine and Surgery Karolinska Institutet, Stockholm, Sweden New insights in parathyroid hormone secretion - with focus on exocytosis and calcium signaling Ming Lu Stockholm 2010
Transcript
Page 1: New insights in parathyroid hormone secretion

1

Department of Molecular Medicine and Surgery

Karolinska Institutet, Stockholm, Sweden

New insights in parathyroid

hormone secretion

- with focus on exocytosis and calcium signaling

Ming Lu

Stockholm 2010

Page 2: New insights in parathyroid hormone secretion

2

Supervisors

Robert Bränström, Associate Professor

Department of Molecular Medicine and Surgery

Karolinska Institutet, Stockholm, Sweden

Lars-Ove Farnebo, Professor

Department of Molecular Medicine and Surgery

Karolinska Institutet, Stockholm, Sweden

Lars Forsberg, Ph.D

Department of Molecular Medicine and Surgery

Karolinska Institutet, Stockholm, Sweden

Faculty opponent

Peter Stålberg, Associate Professor

Department of Surgical Sciences

Uppsala University, Uppsala, Sweden

Examination board

Bo Rydqvist, Professor

Department of Physiology and Pharmacology

Karolinska Institutet, Stockholm, Sweden

Olle Kämpe, Professor

Department of Medical Sciences

Uppsala University, Uppsala, Sweden

Per Uhlén, Associate Professor

Department of Medical Biochemistry and Biophysics

Karolinska Institutet, Stockholm, Sweden

All previously published papers were reproduced with permission from the publisher.

Published by Karolinska Institutet. Printed by Repro Print AB 2010.

Copyright © Ming Lu, 2010

ISBN 978-91-7409-881-5

Page 3: New insights in parathyroid hormone secretion

3

世上唯一不能倒退的是时间,别在意过去,抓住现在,规划未来 No one can turn back the clock, so don’t regret the past, hold the presence and look forward To My Family

- 送给爱我的家人

Page 4: New insights in parathyroid hormone secretion

4

Thesis defense: Wednesday May 19, 2010 at 9 AM Location: L8:00, CMM, Karolinska University Hospital, Stockholm, Sweden

Page 5: New insights in parathyroid hormone secretion

5

Abstract

The parathyroid is distinguished from other endocrine systems since its primary stimuli

inhibit hormone secretion. It is generally considered that extracellular Ca2+

binds to a

membrane receptor (calcium sensing receptor, or CaSR), and via second messengers

modulate parathyroid hormone (PTH) secretion. However, the intracellular signaling

pathway is not completely characterized, but intracellular Ca2+

concentration (Ca2+

i) is

set aside as a centre mediator for this regulation.

In regulated hormone secretion, or exocytosis, synaptosomal-associated proteins

(i.e. SNAPs and others) are one of the key-players in hormone vesicle and membrane

fusion. These proteins also refered to as SNARE-proteins are widly expressed in

neurons and neuroendocrine cells. In paper I, immunohistochemistry and Western blot

were used to investigate expression of SNARE-proteins, i.e. SNAP-25, SNAP-23,

Syntaxin1 and VAMP in normal and pathological human parathyroid tissues. SNAP-25

and Syntaxin1 were absent in normal parathyroid, but expressed in 20% of chief cell

adenoma and 45% of parathyroid carcinomas. SNAP-23 and VAMP were expressed in

all parathyroid samples, indicating that SNAP-23 and VAMP, rather than SNAP-25

and Syntaxin1, play a central role in exocytosis in human parathyroid cell.

Ca2+

i has a pivotal role in the stimulation-secretion coupling in parathyroid cell,

and study II and III focuse on this process. Increased levels of extracellular Ca2+

triggers Ca2+

influx in parathyroid cells, but the mechanism for this influx is not

completely understood. Using patch-clamp technique and Ca2+

i measurements (Fura-2),

we show the presence of store operated calcium current in human parathyroid and that

this is constituted by TRPC1, STIM1 and Orai1 ternary complex (Paper II).

In addition, calmodulin and calmodulin dependent protein kinase II (CaMKII),

two important proteins in Ca2+

i handling, are demonstrated in human parathyroid cells.

Blocking of calmodulin and CaMKII regulated activity results in an increase of PTH

secretion that was disassociated from Ca2+

i. Our findings suggest that calmodulin and

CaMKII are involved in PTH secretion (Paper III). Interesting from a clinical

perspective is that a negative correlation was seen between serum calcium and

phosphorylated CaMKII protein level.

Finaly, in paper IV, we show that at least two types of Ca2+

-activated K+

channels, i.e. KSK and KBK channel, are expressed in the parathyroid cell. These

channels are sensitive to specific peptide toxin blockers (apamin, charybdotoxin) and

activator (NS-11021). Modulation of these channels affects the membrane potential,

and hence the PTH secretion.

In summary, the stimulation-secretion coupling in the parathyroid cell is complex and

in some aspects unique. This thesis characterizes different parts of this process, from

Ca2+

i handling and influx, ion channel events to exocytosis. In vivo, all parts may

influcence each other and regulate PTH secretion.

Page 6: New insights in parathyroid hormone secretion

6

List of Publications

This thesis is base on the following papers, which are referred to by their Roman

numbers in the text:

I. Lu M, Forsberg L, Höög A, Juhlin CC, Vukojević V, Larsson C,

Conigrave AD, Delbridge LW, Gill A, Bark C, Farnebo LO, Bränström R.

Heterogeneous expression of SNARE proteins SNAP-23, SNAP-25,

Syntaxin1 and VAMP in human parathyroid tissue.

Molecular and Cellular Endocrinology 2008:287, 72-80.

II. Lu M, Bränström R, Berglund E, Höög A, Björklund P, Westin G, Larsson

C, Farnebo LO, Forsberg L.

Expression and association of TRPC subtypes with Orai1 and STIM1 in

human parathyroid.

Journal of Molecular Endocrinology 2010: 44, 285-294

III. Lu M, Berglund E, Larsson C, Höög A, Farnebo LO, Bränström R.

Involvement of Calmodulin and Calmodulin dependent protein kinase II

(CaMKII) in human parathyroid hormone secretion.

Submitted.

IV. Lu M, Forsberg L, Mun HC, Vukojevic V, Berggren PO, Conigrave AD,

Larsson C, Farnebo LO, Bränström R.

Expression of Ca2+

-activated K+ channels in human parathyroid cell:

potential role in hormone secretion.

Manuscript.

Page 7: New insights in parathyroid hormone secretion

7

Related Publication

Zhang F, Dey D, Bränström R, Forsberg L, Lu M, Zhang QM, Sjöholm A.

BLX-1002, a novel thiazolidinedione with no PPAR affinity, stimulates AMP-activated

protein kinase activity, raises cytosolic Ca2+

, and enhances glucose-stimulated insulin

secretion in a PI3K-dependent manner.

American Journal of Physiology: Cell Physiology 2009:296, C346-354.

Page 8: New insights in parathyroid hormone secretion

8

Table of content

Introduction ............................................................................................................... 11

Anatomy and histology of parathyroid gland .................................................... 12

Biosynthesis and metabolism of parathyroid hormone (PTH) ........................... 13

Biological effects of PTH ................................................................................. 14

Diseases in parathyroid glands ......................................................................... 16

Primary hyperparathyroidism

Secondary hyperparathyroidism

Hypoparathyroidism

Calcium signaling ....................................................................................................... 17

Ca2+

mobilization and Ca2+

influx .................................................................... 17

Ca2+

binding proteins ....................................................................................... 19

Hormone secretion – exocytosis ................................................................................. 20

Regulation of PTH secretion ..................................................................................... 21

Calcium sensing receptor (CaSR)..................................................................... 21

Vitamin D ........................................................................................................ 23

Phosphate ........................................................................................................ 24

Fibroblast growth factor 23 and α-klotho ......................................................... 24

Other regulators, like divalent cations, Lithium, ATP, estrogen, and prolactin .. 26

Aim of study ............................................................................................................... 28

Materials and methods............................................................................................... 29

Human parathyroid specimens ......................................................................... 29

Immunohistochemistry .................................................................................... 29

Western blot .................................................................................................... 30

Protein complex immunoprecipitation (Co-IP) ................................................ 31

Reverse transcription (RT) polymerase chain reaction (PCR) and

quantitative real-time PCR (qRT-PCR) ............................................................ 31

Human parathyroid cell preparation ................................................................. 33

Hormone secretion ........................................................................................... 34

Intracellular Ca2+

measurements with Fura-2 ................................................... 34

Electrophysiology technique ............................................................................ 36

Page 9: New insights in parathyroid hormone secretion

9

Results and discussion ............................................................................................... 38

Paper I - Heterogeneous expression of SNARE proteins SNAP-23,

SNAP-25, Syntaxin1 and VAMP in human parathyroid tissue ......................... 38

Paper II - Expression and association of TRPC subtypes with Orai1 and

STIM1 in human parathyroid ........................................................................... 40

Paper III - Involvement of calmodulin and calmodulin dependent kinase II

(CaMKII) in parathyroid hormone secretion .................................................... 41

Paper IV - Expression of Ca2+

-activated K+ channels in human parathyroid cell:

potential role in hormone secretion ................................................................... 42

Conclusion ................................................................................................................. 44

摘要............................................................................................................................ 46

Acknowledgements .................................................................................................... 47

References .................................................................................................................. 51

Page 10: New insights in parathyroid hormone secretion

10

Abbreviations

ATP Adenosine-5'-triphosphate

Ca2+

e Extracellular Ca2+

concentration

Ca2+

i Intracellular Ca2+

concentration

CaMKII Calmodulin-dependent protein kinases II

cAMP Cyclic adenosine monophosphate

CaSR Calcium sensing receptor

Co-IP Protein complex immunoprecipitation

ER Endoplasmic reticulum

FGF23 Fibroblast growth factor 23

IP3 Inositol-1,4,5- trisphosphate

KBK channel Big conductance Ca2+

activated K+ channels

KSK channel Small conductance Ca2+

activated K+ channels

Orai1 Calcium release-activated calcium channel protein 1

pHPT Primary hyperparathyroidism

PKA, PKC Protein kinase A, and C

PLA2, PLC, PLD Phospholipase A2, C and D

PTH Parathyroid hormone

qRT-PCR Quantitative real-time polymerase chain reaction

RT-PCR Reverse transcription polymerase chain reaction

RYRs Ryanodine receptors

SERCA Sarco (endo) plasmic reticulum Ca2+

-ATPase

sHPT Secondary hyperparathyroidism

SNAP-23/25 Synaptosomal-associated protein 23/25

SNARE Soluble NSF Attachment Protein receptors

SOCS Store operated calcium channels

STIM1 Stromal interaction molecule 1

TRP channel Transient receptor potential channel

VAMP2 Vesicle-associated membrane protein 2 (Synaptobrevin 2)

VOCCs Voltage operated calcium channels

Page 11: New insights in parathyroid hormone secretion

11

Introduction

There is a tiny endocrine organ in the neck called parathyroid gland. It produces

parathyroid hormone (PTH), an important hormone for calcium regulation in our body.

Loss or gain of function in this organ leads to multiple clinical features. However, the

parathyroid gland has been unobserved until middle of 18th century. Richard Owen, a

conservator of Hunterian Museum at the Royal College of Surgeons of England, gave

the first description of parathyroid gland in 1852. By discovery in Indian rhinoceros, he

reported ‘a small compact yellow glandular body was attached to the thyroid at the

point where the veins emerge’ (Rhys Evans et al. 2004). That small gland was named

as parathyroid by Ivar V. Sandström, a postgraduate student in Uppsala, Sweden in

1877 (Carney 1996; Eknoyan 1995). It was observed that removal of parathyroid

glands led to ‘tetany’ in both animals and human, such as symptoms of involuntary

systemic muscles contraction known as Chvostek’s sign, Trousseau’s sign and

carpopedal spasm (Pool 1907; Vincent 1904), but the reason was not clear at that

moment. One claim was that parathyroid glands might be responsible for cleaning of

toxic substances, e.g. dimethylguanidine. The idea was that after removal of

parathyroid glands, accumulation of toxicants in the body caused tetany. However, the

scientific proof for this idea was poor. Simultaneously, more and more evidences

showed that tetany was associated with low serum calcium, and that the parathyroid

glands were involved in regulation of serum calcium since both calcium supplement

and injection of parathyroid extracts prevented tetany (Branham 1908; Collip 1925;

Shepardson 1927; Stewart & Percival 1927; Willard 1935).

Besides calcium, phosphate was also observed to be regulated by PTH. It was figured

out lately that not only parathyroid but also kidney and bone were involved the

maintenance of serum calcium and phosphate (Ellsworth 1932). In the late decades of

1900’s, many studies were done to understand the secretion of PTH. It was found that

calcium was the main regulator of PTH secretion. Within a narrow range, PTH is

negatively regulated by calcium. A break trough in the understanding of parathyroid

physiology was the identification and cloning of calcium sensing receptor (CaSR)

(Brown et al. 1993). CaSR are located in cell membrane, facing the calcium sensing

epitope towards the extracellular space whereby sensing small changes in circulating

calcium concentration. CaSR is a G-protein linked receptor, and acts via a

phospholipase C (PLC) pathway including production of inositol-1,4.5-trisphosphate

(IP3) and release of Ca2+

from intracellular stores (Brown 1991). Intracellular Ca2+

Page 12: New insights in parathyroid hormone secretion

12

(Ca2+

i) has been shown to have a pivotal role in regulation of PTH secretion (Shoback

et al. 1984). The central role of CaSR in the stimulus-secretion coupling is well

understood since mutations in CaSR cause changes in PTH secretion. Mutations in

CaSR in the population are rare and can only explain a minority of parathyroid

diseases. However, the finding of CaSR has provided a new target for treatment of

parathyroid diseases (Brown 2007). Another breakthrough in parathyroid gland

physiology was the cloning of PTH/PTHrP (parathyroid hormone related peptide)

receptor by Juppner in 1991 and Schipani in 1993 (Juppner et al. 1991; Schipani et al.

1993). The discovery of PTH receptor on bone and kidney enables us to know more

about the interaction of PTH and its target organs (Potts 2005).

Anatomy and histology of parathyroid gland

Anatomy

Parathyroid glands are usually composed of four glands, two superior glands and two

inferior glands; the location is on the backside of the thyroid gland in the neck (Figure

1). The superior glands arise from the fourth endodermal pharyngeal pouches, and the

inferior glands originate from the third endodermal pharyngeal pouches. The weight of

each gland is on average 40-50 mg. The location and the number of the glands may

vary in normal population. For example, they can be located down in the thymus or be

five to eight glands in a normal person. Their color is yellow-brown. In theory, they are

easily distinguished from the reddish structure of thyroid; nevertheless, they are

sometimes hard to recognize from thyroid and fat in reality. It is high demand for the

experience of the surgeon for identification (Gardner & Shoback 2007).

Figure 1. Location of parathyroid glands.

Page 13: New insights in parathyroid hormone secretion

13

Histology

Histologically, normal parathyroid glands are composed of epithelial cells and

stromal fat. Stromal fat is around 30% at age of 25 and decreases with aging. The

epithelial cells include chief cells and oxyphilic cells, and majority is chief cells. The

chief cells are small (8 µm), light, centrally round nucleus, and contain intracellular

fat. Some of them have extremely clear cytoplasm and well-defined cell membrane

due to abundant cytosolic glycogen, namely water clear cells. Chief cells are PTH

producing cells in parathyroid glands. Oxyphilic cells are usually slightly larger (12

µm) with acidophilic cytoplasm due to large amounts of mitochondria, which is

distinguished from smaller and weakly acidophilic cytoplasm of chief cells. Their

numbers increase with age. The number of oxyphilic cells is much less than chief

cells in normal parathyroid and their function are still not very clear. However,

oxyphilic cells was found to release PTH in sHPT patients (Rudberg et al. 1986;

Tanaka et al. 1996) and PTH producing oxyphilic cell adenomas are reported from

time to time (Allen & Thorburn 1981; Bedetti et al. 1984; Zhou et al. 2003), therefore,

it is also considered as functional cells.

Figure 2. Transmission Electron Microscopy image of normal parathyroid cell

(1: oxyphilic cell, 2: chief cell)

.

Biosynthesis and metabolism of PTH

Mature PTH is an 84-amino-acid long peptide. Its gene is located on chromosome 11.

The same as other hormone, PTH is initially synthesized as prepro-PTH with 115

Page 14: New insights in parathyroid hormone secretion

14

amino acids in the rough endoplasm reticulum. After the first cleavage removing 25-

amino-acid on the N-terminal, it becomes pro-PTH. The leader sequence of pro-PTH

serves as a signal transporting pro-PTH to Golgi apparatus. Then a second cleavage

with six amino acids on the N-terminal results in the formation of mature PTH. After

mature PTH leaves the Golgi apparatus, it is stored in secretory-vesicles and waiting

for secretion signal (Habener 1981).

Full length PTH (1-84) has a half-life of 2-4 minutes in blood. After released from

parathyroid glands into blood circulation, it is further cleaved in liver and kidney at

the 33–34 and 36–37 positions to produce an amino terminal fragment and a carboxyl

terminal fragment. The amino terminal fragment (1-34) is the biologically active part

of PTH. It binds to PTH receptor and activates cell signaling in target tissues. The

active fragments only constitute small part of circulating PTH and soon are degraded,

whereas the carboxyl terminal fragments consist of majority of circulating PTH with

longer life. They are mainly cleared by kidney, so they accumulate in renal failure.

Instead of acting with PTH receptor, they have their own receptors but the

physiological importance is not completely understood.

The first immunoassay of PTH was invented by Berson and Yalow in 1963 (Berson

et al. 1963). After decade’s development, the measurement of intact PTH has become

precise and fast. Current assays compose of two-site immunoradiometric assay or

immunochemiluminescent assay techniques, by which the normal range for PTH is

approximately 10–60 ng/L (1–6 pmol/L).

Biological effects of PTH

Calcium is a key intracellular messenger and co-factor of various enzymes. It plays

diverse roles in multiple organs. It modulates neuromuscular excitability,

proliferation, apoptosis, exocytosis and so on. Calcium levels vary greatly between

extracellular and different cellular compartments in the human body; ranging from

1.15-1.35 mmol/L in blood (reference value at Karolinska University Hospital).

Maintenance of serum calcium level and calcium gradient in cells are very important

for the physiological and pathophysiological functions of our body. PTH and Vitamin

D are the main regulators of blood calcium level.

Page 15: New insights in parathyroid hormone secretion

15

When serum calcium is low, it gives a positive signal to parathyroid gland resulting in

an increase of PTH secretion. The release of PTH causes mobilization of calcium

from bone and calcium absorption from kidney and intestine. The increased blood

calcium in turn gives a negative feedback to parathyroid gland and inhibits PTH

secretion. This response feedback system controls serum calcium within a narrow

range (Figure 3).

An increase of PTH affects renal tubular cells within minutes. It facilitates calcium

influx into distal renal tubules by transporting and activating dihydropyrine-sensitive

Ca2+

channel in apical plasma membrane (Bacskai & Friedman 1990; Lau &

Bourdeau 1995). It promotes phosphate excretion by decreasing type II Na+/Pi co-

transporter activity and protein content at the apical brush-border membrane of

proximal tubules (Kempson et al. 1995; Murer et al. 1996).

It was originally believed that the effect of PTH on intestinal calcium absorption was

only indirect. PTH stimulates the biosynthesis of 1α-hydroxylase in kidney resulting

in increased production of active Vitamin D metabolite 1,25(OH)2D3 (1,25-

dihydroxycholecalciferol) (Henry et al. 1974), which directly enhances both calcium

and phosphate reabsorption through the small intestine (Norman 1979). However,

recent studies found the expression of functional PTH receptor in intestine; and more

evidence indicates that PTH may have a direct effect on calcium transport and uptake

in intestine (Nemere & Larsson 2002).

Bone is also a direct target of PTH. The effect of PTH on bone is dualistic, including

both anabolic and catabolic effect. High level of PTH causes bone loss and

intermittent administration induces bone remodeling. PTH enhances proliferation and

differentiation of osteoblast cells responsible for bone formation. But it can also

stimulate ostoclast cells through activation of Receptor Activator for Nuclear Factor

B Ligand/Receptor Activator for Nuclear Factor B (RANKL/RANK) signaling which

is required for osteoclastogenesis, resulting in increase of calcium mobilization (Qin

et al. 2004). It is the reason why hyperparathyroidism patients develop osteoporosis

and osteitis fibrosa cystica. However, intermittent administration of PTH induces

bone reconstruction and rarely causes bone resorption, therefore it has been approved

for treatment of osteoporosis.

Page 16: New insights in parathyroid hormone secretion

16

The action of PTH on target organ is mediated by type 1 PTH receptor, a G protein

receptor that is highly expressed in kidney and bone. Interaction of PTH and PTH

receptor activates adenylate cyclase/cAMP/protein kinas A (PKA) pathway or

phospholipase C/protein kinase C (PKC) pathway, which in turn regulate down-

stream biological actions (Schluter 1999).

Figure 3. Simplified model of human calcium homeostasis.

Diseases in parathyroid glands

Primary Hyperparathyroidism

Hyperparathyroidism refers to excess PTH secretion. The cause of primary

hyperparathyroidism (pHPT) is parathyroid adenoma (80-90%), hyperplasia (10-20%)

and carcinoma (<1%). The prevalence is about 1 to 4 cases per 1000 of the general

population, but can be much higher in the elderly population above the age of 50

(Nilsson et al. 2002). The sex ratio between women and men is about 3:1 (Heath

1989). The peak is among menopause women. It has been reported the prevalence in

menopause women reaches 3.4% in Sweden (Lundgren et al. 2002; Lundgren et al.

1997; Palmer et al. 1988). It is the most common cause of hypercalcemia in

unselected patients.

Page 17: New insights in parathyroid hormone secretion

17

Overproduction of PTH increases mobilization and absorption of calcium from bone

and increases renal excretion of phosphate, resulting in hypercalcemia and

hypophosphatemia. Clinic features include osteoporosis, peptic ulcer, pancreatitis,

hypertension, anxiety, kidney stone, and so on. However, many patients lack

symptoms and some of them have normal level of calcium.

Secondary hyperparathyroidism

One important cause for secondary hyperparathyroidism (sHPT) is vitamin D

deficiency; it is a common complication of chronic renal failure. Initially, reduction

of Vitamin D and low ionized Ca2+

stimulate PTH synthesis and release. As disease

progresses, Vitamin D receptor and CaSR decrease in parathyroid gland (Kifor et al.

1996), promoting proliferation of parathyroid gland (Tokumoto et al. 2005; Yano et

al. 2000) and resulting in insensitivity of parathyroid to calcium, which further

enhances secretion of PTH. Moreover, high phosphate and uremia can induce

hyperplasia of parathyroid gland independently.

Hypoparathyroidism

Hypoparathyroidism refers to impairment of PTH secretion. It is mostly a

complication to thyroidectomy. Primary hypoparathyroidism is rare and due to

autoimmune destruction of parathyroid gland. It may also be due to activating

mutations in CaSR. Hypocalcemia and hyperphosphatemia are major laboratory

findings, and clinical magnificence is hypocalcemia tetany. Supplements of calcium

and vitamin D are fundamental treatment.

Calcium signaling

Calcium mobilization and calcium influx

Calcium is a critical divalent ion involved in all cellular processes. It regulates

contraction, secretion, apoptosis, proliferation, differentiation, fertilization, and so on.

Normally, extracellular free calcium ion concentration around 1.2 mM, whereas

calcium concentration within the cell (intracellular Ca2+

concentration, or Ca2+

i) can

be as low as 100 nM in resting stage and rise to 1-10 M when cell is excited.

Increased level of Ca2+

i is the result of calcium mobilization from ER and calcium

influx from extracellular spaces. This action involves efforts of several ion channels,

Page 18: New insights in parathyroid hormone secretion

18

exchangers and second messengers and consumes lots of energy (Clapham 1995;

Clapham 2007).

Within calcium signaling, SERCA pump on ER helps to take in Ca2+

and channels

with IP3 receptor or Ryanodine receptors are transporting Ca2+

out to cytosol. IP3

receptors usually are expressed in non-excitable cells and are activated by IP3,

whereas calcium or Cyclic ADP Ribose activates Ryanodine receptors in excitable

cells. Calcium permeable channels in plasma membrane mediate calcium influx,

including voltage-operated calcium channels (VOCCs) and voltage-independent

calcium channel. VOCCs are found in excitable cell and are activated by membrane

depolarization. Voltage-independent calcium channels comprise three channels. 1)

Ligand-gated ionotropic channels are channels operated by ligands (chemical

messenger), e.g. ATP, serotonin, Gamma-aminobutyric acid (GABA). They are

usually non-selective channels for Ca2+

permeation. 2) Transient receptor potential

channel (TRP) is a big non-selective cation channels family composed of three

homologous: TRPC (canonical), TRPV (vanilloid), TRPM (melastatin) and three

distantly related subfamilies (TRPA (ankyrin), namely, TRPP (polycystin) and

TRPML (mucolipin)) in mammals. Most of TRP channels are Ca2+

permeable

channels (Inoue 2005; Inoue et al. 2003; Pedersen et al. 2005). They are widely

expressed in body and are activated by a variety of stimulus. For example, TRPV1

channel is activated by heat and capsaicin, TRPV4 is stimulated by osmolarity and

TRPC channels are regulated by PLC signaling cascades activated by G-protein

coupled receptor or tyrosine-kinase receptor. 3) Store operated calcium channels

(SOCs) are activated by the depletion of intracellular calcium store. This is the

dominant calcium entry pathway in non-excitable cells, such as epithelial cells,

hepatic cells and blood cells. Two well established components of SOCs are STIM1

(Stromal interaction molecule 1) located on ER membrane and Orai1 (Calcium

release-activated calcium channel protein 1) on plasma membrane (Frischauf et al.

2008; Hewavitharana et al. 2007). STIM1 senses decrease of Ca2+

in ER and drives

ER membrane to plasma membrane (Zhang et al. 2005). The binding of STIM and

Orai1 facilitates the opening of Orai1 channels and calcium influx (Soboloff et al.

2006). Recently, TRP channels, especially TRPC1 channel, are also considered as

components of SOC channels. The interaction of TRPC channel, STIM1 and Orai1

has been reported in many cells types (Cheng et al. 2008; Ong et al. 2007).

Thapsigargin, a specific blocker of SERCA, is the most common tool for

Page 19: New insights in parathyroid hormone secretion

19

investigation of SOCs. Moreover, Na+/Ca

2+ exchanger and plasma membrane calcium

ATP pump (PMCA) are pumps for moving out Ca2+

.

Figure 4. Schematic illustration of calcium signaling.

Calcium binding proteins

There are many Ca2+

binding proteins involved in calcium signaling. They may act as

Ca2+

sensors or receptors for signal transduction or may exist as Ca2+

buffers.

Depending of the type of Ca2+

binding domain, they are classified into EF-hand,

annexin and C2 region. EF hand proteins, for example Troponin C modulates muscle

contraction, and calmodulin regulates several enzymes (e.g. CaMKI, II and IV,

phosphodiesterase) involved in most cellular processes, such as exocytosis, muscle

contraction, ion channel activation, and cell proliferation. Calbindin-D9K and D28K

regulate calcium transport in intestine and kidney, and S100 is involved in cell

growth. Annexin proteins modulate phospholipase-A2 (PLA2) inhibition and ion

channel activity. C2 domain proteins, for example, Rab3A regulates secretory

vesicles trafficking, and synaptotagmin regulates exocytosis. Some proteins, such as

calsaquestrin, calreticulin, bind to calcium as storage (Clapham 1995; Niki et al.

1996).

Page 20: New insights in parathyroid hormone secretion

20

Hormone secretion-exocytosis

The process of packing proteins into vesicles and releasing it out to the extracellular

space is called exocytosis. Generally, exocytosis can be categorized into regulated

exocytosis and constitutive exocytosis. Constitutive exocytosis occurs in all cells. It

spontaneously and continuously transports proteins from the Golgi apparatus to the

plasma membrane. However, in neurons, as well as in endocrine cells, proteins are

stored in secretory vesicles and released only in response to stimulation of certain

external signal, namely regulated exocytosis (Alberts et al. 2007). The same as in

other endocrine cells, parathyroid hormone was observed to be stored in large

secretory vesicles waiting for release using electron microscopy by Setoguti and

colleges. The same authors also showed that PTH is rapidly released by exocytosis

when serum calcium was decreased (Setoguti et al. 1981; Setoguti et al. 1988).

Exocytotic process includes vesicle trafficking, tethering, docking, priming and

fusion, which involve several proteins and protein-protein interactions. Among them,

SNARE complex, munc-18 proteins and small GTP binding protein Rab3A are

essential for Ca2+

-regulated exocytosis. SNARE refers to Soluble N-ethylmaleimide-

sensitive factor Attachment protein REceptors. It is a big membrane-bound proteins

family categorized into vesicle-SNARE (v-SNARE) on vesicle membrane and target-

SNARE (t-SNARE) on plasma membrane. The formation of a stable 4-α-helix

SNAREs complex is the core-step driving vesicle fusion. The most well established

SNARE complex consists of t-SNARE proteins SNAP-25 and Syntaxin1A and v-

SNARE protein Synaptobrevin (VAMP). Protein Rab3A transports vesicles to

docking site, and then the disassociation of munc-18 proteins from Syntaxin1A

facilitates the reversible contact of SNARE complex to plasma membrane. Following

by Ca2+

influx, Ca2+

binding to synaptotagmin, a calcium sensor promotes assembly

of SNAREs into 4-α-helix bundles and accelerates SNARE-catalyzed fusion (Chicka

et al. 2008; Gerst 1999; Tang et al. 2006). The components of SNAREs proteins vary

in different cell types.

Page 21: New insights in parathyroid hormone secretion

21

Figure 5. Model of Ca2+

regulated exocytosis.

Regulation of parathyroid hormone secretion

Calcium sensing receptor

Serum calcium is the major physiological regulator of PTH secretion. In contrast to

other endocrine cells, low calcium stimulates PTH secretion, whereas high calcium

inhibits. The calcium-PTH curve is very steep between 1.1 mM to 1.3 mM, indicating

that parathyroid cells are extremely sensitive to the change in Ca2+

e level (Brown

1991). The high Ca2+

sensitivity enables us to control serum calcium within a tight

range. Practically, a calcium-PTH set point (calcium required for half-maximal

inhibition of PTH release) has been used to evaluate the individual Ca2+

sensitivity. It

has been shown cultured normal human parathyroid cells have a set-point close to 1.0

mM, and around 1.1-1.3 mM in vivo (Brown 1991). The set-point is shifted to the

right in hyperparathyroidism which means decreased Ca2+

sensitivity and shifted to

left in hypoparathyroidism indicating increased Ca2+

sensitivity. The Ca2+

sensitivity

varies between individuals. CaSR was discovered as an important factor for the

regulation of Ca2+

sensitivity in parathyroid cells (Brown et al. 1993). CaSR senses

small increase of Ca2+

e resulting in hundreds times rise of Ca2+

i which downstream

leads to inhibition of PTH secretion.

The CaSR is a member of subfamily C of G-protein coupled receptors. The gene is

located on chromosome 3q13.3-2.1. The molecular weight of the protein is 120 kDa,

and is coupled to Gαq-PLCβ complex (Brown et al. 1993). Activation of CaSR

enhances hydrolysis of phosphoinositides resulting in increased generation of IP3 and

diacylglycerol (DAG). IP3 binds to IP3 receptor in endoplasmatic reticulum inducing

Page 22: New insights in parathyroid hormone secretion

22

transient calcium release from intracellular stores, followed by sustained influx of

extracellular calcium. The pathway of calcium influx is still unclear in parathyroid

cells. It has been widely debated if there are voltage gated calcium channels or not.

Although two studies have shown the expression of dihydropyridine-sensitive L-type

calcium channels in parathyroid cells (Chang et al. 2001; Yokoyama et al. 2009), K+-

induced depolarization reduces Ca2+

i (Shoback & Brown 1984b) which do not

support the present of VOCCs. On the contrary, thapsigargin, an inhibitor of SERCA,

induces a rapid increase of Ca2+

i and lowered PTH secretion, which indicate the

existence of store operated calcium entry (Ferzandi & MacGregor 1997; Shoback et

al. 1995).

Calcium is not the only ligand for CaSR. CaSR can also be activated by a number of

divalent or trivalent cations and various amino acids. Increased Ca2+

i due to the

activation of CaSR is strongly correlated to decreased PTH secretion (Shoback et al.

1983). Notable, in endocrine cells as well as in neurons, an increased Ca2+

i generally

promotes hormone secretion. It is unknown why increased Ca2+

i causes an inhibition

of PTH secretion in parathyroid cells. By using the model of permeabilizing cells,

Oetting and his colleagues found that increased Ca2+

i induced PTH secretion after

abolishment the effect of membrane potential, which implied that membrane potential

is a critical regulator for PTH secretion (Oetting et al. 1987). Our group has

previously showed that high Ca2+

e results in hyperpolarization, whereas low Ca2+

e

leads to depolarization in human parathyroid cells, most likely via Ca2+

-activated K+

channels (Valimaki et al. 2003).

Besides PLC, PLA2 and PLD signaling cascades are also activated by CaSR in

parathyroid cells (Kifor et al. 1997). The phosphorylation and activation of PLA2

generates arachidonic acid that inhibits PTH secretion via the 12- and 15-

lipoxygenase pathway (Bourdeau et al. 1992). Activation of CaSR also couples to Giα

inactivating adenylyl cyclase and reducing the production cAMP, thereby decreasing

PTH secretion (Brown et al. 1990; Brown et al. 1978; Shoback & Brown 1984a).

CaSR is known as the most important receptor for calcium regulated PTH secretion.

This statement rests on several findings; 1) Familial Hypocalciuric Hypercalcemia

(FHH), which is characterized by hypercalcemia, lower urinary calcium excretion

(hypocalciuria) and normal or increased PTH levels, is associated with inactivated

mutations of CaSR gene. 2) expression of CaSR was found to be reduced in pHPT

Page 23: New insights in parathyroid hormone secretion

23

and sHPT at both mRNA and protein level (Farnebo et al. 1997; Farnebo et al. 1998).

3) reduction of CaSR is also associated with right-shift of calcium-PTH set point in

hyperparathyroidism (Corbetta et al. 2000; Kaneko et al. 1999). 4) activating

antibody binds to CaSR results in hypoparathyroidism (Goswami et al. 2004). 5)

pharmacological activation of CaSR using calcimimetic agonists (Cinacalcet or R-

568) have dose-dependent effect in reducing PTH secretion and serum calcium, and

have been used for treatment of both primary and secondary hyperparathyroidism

(Lindberg et al. 2005; Peacock et al. 2005; Shoback et al. 2003; Torres 2006). All

this point to the central role of CaSR in the parathyroid physiology. However, it has

been shown that changes of CaSR directly contributes to enhanced or impaired PTH

secretion, but it has not been possible to correlate the level of CaSR expression with

individual Ca2+

sensitivity (Cetani et al. 2000; Corbetta et al. 2000).

Vitamin D

Another important regulator of PTH secretion is the active form of Vitamin D. Vitamin

D in our body comes from skin and food. When skin is exposed to sunlight, 7-

dehydrocholesterol, a derivative of cholesterol, is converted by UVB light to vitamin-

D3. However, vitamin-D3 has low biological activity. In order to be functional, it needs

to be hydroxylated to 25(OH)D3 (25-hydroxycholecalciferol, calcidiol) by the enzyme

25-hydroxylase in the liver, and then further converted into 1,25(OH)2D3 (calcitriol) by

the enzyme 1α-hydroxylase in the kidney. Calcitriol, the most active from of vitamin D,

is critical for maintenance of blood calcium and phosphate via interaction with

intestine, kidney and bone. In case of kidney failure, inefficient 1α-hydroxylase reduces

the production of calcitriol resulting in low blood calcium. The following compensative

increase of PTH secretion is named secondary hyperparathyroidism (sHPT). Besides

regulating blood calcium, calcitriol directly suppresses PTH synthesis and secretion by

binding Vitamin D receptor on the 5’-untranslated region of PTH gene and down-

regulating PTH gene transcription (Brown et al. 1995; Demay et al. 1992). It has been

shown that calcitriol inhibits PTH secretion in cultured parathyroid cells from sHPT

patients in a dose-dependent manner and independent of vitamin D receptor

polymorphisms (Alvarez-Hernandez et al. 2003). Calcitriol also has a similar action on

PTH secretion, but is a hundred times less active than calcitriol (Ritter et al. 2006).

Clinically, analogs are an essential treatment for sHPT, especially in the early stage.

Page 24: New insights in parathyroid hormone secretion

24

Phosphate

Phosphate is a positive regulator of PTH secretion. It has been found that there are three

phosphate binding proteins on 3’-UTR of PTH mRNA: AU-rich binding factor

(AUF1), N-ras(Unr) and KH-splicing regulatory protein (KSRP). High phosphate

enhanced the binding of AUF1 and Unr to PTH mRNA, which increases mRNA

stability. In low serum phosphate, KSRP recruited exosome to PTH mRMA facilitating

PTH mRNA degradation (Levi et al. 2006; Nechama et al. 2008; Sela-Brown et al.

2000). But it is still unclear that how parathyroid cells recognize phosphate. A

complementary DNA encoding Na+-Pi co-transporter, namely PiT-1, was cloned in rat

parathyroid tissue in 1998 (Tatsumi et al. 1998). Low phosphate diet increased

expression of PiT1, whereas high phosphate diet inhibited, and calcitriol also raised the

expression of PiT1, indicating that PiT1 was contributing to both phosphate and

calcitriol regulation in parathyroid. But there is no evidence that phosphate needs to be

transported into cells to exert its function. So PiT1 can be a sensor of phosphate

(Miyamoto et al. 2000; Miyamoto et al. 1999). Phosphate can also indirectly regulates

PTH secretion via production of fibroblast growth factor 23 (FGF23) from bone cells

(see discussed as below).

Fibroblast growth factor 23 and α-Klotho

Fibroblast growth factor 23 (FGF23) is a hormone released from osteblast and

osteocytes cells in bone. It is very critical for phosphate and vitamin D metabolism. It

stimulates urinary phosphate excretion and reduces production of calcitriol by

suppression of 1α-hydroxylase activity in the kidney. Ablation of FGF23 gene causes

hyperphosphatemia, hypercalcemia, high serum calcitriol, and suppressed PTH

(Shimada et al. 2004). Active mutation of FGF23 in human causes autosomal dominant

hypophosphatemic rickets (Shimada et al. 2002). Over-production of FGF23 by tumor

is the main reason for tumor-induced osteomalacia (Shimada et al. 2001). Recently, it

was found that parathyroid gland is also a target of FGF23. FGF23 receptor type 1 and

3 are found in parathyroid gland (Ben-Dov et al. 2007). Serum FGF23 was increased in

pHPT mouse model and the level was significantly correlated with serum PTH and

calcium. The level of FGF23 decreased after parathyroidectomy (Kawata et al. 2007).

But the level of serum FGF23 in HPT patients is still controversial (Kobayashi et al.

2006; Tebben et al. 2004). In vitro study on bovine parathyroid cells has shown that

Page 25: New insights in parathyroid hormone secretion

25

FGF23 reduced PTH mRNA level and inhibited PTH secretion in a dose-dependent

manner (Krajisnik et al. 2007).

Klotho gene was first described in 1997 by Kuro-o and his colleagues. Mutant mice

showed multiple aging-related disorders, including short lifespan, infertility,

arteriosclerosis, osteoporosis, age-related skin changes etc. The name was given

according to Geek mythology in which klotho is a goddess who spins the thread of life

(Kuro-o et al. 1997). Interestingly, hyperphosphatemia, high serum calcitriol and

hypercalcemia were also found in klotho mutant mice, which indicate that klotho is

involved in calcium and phosphate homeostasis. Later on, α-Klotho, an isoform of

Klotho, was found to be important for PTH secretion and calcium reabsorption in

kidney. In response to low Ca2+

e, it recruits Na+/K

+-ATPase from ER to cell surface and

induces PTH secretion in parathyroid glands (Imura et al. 2007). In kidney, recruitment

of α-Klotho activates TRPV5 channels that increases Ca2+

intake from lumen into distal

convoluted tubule cell, then Ca2+

is further absorbed into blood by Na+-Ca

2+ exchanger

(Chang et al. 2005). It has reported that the expression of α-Klotho was reduced in

pHPT patients, and the level of α-Klotho was inversely correlated to serum calcium, i.e.

higher level of serum calcium, lower expression of α-Klotho. But there is no correlation

between PTH level and α-Klotho expression (Bjorklund et al. 2008). Distinguished

from CaSR, α-Klotho/Na+/K

+ATPase pathway may be more important for low Ca

2+

stimulated PTH secretion.

The fact that the α-Klotho deficient mice showed all phenotypes of FGF23-null mice

indicated an association between FGF23 and α-Klotho (Kuro-o et al. 1997; Shimada et

al. 2004). Using renal homogenate, Urakawa et al. was for the first time to show that α-

Klotho binds to FGF23. They also found that the binding of α-Klotho was essential for

the activation of FGF23 signaling since FGF receptor 1 converts to a receptor specific

for FGF23only in the presence of α-Klotho (Urakawa et al. 2006). Supplement of

FGF23 did not reduce serum phosphate in FGF23(-/-)/klotho(-/-) and klotho(-/-) mice,

nevertheless, it improved serum phosphate in wild-type and FGF23(-/-) mice, which

further proved that FGF23 regulated phosphate homeostasis dependent on α-Klotho

(Nakatani et al. 2009).

Page 26: New insights in parathyroid hormone secretion

26

Other regulators

Divalent cations (Mg2+

, Mn2+

, Ba2+

, and Sr2+

)

Besides Ca2+

, many other divalent cations also influenced PTH secretion. Mg2+

was

found to have duplicate effect on Ca2+

regulated PTH secretion. When Mg2+

was less

than 0.8mM, it impaired low Ca2+

stimulated PTH release. 0.8-1 mM Mg2+

gave the

maximal stimulation of PTH secretion, and when it was higher than 1 mM, it

progressively inhibits PTH release (Takatsuki et al. 1980). When compared the

efficiency on PTH secretion, Mn2+

>Ca2+

=Sr2+

>Mg2+

. Ba2+

has not obvious effect at

concentration of 0.5-3.25 mM (Wallace & Scarpa 1982). The inhibition of PTH

secretion by other cations was not due to Ca2+

influx because Sr2+

, Mg2+

and Ba2+

were

only evoked rapid transient but no sustained increased in Ca2+

i (Nemeth & Scarpa

1987). Mn2+

also induced transient increased in Ca2+

i, but followed by a decrease of

Ca2+

uptake (Johansson et al. 1988). Similarly, those ions were found to increase IP3,

IP2, and IP1 in parathyroid cells (Shoback et al. 1988). Therefore, there it is likely that

membrane receptors are activated by divalent cations and induced IP3 related Ca2+

mobilization. The mechanism is still not clear.

Lithium

After the first case of Lithium associated hyperparathyroidism was reported in 1973

(Garfinkel et al. 1973), several studies have further conformed the stimulation effect of

Lithium on PTH secretion (Birnbaum et al. 1988; Graze 1981; Saxe et al. 1995). The

stimulation of PTH is likely due to decreased Ca2+

e sensitivity on receptor level (Brown

1981; McHenry et al. 1991), since Lithium do not change the level of Ca2+

i and cAMP

(Wallace & Scarpa 1983). Preincubation with LiCl caused a dose-dependent right-shift

of ‘calcium set point’ in bovine parathyroid cells (Brown 1981). Interestingly, CaSR

agonist, normalizes the hypercalcemia and hyperparathyroidism induced by lithium

(Sloand & Shelly 2006).

Adenosine-5'-triphosphate

Adenosine-5'-triphosphate (ATP) and its non-metabolizable analog Adenosine 5′-O-(3-

thio) triphosphate (ATPγS) were found to inhibit PTH secretion (Nemeth & Kosz

Page 27: New insights in parathyroid hormone secretion

27

1989). When applied to parathyroid cell, they caused a transient increase of Ca2+

i due

to mobilization of intracellular calcium store, which may suggest the presence of

purinoceptor P2 receptor (Nemeth & Kosz 1989). But more studies are needed to

further elucidate this pathway.

Estrogen

The prevalence of hyperparathyroidism is increased in postmenopausal women, which

indicates a possible association between estrogen and parathyroid disease. Estrogen

receptor α and β has been demonstrated in human parathyroid cells, and it was

predicted an increased expression in pathological parathyroid tissue compared with

normal tissue (Wong et al. 2002). In vitro studies show that physiologic concentration

of estrogen or estrogen receptor modulators increased PTH secretion (Duarte et al.

1988; Greenberg et al. 1987). In vivo, estrogen replacement improved calcium

sensitivity of parathyroid glands (Boucher et al. 1989; Zofkova et al. 1993).

Prolactin

Many studies have shown that hyperprolactinemia is associated with increased bone

loss (Palmer et al. 1988). This correlation is not fully elucidated. Some studies have

shown it may be due to hypogonadism, others found that prolactin has direct effects on

vitamin D and calcium metabolism, whereas, others suggests that prolactin has its

mode of action on bone directly (Seriwatanachai et al. 2008). From our perspective, it

is interesting that one study showed that prolactin directly stimulates PTH secretion in

bovine parathyroid cells (Magliola & Forte 1984).

Page 28: New insights in parathyroid hormone secretion

28

Aim of the study

The overall aim of the thesis was to enhance the understanding of stimulus-secretion

coupling in human parathyroid.

More specific, the aims for individual papers were:

1. to characterize the exocytotic proteins in human parathyroid, with special

focus on SNAP-23, SNAP-25, Syntaxin1 and VAMP (paper I).

2. to identify the mechanism of Ca2+

entry and Ca2+

handling proteins in human

parathyroid cells (paper II and III).

3. to investigate the role of Ca2+

-activated K+ channel and membrane potential in

relation to PTH secretion (paper IV).

Page 29: New insights in parathyroid hormone secretion

29

Materials and Methods

Human parathyroid specimens

All molecular analysis in this study was done on human parathyroid samples. All

parathyroid tissues were classified according to the World Health Organization (WHO)

guideline (DeLellis RA et al. 2004). In paper I, besides seven parathyroid adenomas

and fifteen normal parathyroids that were collected from the Mater Private Hospital,

North Sydney, NSW, Australia or the Royal North Shore Hospital, St. Leonards, NSW,

Australia and seven parathyroid carcinomas that were collected world widely, all other

parathyroid tissues were obtained from ‘Endocrine BioBank’ in the Department of

Oncology and Pathology at Karolinska University Hospital. The same twenty

parathyroid adenomas from Karolinska University Hospital were used in Paper II, III

and IV for protein and gene expression analysis. All parathyroid adenomas used for

PTH secretion study, electrophysiological study and Ca2+

i measurement in paper II, III

and IV were gotten freshly from surgery at Karolinska University Hospital between

2006 and 2009. All samples collection was under patient’s consent and approved by

local ethic committees.

Immunohistochemistry (Paper I and IV)

Immunohistochemistry (IHC) is a method to study the spatial expression pattern of a

certain protein within a tissue or other cellular structures. The process includes the use

of an antibody specifically binding to one antigen and then visualization of the antigen-

antibody complex by fluorescent dye, enzyme or radioactive element.

Generally, IHC is performed in formalin fixed paraffin-embedded tissue sections.

Paraffin-embedded sections provide a clear tissue structure and cell morphology, but

some antigens are easily degraded by formalin fixation and paraffin embedding.

Therefore, frozen tissue section is required in certain analysis. The tissues are freshly

frozen, then cut and fixed with cold acetone or formalin. Frozen secretions conserve

more antigens, but poor morphology reduces the specificity and quality. Antigen

retrieval is a process specifically used for Paraffin-embedded sections, in which

sections are heated with Citrate or EDTA or digested with enzyme to break the protein

Page 30: New insights in parathyroid hormone secretion

30

cross-linking formed by formalin fixation and unmask antigenic binding sites. Before

adding the antibody, fat-free milk or normal serum is usually applied to reduce the

background staining. The detecting method can be either direct or indirect. When using

the direct method, the indicator (fluorescent dye, biotin or Peroxidase) is conjugated to

an antibody, including only one step staining. The indirect method includes an

unlabeled primary antibody that binds to antigens, a conjugated secondary antibody that

reacts to primary antibody, and then a third layer with a complex recognizing the

secondary antibody. The indirect method is thereby more specific and provide higher

signal compared with direct method.

The method used in Paper I is a

classical indirect method called ABC.

The technique involves three layers:

unlabeled primary antibody,

biotinylated secondary antibody and a

complex of avidin-biotin peroxidase.

The signal is visualized by DAB

because DAB is converted to insoluble

brown substance by peroxidase in the

presence of H2O2 (hydrogen peroxide).

Figure 6. Illustration of ABC method.

Western blot (paper I-IV)

Western blot is also a method to expression by using an antigen-antibody reaction.

Instead of tissue sections, cell extracts were used for Western blot. SDS is a detergent

that can denature multiple proteins into linear structure and coated with negative

charges. Polyacrylamide gel contains tunnels with different diameters, which allow

proteins going through by their sizes under the same electric current. Therefore, after

SDS-PAGE (sodium dodecyl sulfate polyacrylamide gel electrophoresis), proteins are

separated on gel according to their molecular weight. Separated proteins were then

blotted on nitrocellulose or Polyvinylidene fluoride (PVDF) membranes for antigen-

antibody reaction. Generally, nitrocellulose membrane is easier to handle since it wets

naturally and gets high signal and low background. PVDF membrane, on the other

hand, needs to be prewetted in methanol and is sensitive to SDS level on the gel.

Page 31: New insights in parathyroid hormone secretion

31

Detection of the protein includes a primary antibody specific to the protein of interest, a

secondary antibody conjugated with an indicator or substrate interacting with indicator.

The commonly used indicator is horseradish peroxidase (HRP). An Enhanced luminol-

based chemiluminescent (ECL) converts the HRP signal into light and detects by film.

The representativity of Western blot is very much depending on the specificity and

sensitivity of the primary antibody. In general, monoclonal antibody is more specific

than polyclonal antibody.

Protein complex immunoprecipitation (Co-IP) (Paper II)

Co-IP is a powerful method used to study of intact protein protein interaction. In order

to evaluate suspected protein interaction, an antibody that targets on a known protein of

the study protein complex is mixed with cell or tissue lysis buffer. After incubation, a

complex of antibody against proteins of interest is formed. Protein A or G agarose is

then added to capture the complex. Proteins that interact with the antigen-antibody

complex are also pulled down after centrifugation. Free proteins are washed way.

Precipitated products are finally analyzed by immunoblotting to verify the interacted

proteins. To further confirm the results, experiments are repeated by using antibodies

identified other proteins in the complex. The proteins interaction will be questioned if

antibody against any membrane of the complex cannot precipitate other proteins in the

complex.

Reverse transcription (RT) polymerase chain reaction (PCR) and quantitative real-

time (qRT) PCR (Paper II and III)

Polymerase chain reaction (PCR) is a technique to amplify a certain region of DNA

strand, which can provide quantitative or qualititative information about a specific

gene. The reaction includes denaturation, annealing and extension. Double strand DNA

is denatured into single strand at 94-98C. When temperature goes down to 50-65

C,

primers will bind to DNA template and the DNA polymerase can subsequently

facilitate the duplication of the sequence of interest. After that, when temperature is

raised to 68-75C, the optimal working temperature for polymerase, primers are

continuously extended by DNA polymerase. The same cycles are usually repeated for

30-45 times resulting in millions of copies of DNA fragment of interest. If real time

monitoring the process, the PCR reaction progress can artificially be divided into three

Page 32: New insights in parathyroid hormone secretion

32

phases: exponential phase in which reaction goes quickly because of sufficient working

materials, linear phase in which amplification starts to slow down, finally plateau phase

in which reaction has stopped and products start to degrade.

RT-PCR and qRT-PCR were

the two most widely used PCR

for gene analysis. In RT-PCR,

single strand RNA is firstly

reverse transcribed into its

complementary DNA (cDNA)

using reverse transcriptase and

primers, then cDNA is used as

template to amplify specific

DNA region by traditional

PCR. The PCR products were

detected using agarose gel

electrophoresis and ethidium

bromide or Gelred. By this

method, we verify the

expression of target gene. The

product size or sequence

analysis of the PCR products

helps to verify the correct

products. Because the agarose

gel only detects the endpoint

(plateau phrase) products, it

only provides quality of the

gene and is imprecise to predict

the quantity of templates.

qRT-PCR, on other hand, provides information of the whole PCR proceeding, which is

more accurate for DNA or RNA quantification. The data collection of qRT-PCR is

fluorescence based. Two commonly used chemistries for detection are Taqman probe

and SYBR green. SYBR green directly binds to double-stranded DNA and emits light.

TaqMan probes are oligonucleotides with a high energy reporter dye on the 5' end and a

Page 33: New insights in parathyroid hormone secretion

33

low energy quencher dye at the 3' end. Before reaction, two dyes sit together by which

the emission of reporter dye is suppressed by quencher dye. During PCR, when DNA

replication reaches to the TaqMan probes bonded on template, the 5'-nuclease of

polymerase cleaves the probe. The release of reporter from quencher increases

fluorescent emission of reporter. The signal increases in direct proportion to the amount

of PCR product in the reaction. SYBR green dye, on the other hand, binds to any

double-stranded DNA molecular including primer-dimers and other non-specific

reaction products, which may lead to an overestimate of the PCR product. Therefore,

Taqman method is a more precise and advance method compared to SYBR green.

Figure 8. Illustration of TaqMan qRT-PCR.

Human Parathyroid cells preparation

In this thesis, primary cultured human parathyroid cells were used for hormone

secretion, intracellular calcium measurement and electrophysiological studies. Fresh

human parathyroid tissue was put into MEM medium (Hank’s) right after dissection

and transported to the lab for isolation within one hour. Tissue was cut into small pieces

and digested in 1.5 mg/ml Type II collagenase for 30 min, then triturated with syringe.

After washed twice with MEM medium, cells were placed and cultured within

DMEM/F-12 medium with supplement of 10% fetal calf serum and 1% antibiotic.

Cultivation of parathyroid cells has been a challenge because cells quickly lose their

secretion property during culture. In my hands, cells completely lose their ability to

release PTH on the third week of normal tissue culture. Type I collagenase and other

cultured mediums, e.g. DMEM, RPMI-1640, and SFM (serum-free keratinocyte)

Page 34: New insights in parathyroid hormone secretion

34

supplied with growth factor, have been used for isolation and culture by other groups.

SFM is a serum free medium containing only 0.09 mM Ca2+

. It has been shown to

provide functional attached culture for up to 2 months, but we did not use it for our

culture because of the poor attachment of the cells in SFM. The DMEM/F-12 medium

used in our study has a more physiological level of calcium (1.05 mM Ca2+

) and was

shown to give functional suspension culture for three months (Kanai et al. 2009).

Hormone secretion (Paper II, III and IV)

There are two ways to assess hormone secretion. One is batch incubation of cells on

tissue culture dishes. Intervenors are directly applied to cells after cell attached and

supernatant is collected for hormone measurement at certain time points. The weakness

of this method is the inconstant amount of cells which makes it imprecise to compare

samples. Another method that is used in this thesis is cell perfusion. Cells are put in a

column between gel filters. Chemicals are added to cells by a perfusion system and

samples are collected at certain times. In this method, results are more reliable and

comparable because of the constant number of cells.

Figure 9. Illustration of parathyroid cell preparation and hormone secretion.

Ca2+

i measurement with Fura-2 (Paper II, III and IV)

In this study, intracellular calcium was detected by fluorescence measurement using

Fura-2. Fura-2 is a commonly used fluorescent indicator of Ca2+

. With the binding of

acetoxymethyl (AM) esters, Fura-2 is transported into cells. After hydrolysis by

Page 35: New insights in parathyroid hormone secretion

35

intracellular esterases, Fura-2 is free

in the intracellular space. Fura-2 has

excitation wavelength at 340 nm and

380 nm. As shown in figure, when it

is free from Ca2+

, excitation is equal

at both wavelengths. When it binds

to Ca2+

, the excitation signal at 340

nm increases in a Ca2+

dependent

manner, whereas signals at 380 nm

are reduced. Therefore, measurement

of fluorescence at two wavelengths gives information of Ca2+

i independent of cytosolic

dye concentration, cell thickness and excitation light intensity (Hayashi & Miyata 1994;

Takahashi et al. 1999; Tsien et al. 1985).

The equipments required for this method include an inverted microscope with a 40×

objective; a perfusion system with heating system which keeps cells at 37C; xenon arc

lamp for light source; monochromaters for selecting wavelength; a shutter controlling

the duration of excitation pulses; a charge-coupled device (CCD) camera detecting

fluorescence signal and

image processor to

convert fluorescent

image to digital number

displayed by computer.

The working flowchart

is shown as Figure 11

(Hayashi & Miyata

1994).

Figure 11. Working flowchart of Ca2+

i measurement.

However, we have to consider some limitations when using a fluorescent dye. The

common problem is Ca2+

insensitivity. The main reason is Fura-2/AM doesn’t

completely convert to free dye in the cells which results in a high signal of fluorescence

but insensitive to the change of calcium concentration. Furthermore, longer loading

Page 36: New insights in parathyroid hormone secretion

36

time can cause compartmentation which means that Fura-2 is accumulating in

intracellular organelles. High concentration of dye can buffer Ca2+

i lowering Ca2+

i or

delaying Ca2+

transit. Bleaching and autofluorescence can also reduce the reliability of

Ca2+

i measurement.

Electrophysiological technique (Paper II and IV)

Patch clamp technique is one of the electrophysiological methods used for the study of

ion channels. It was developed by Erwin Neher and Bert Sakmann in 1970’s (Neher &

Sakmann 1976; Neher et al. 1978).

In this method, a glass micropipette with a flat small tip (around 1 µm) is used as an

electrode. By direct contact and additional suction, a high resistance seal (> 1 GΩ), is

formed between micropipette and cell plasma membrane. The high resistance seal

provides reduced noise and stable recording. Proper buffer that depends on the purpose

of measurement and recording configuration is filled in the pipette. A chloride silver

wire contacts with buffer and conducts electrical current to the amplifier. The way for

recording is either voltage-clamp or current-clamp. In voltage-clamp, the voltage is

fixed and the changes on current are observed, for example recording of ion channel

current. The other way around, current-clamp monitors the changes in voltage while

current is constant, such as the measurement of membrane potential.

Five patch techniques can be used for different purposes. As shown in Figure 12, on-

cell patch is the fundamental configuration from which the other four configurations are

derived. In on-cell patch, the pipette is tightly sealed to the cell membrane without

damage. This method allows us to measure the current from one or few ion channels

within pipette. If deeper suction was given after gigaseal to break the membrane

between pipette and cells, we get whole-cell configuration. Instead of single channel,

whole-cell patch record multiple ion channels on the entire cell. Since there is an open

connection between cell and pipette, the intracellular content is dialyzed by pipette

solution. Therefore, it will not be a good choice for the study of channel activated

through second messengers. To avoid this problem, other type of whole-cell patch

called perforated-patch can be used. Instead of breaking the membrane violently, small

amounts of antibiotic, such as amphothericin-B or gramicidin, are used to create small

pores for monovalent ions going through and generating current. This method reduces

Page 37: New insights in parathyroid hormone secretion

37

the dialysis of cell contents, but also creates some problems. The antibiotic in pipette

makes it difficult to get gigaseal to cell membrane, it takes a long time to get

perforation, contact may be lost or rupture of the membrane may occur. After on-cell

patch is formed, if the pipette is quickly pulled out from the cell, cell membrane on the

tip of pipette is parted from the cell and sticks onto pipette, the inner surface of cell

membrane will be exposed to the bath solution. This configuration is called inside-out

patch. It is useful to study channels activated by inner ligand, such as ATP-dependent

K+ channel. If the pipette is slowly pulled out from the cell after whole-cell patch, the

cell membrane around pipette can be ripped out and then form a bulb on the top of

pipette. That is called outside-out patch. This method reduces the effect of second

messengers and provides better way to study ion channels activated on outer face.

In this thesis, whole-cell patch was used for measurement of voltage operated calcium

channel, store operated calcium channel (Paper II) and Ca2+

-activated K+ channel and

perforated-patch was performed to measure membrane potential (Paper IV).

Figure 12. Illustration of patch-clamp technique and configurations.

Page 38: New insights in parathyroid hormone secretion

38

Results and discussion

Paper I - Heterogeneous expression of SNARE proteins SNAP-23, SNAP-25,

Syntaxin1 and VAMP in human parathyroid tissue

Like in other endocrine cells, parathyroid hormone is stored in vesicles and released via

exocytosis by stimulation of external signal. Exocytosis is a complicated process

requiring participation of several proteins. SNAREs are the central players in

membrane fusion. According to the location, SNAREs are classified into two groups:

v-SNAREs and t-SNAREs. v-SNAREs include VAMP family and are located on

vesicle membrane. t-SNAREs are composed of SNAP and syntaxin families located on

plasma membrane. It is well established that the formation of SNAREs complex by

SNAP-25, Syntaxin1 and VAMP2 is essential for neurotransmitter and insulin secretion

in synapse and pancreatic β-cells (Chen & Scheller 2001). The exocytotic machinery

has so far not been investigated in parathyroid cells. Instead of triggering by increased

Ca2+

i, parathyroid hormone is released when serum calcium is decreased. Therefore, it

is possible that parathyroid cells have a different setup of exocytotic proteins.

To answer this question, we first analyzed the expression of SNAP-25, Syntaxin1 and

VAMP proteins in 20 human parathyroid chief cell adenomas and a pool of three

normal tissues using Western blot analysis. Surprisingly, we did not detect any signal

of SNAP-25 and Syntaxin1 in normal parathyroid tissues. And only 5 out of 20

adenomas did show the expression of SNAP-25 and Syntaxin1 proteins. The absence of

SNAP-25 protein in parathyroid tissues indicated that SNAP-25 is not responsible for

hormone secretion machinery in general. We therefore extended our search and turned

to SNAP-23, a homologue that shares 59% identity of SNAP-25. In contrast to SNAP-

25, which requires relatively high Ca2+

(1 M) to trigging the formation of SNARE

complex, SNAP-23 is able to mediate vesicle docking and fusion at resting condition

(100 nM Ca2+

) (Chieregatti et al. 2004).

Using a specific anti-SNAP-23 antibody, we detected the expression of SNAP-23 in all

parathyroid adenoma and a pool of normal tissues (Figure 1, paper I). An antibody

recognized VAMP1-3 proteins also detected clear signals in all adenoma and normal

parathyroid tissues. These data implied that SNAP-23 and VAMP are more likely to be

Page 39: New insights in parathyroid hormone secretion

39

involved in parathyroid hormone secretion.

To further evaluate the possible role of SNAP-25 and SNAP-23 in human parathyroid

cells, a total of 51 parathyroid specimens, which included 27 chief cell adenomas, 15

normal parathyroid and 9 carcinomas, were used for immunostaining of SNAP-25,

SNAP-23 and PTH. Similar to Western blot analysis, immunohistochemical

investigation showed even expression of SNAP-23 in all examined samples. SNAP-23

was expressed both in the plasma membrane and in the cytoplasm, whereas SNAP-25

is mainly accumulated in plasma membrane (Figure 4, paper I). Parallel to Western

blot, immunoreactivity of SNAP-25 was also absent in all normal parathyroids.

Interestingly, the expression pattern of SNAP-25 in parathyroid adenoma varied.

Thirteen out of them were totally blank, five out of them were widely positive, three

out of them showed nodular expression, while six of them only has staining in single

cells. No correlation was found between SNAP-25 expression and PTH

immunoreactivity. Neither was there a correlation between serum calcium, tumor

weight and plasma PTH level and SNAP-25 level, implying lack of function of SNAP-

25 in parathyroid physiology and pathophysiology. Notably, SNAP-25 has higher

expressions in carcinomas (45%) compared with adenomas (20%). The specific

expression of SNAP-25 in tumor may indicate low differentiation of tumor since

increased SNAP-25 has been shown in human undifferentiated colon and rectum

carcinomas and small cell lung carcinomas with poor prognosis (Grabowski et al. 2004;

Graff et al. 2001). Another hypothesis of increased SNAP-25 is the amplification of

SNAP-25 gene (located in chromosome 20p11). However, published genetic analysis

papers did not show a chromosomal unbalance in chromosome 20 in parathyroid

adenomas and carcinomas (Kytola et al. 2000; Palanisamy et al. 1998). The nodular

expression of SNAP-25 in a few cases suggested the occurrence of an additional

genetic hit during tumorigenesis.

In summary, in contrast to other endocrine cells, SNAP-23 and VAMP play a role in

calcium regulated parathyroid hormone secretion. SNAP-25 and Syntaxin1, on the

other hand, may serve an undefined tumor related function in parathyroid cells.

Page 40: New insights in parathyroid hormone secretion

40

Paper II - Expression and association of TRPC subtypes with Orai1 and STIM1 in

human parathyroid

It has been well established that increased Ca2+

e induces increase of Ca2+

i characterized

by a transient spark followed by a steady-state elevation in parathyroid cells (LeBoff et

al. 1985). Using radioactive labeled Ca2+

, it has been shown that the first transient spark

is due to calcium release from intracellular calcium stores activated by increased IP3

following CaSR activation, while the second phase is dependent on calcium influx

(Wallfelt et al. 1985). However, it is still unclear what type of calcium channel that

mediates calcium influx in parathyroid cells.

The first question was if there is voltage operated and or voltage insensitive calcium

channels in parathyroid cells. Therefore, patch-clamp studies were performed to

investigate possible presence of voltage-gated calcium channels. However, step

depolarization from -70 mV to 70 mV on whole cell configuration did not trigger any

detectable current neither in normal cells nor in cells from parathyroid adenoma. As a

control, on the other hand, the same protocol evoked a clear inward current in mouse

pancreatic β-cells. Besides voltage-gated calcium channel, store operated calcium entry

is another way for calcium influx, which occurs after emptying of intracellular calcium

stores. Thapsigargin, an inhibitor of SERCA, is a reliable tool to deplete calcium store

and trigger calcium entry. To begin with, 2 M thapsigargin was applied to single

parathyroid cells when performing voltage-ramps. The use of ramps demonstrates that

no voltage-gated channels are activated. Our results show a marked increase of current

after exposure to thapasigargin. In parallel, Fura-2 measurements showed a larger rise

of Ca2+

i at 1.5 mM Ca2+

e after addition of thapsigargin. 2-APB, a store operated

calcium channel blocker, totally blocked thapsigargin induced Ca2+

i increase. This

finding implies the presence of store operated calcium influx in human parathyroid

cells.

Recent findings in other cell types show that TRPC channels, Orai1 and STIM1 are key

players in store operated calcium entry (Ambudkar et al. 2007; Potier & Trebak 2008).

In TRPC family, besides of TRPC2 which is known as pseudogene, all other members

TRPC1, 3-7 have been shown to be activated by stored depletion (Salido et al. 2009).

Screening of normal and adenoma parathyroid tissue using RT-PCR showed expression

of TRPC1, 4 and 6, but not TRPC3, 5 and 7. The level of TRPC1 expression was

Page 41: New insights in parathyroid hormone secretion

41

significantly higher than that of TRPC4 and 6, detected by qRT-PCR. However, no

statistical comparisons of these genes could be made between normal and adenomas.

qRT-PCR and Western-blot showed the general expression of STIM1 and Orai1.

Finally, co-IP gave strong evidence of the interaction between TRPC1, STIM1 and

Orai1.

In conclusion, we show that store operated calcium channels composed by TRPC1-

STIM1-Orai1 complex mostly likely are involved in mediating calcium influx in

human parathyroid.

Paper III - Involvement of calmodulin and calmodulin dependent protein kinase II

(CaMKII) in human parathyroid hormone secretion

CaSR senses small elevation in Ca2+

e and converts to several folds increase of Ca2+

i

inhibiting PTH secretion via Ca2+

binding proteins pathway. Among the numerous of

Ca2+

binding proteins described in the literature, calmodulin is the most ubiquitous

protein mediating many Ca2+

regulated cellular functions by binding to different protein

targets. One of the best-described targets and also shown to be involved in exocytosis is

calmodulin dependent protein kinase II (CaMKII)(Wang 2008). Both calmodulin and

CaMKII have been found in parathyroid cells in the early 1980’s and 1990’s (Brown et

al. 1981; Kato et al. 1991; Kinder et al. 1987; Oldham et al. 1982). However, the

correlation between function of calmodulin and CaMKII is still unclear.

Since the technical limitation in the earlier years, calmodulin and CaMKII were

identified indirectly by detection of calmodulin dependent phosphodiesterase activity.

Therefore, to further demonstrate their expression, we performed RT-PCR with primers

specific recognized calmodulin isoforms 1-3 and four CaMKII subunits mRNA and

Western-blot with specific antibodies against those proteins. Our results clearly show

the expression of calmodulin and CaMKII in human normal and adenomatous

parathyroid cells at both mRNA and protein levels (Figure 1, paper III).

To investigate potential association between calmodulin/CaMKII and PTH secretion,

calmodulin and CaMKII antagonists, Calmidazolium and KN-62, were used to block

Calmodulin and CaMKII activity, respectively. Exposing parathyroid cell suspension to

1 µM and 10 µM Calmidazolium caused 309.4% and 260130% increase of PTH

Page 42: New insights in parathyroid hormone secretion

42

secretion, respectively. KN-62 resulted in a 344% increase of PTH release (Figure 2,

paper III). This finding suggests that calmodulin and CaMKII are involved in PTH

secretion.

As stated above, Ca2+

i is negatively related to PTH secretion. Therefore, Ca2+

i

measurement was done to investigate the mechanism of calmodulin and CaMKII

regulated PTH secretion. Our studies shows that calmodulin and CaMKII regulates

PTH secretion regardless of Ca2+

i. since Calmidazolium induced an increase of Ca2+

i,

and 10 µM KN-62 has no effect on Ca2+

i (Figure 3, paper III). It also indicates that

Ca2+

i may not be pivotal regulator in stimulus-secretion coupling in parathyroid.

Calmodulin and/or CaMKII may directly or indirectly interact with exocytotic proteins

and influences exocytosis.

CaMKII needs to be phosphorylated in order to be physiological active (pCaMKII).

Reviewing a cohort of 20 chief cell parathyroid adenomas for calmodulin and

pCaMKII, we found that calmodulin was equally distributed in all adenomas. There is

no correlation between S-Ca2+

, S-PTH, tumor weight and calmodulin protein level.

However, a negative correlation was shown between the level of pCaMKII and S-Ca2+

.

In vitro studies also found the reduction of pCaMKII after treated with 2.5 mM Ca2+

. It

is concluded that high Ca2+

e suppress the phosphorylation of CaMKII.

In conclusion, this study demonstrates the expression of calmodulin and CaMKII in

human parathyroid, and that these proteins are involved in PTH secretion. Reduction of

phosphorylation of CaMKII at high Ca2+

e may have pathophysiological implication that

needs to be further addressed.

Paper IV - Expression of Ca2+

-activated K+ channels in human parathyroid cell:

potential role in hormone secretion

Membrane potential is a key player of hormone secretion in neurons and

neuroendocrine cells. In pancreatic -cells, for example, it has been well known that

membrane depolarization induced by the closure of ATP-dependent K+ channels give

rise to insulin secretion (Ashcroft 2000). Similarly, membrane depolarization increases

of PTH release in parathyroid cells (Dempster et al. 1982). Wallfelt and co-workers

found that elevated cytoplasmatic Ca2+

triggered K+ efflux when monitoring by K

+

Page 43: New insights in parathyroid hormone secretion

43

tractor 86R (Wallfelt et al. 1985). Taken together, it suggests the existence of Ca2+

activated K+ permeability. This hypothesis was latterly confirmed by our group by

detection of Ca2+

-activated K+ current using patch-clamp (Valimaki et al. 2003).

Distinguishably, high Ca2+

o, which inhibits PTH secretion also hyperpolarize

parathyroid cell membrane (Valimaki et al. 2003). In paper IV, we show that

parathyroid express at least two types of Ca2+

-activated K+ channels in human

parathyroid cell membrane, namely the big conductance Ca2+

-activated K+ channels

(KBK channel) and small conductance Ca2+

-activated K+ channels (KSK channel) by

applying specific channel blockers (Figure 1, paper IV). The presence of these channels

was further verified by Western blot and immunocytochemistry.

When we investigated the relationship of Ca2+

i and Ca2+

-activated K+ current, we found

that adenomas cells had significantly lager K+ current at 0.3 µM than normal

parathyroid cells. However, Western blot analysis could not detect any difference in

KBK channel protein level.

Using perforated-patch technique, we show that both high extracellular K+

concentration and KBK channel blocker toxins lead to depolarization of parathyroid cell,

while KBK channel activator (NS-11021) caused hyperpolarization. Using a perfusion

chamber, exposing parathyroid cells to KBK channel blockers (TEA and charybdotoxin)

induce PTH secretion at 1.0 and 1.2 mM Ca2+

e, but no KSK channel blocker (apamin).

The KBK channel activator NS-11021 showed an inhibition of PTH secretion at 0.5-1.5

mM Ca2+

e without alterations in Ca2+

i.

In summary; we conclude that parathyroid cells, normal and adenoma, express Ca2+

-

activated K+ channels. The majority of Ca

2+-activated K

+ channel current can be

assigned to KSK and KBK channel. Moreover, these channels contribute in maintaining

the membrane potential in the parathyroid cell, where opening of these channels

hyperpolarize the membrane and vice verse. Membrane potential seems to be, at least

partly, a Ca2+

i independent modulator of PTH secretion. Our finding that parathyroid

adenoma cells have higher K+ current compared to normal cells is potentially very

interesting, even though we are not able to verify this finding on protein level.

However, there are several explanations for our incongruent finding. Even so, one may

speculate that an increased K+ channel activity could result in a more hyperpolarized

cell. Down-stream in the signaling pathway this could decrease PTH secretion. Since

Page 44: New insights in parathyroid hormone secretion

44

KBK channel gene is located in chromosome 10q22, a genomic area which has not been

shown to harbor chromosomal unbalance (Kytola et al. 2000; Palanisamy et al. 1998),

it is plausible that an increased activity represents a compensatory mechanism for a

decrease PTH secretion in parathyroid adenoma.

Conclusions

The aim of this thesis was to characterize the stimulus-secretion coupling in human

parathyroid. The major findings in summary:

1. In contrast to other endocrine cells, SNAP-23 and VAMP are widely

expressed in normal and adenoma parathyroid tissue. It is likely that these

proteins play a role in calcium regulated hormone secretion. SNAP-25 and

Syntaxin1, on the other hand, may serve an undefined tumor related function

in parathyroid cells.

2. Store operated calcium channels composed by TRPC1-STIM1-Orai1 complex

mostly likely are involved in mediating calcium influx in human parathyroid.

Calmodulin and CaMKII are involved in Ca2+

regulated PTH secretion. High

calcium suppresses the phosphorylation of CaMKII, which may have a

pathophysiological implication.

3. At least two types of Ca2+

-activated K+ channels, namely KBK and KSK channels

are present in the human parathyroid cell. These channels contribute in

maintaining the membrane potential in the parathyroid cell, and membrane

potential seems to be, at least partly, a Ca2+

i independent modulator of PTH

secretion.

Page 45: New insights in parathyroid hormone secretion

45

Figure 13. Summary of main findings in the thesis.

Page 46: New insights in parathyroid hormone secretion

46

摘要 甲状旁腺是一个独特的内分泌腺体,因为离子钙抑制激素分泌,而不象在别的

内分泌腺体内刺激激素分泌。它的机制一般认为是,当离子钙结合到细胞膜上的钙

敏感受体,激活了一系列的第二信史从而影响甲状旁腺激素分泌。其中细胞内钙离

子被认为在这激素分泌过程中起着中心的作用,然而许多细胞内的信号机制仍未完

全清楚。

胞吐是激素分泌的主要方式,在调节性胞吐中,突触小体相关蛋白,比如 SNAP

蛋白在激素囊泡和细胞膜融合过程中着关键作用。突触小体相关蛋白广泛存在于神

经和神经内分泌细胞。在第一篇文章中,我们通过免疫组化和蛋白印迹探索突触小

体相关蛋白 SNAP-23,SNAP-25, Syntaxin1 和 VAMP 在人甲状旁腺正常和病理组织的

表达。结果发现 SNAP25 和 Syntaxin1 不存在于正常甲状旁腺内,但表达在 20%的主

细胞腺瘤和 45%的甲状旁腺癌里。反之 SNAP-23和 VAMP 则存在于所有的甲状旁腺标

本里。这些发现暗示蛋白 SNAP-23 和 VAMP,而不是 SNAP-25 和 Syntaxin1,在人甲

状旁腺细胞胞吐中起主要作用。

细胞内钙离子在甲状旁腺激素分泌中起着重要作用,课题 II 和 III 侧重于研

究这个过程。在甲状旁腺中,当细胞外钙离子升高时,钙离子的内流就增加,但机

制并不清楚。使用膜片钳和细胞内钙离子测定技术,我们发现在甲状旁腺细胞中存

在钙池操纵性钙通道,蛋白 TRPC1,STIM1 和 Orai1 组成的复合体可能是这个通道的

主要元素。

另外,两个在钙信号通道中很重要的钙结合蛋白 calmodulin 和 calmodulin 依

赖性激酶也被发现存在于人甲状旁腺中。当他们失活时,甲状旁腺激素分泌增加,

而这个作用与细胞内钙离子的高低没有直接联系。我们的发现首次表明了钙结合蛋

白 calmodulin 和 calmodulin 依赖性激酶参与了甲状旁腺激素分泌的调解。有趣的

是我们还发现了在腺瘤病人中血钙水平和腺瘤组织中磷酸化的 calmodulin 依赖性

激酶水平成反比关系,这也许暗示着一定的临床意义。

最后在文章四,我们发现了甲状旁腺细胞至少存在两种钙激活性钾离子通道,

分别是大电导型和小电导型钙激活性钾离子通道。这些通道对相应的钙激活性钾离

子通道的激活剂和拮抗剂都很敏感。这些通道的开放和关闭直接影响到膜电位的改

变,并因而影响甲状旁腺激素分泌。

概括而言,甲状旁腺细胞的刺激-激素分泌复杂而特别。这本论文从钙内流,

信号传递,离子通道和胞吐不同的方面分别阐述了这个过程。但在体内,这些不同

的部分也许互相影响,共同调节甲状旁腺激素分泌。

Page 47: New insights in parathyroid hormone secretion

47

Acknowledgment

I would like to express my sincere gratitude to all the people who have been involved in

my postgraduate study. Without you, my life in the past four years cannot be so

wonderful. In particular, I would like to thank:

Robert Bränström, my main supervisor, for accepting me as a PhD student and

guiding me into scientific field, for enthusiasm and intelligence in parathyroid research.

Thank you for always encouraging me and giving me time and space to develop

myself, for all the time spent on my projects and thesis.

Lars-Ove Farnebo, my co-supervisor, for financial support for my PhD study, for

broad knowledge in research, for patiently correction and discussion for my

manuscripts and thesis.

Lars-Forsberg, my co-supervisor, for helping me adapting to Swedish life and

research work, for helping me to improve my oral presentation skills. You are like an

elder brother giving me lots of encouragements and advice. My earlier life in Sweden

became much easier with your help.

Zhao Bian, who introduce me to Karolinska Institute, for providing me the opportunity

to study abroad, for trust and encouragements, for personal concern and supports for

my family.

Xia Ning, supervisor of my Master degree in China, for warm greeting cards in every

New Year, for always generously expressing your supports, for help whenever I need.

Other parathyroid coworkers

Catharina Larsson, for organizing wonderful parathyroid meeting, for kindness and

deep knowledge in parathyroid research, for all brilliant comments to my manuscripts.

Anders Höög, the super expert in endocrine pathology and master of Endocrine

Biobank, for arrangement of study samples, for help in reading immunohistochemistry

slides. Lisa Ånfalk, for taking care of Endocrine BioBank.

Page 48: New insights in parathyroid hormone secretion

48

Inga-Lena Nilsson, for sharing your epidemiological study related to parathyroid

tumor. Jörgen Nordenström, for brilliant idea and interesting discussion in

parathyroid meeting, for introduce me to Robert.

Christofer C Juhlin, an outstanding Ph.D, I am very impressed in your thesis. Thank

you for establishing the immunohistochemistry lab and technique helps. Felix

Haglund, for interesting presentation and collaboration. Jamileh Hashemi and

Luqman Sulaiman, big parathyroid adenoma workers, for common interest and

interesting discussion.

Erik Berglund, an enthusiasm medical student and next PhD in our lab, for help and

discussion in patch-clamp and intracellular Ca2+

measurement, for all nice chats.

Collaborators in other lab, Vladana Vukojević, for help in Fluorescent

immunocytochemistry, for kindness and nice collaboration. Christina Bark, for

knowledge in exocytosis, for advice in primer design.

Collaborators from other Universities: Arthur D. Conigrave, Leigh W. Delbridge,

Anthony Gill, Hee-Chang Mun from University of Sydney, Australia and Peyman

Björklund, Gunnar Westin from Uppsala University, for common interest and fruitful

collaborations.

Thank you for all surgeons who have helped me for samples collection in Section of

Endocrine Surgery, Karolinska University Hospital. Thank you for all the patients who

have been involved in my studies.

Other members in surgical research: Bertil Hamberger, Martin Bäckdahl, Marja

Thorén for kindness and interest, Ulla Enberg, a nice lady with strong will, for your

optimism attitude, for organizing nice group parties and delicious dinners in your small

cottage, for tasty cakes for Fika, for nice companion and tips for baking, wish you

endless happiness in another world. Cristina Volpe, a nice roommate and friend during

last period of my PhD study, for so many fun chats and nice companion for thesis

writing. Jan Zedenius, for organizing the wine seminar, for your vast knowledge in

tumor genetic and wine. All the former and present members of wine seminar between

2006 to present, for interesting presentation, discussion and wine tasting.

Page 49: New insights in parathyroid hormone secretion

49

Other former members of our research group: Jussi Sane and Viljam Lindqvist, for

kindness and companion, for technique help for Patch clamp. Cecilia Laurell for

kindly letting me your apartment when I was homeless.

Former and present, Corridor mates in L5:02, L8:01 and L1, thank you for kindness,

help and friendly working environment, Members in Professor Catharina

Larsson’s group: Stefano, Andrii, Anastasios, Lui, Nimrod, David ;

Professor Gunnar Norstedt and members in his group, especially, Yin-Choy (for

humors and always help for my methodological problems), Fahad, Mattias, Anenisia,

Roxana, Cecilia, Louisa, Diego, Irina, Hong-yan, ZiCai; Professor Lars Tenenius

and his group members, especially Yu Ming, Agneta, for nice chats and

encouragements; Professor Tomas Ekström and his group members, Monira,

Zahidul, Mohsen and Anna-Maria; Professor Kerstin Brismar and her group

members: Jacob, Vivek, Stina, Katrin, Michael, Jing; Professor Per-Olof Berggren

(for inviting our group moving to such nice work place) and all his members: Lisa,

Martin and Tilo (all for help in Ca2+

i measurements), Ingo, Barbara, Pilar,

Sergei, Irina, Gian-Carlo, Luo-Sheng, Andrea, Erwin, Jantje, SengEun, Essam,

Shao-Nian, Jia, Yue, Xiao-Lei, Lina, Slavena, Rebecka, Lola, Karin, Ulrika,

Elisabetta (for helpful advice in primary cell culture and hormone secretion and

nice lunch chats), Chris, Christopher, Dominic, Gustavo, Yusuf, Lars, Alan,

Patrick, Aarti, Craig (for help on many scientific questions, for nice collaboration),

Annika and Yvonne for help in the lab, Hannelore for showing perfusion experiment

and issues in laboratory safety.

People in Administration board, Katarina Breitholtz, Christina Bremer, Kerstin

Florell, Anne-Marie Richardsson, Helena Nässén, especially, Ann-Britt Wikström,

for kindness and administrative help for my PhD study. Lennart Helleday and

Thomas Westerberg, for IT maintenance.

My friends: Lin Rui, my dear friend, for accompany in the special time of my life,

watching the birth of my son. Hu Jin and his family, thank for all time spending

together. Zhao Yun-Gang (for always patient ly listening and answering

my idiot scientific questions and helps in molecular biological methods), Xie Hong

and their son, for nice trip in Germany. My neighbors, Sun Ying, Qiu Ping and their

Page 50: New insights in parathyroid hormone secretion

50

families, for good company and charing happiness and secretes. Li Xue-Lian, latifa

bikndarne and Linh Dang for all nice chats and dinners. Xu Hong and Ma Zhu-

Heng family, for invited dinners in your house. Other friends, Liu Tong, Zheng Hui-

Yan, Wei Tian-Ling, Jiao Wei, Meng Chun-Xia, Lin Hui-Qiong, Zhou Ying, Wu

Liang, Tu Dan, Zhang Yun, Zhang Xiao, Li Zheng-Hong, Zhang Dong-Ying,

Jiang Li-Ying. My friends in China, Huang Bo, for teaching me English and

encourage me to come to Sweden, you will be in my memory forever.

My colleagues in my home country, Zhao Jin-Min, President of Hospital, Liu Hong,

chief of department, Huang Hong, Yang Xi, Sun Wen, Qin Bao-Yu, Yao Yan-Bing,

thank all for supports.

我的家人,在此要感谢父母培养了我的个性和引导我走上医学的道路,特别

感谢母亲给我生活上的关心和帮助,感谢弟弟陆泉和弟媳宪春对我的理解,支

持和这几年中对母亲的照顾。感谢一直关心和帮助我们家人的亲戚朋友。感谢

婆婆在我需要的时候,来到瑞典帮我带儿子,支持我的学业。

感谢我的丈夫Robin为了我和我们的儿子来到瑞典, 感谢你多年来对我的督

促和给我们无私的爱。感谢上天给了我一个聪明可爱的儿子典典,你的来到带

给我无限的希望和快乐。

最后,我还要感谢基督教会的沈牧师以及所有给过我丈夫和家庭帮助的人。

Thank you for the First Affiliated Hospital of Guangxi Medical University and

Department of education of GuangXi province, NanNing, Peoples Republic of China

for funding my PhD study within 2006-2007.

The study was financially supported by grants from Swedish Research Council, the

Novo Nordisk Foundation, Funds of Karolinska Institutet, the Tore Nilsson Foundation,

the Thuring Foundation, the Jeansson Foundations, the Åke Wiberg Foundation, the

Göran Gustavsson Foundation for Research in Natural Sciences and Medicine, Magn.

Bergvall Foundations, the Stockholm County Council, the Swedish Cancer Foundation,

and the Knut and Alice Wallenberg Foundation.

Page 51: New insights in parathyroid hormone secretion

51

References

Alberts B, Johnson A, Lewis J, Raff M, Roberts K, et al. 2007. Molecular Biology of

the Cell. Fifth Edition.

Allen TB & Thorburn KM 1981 The oxyphil cell in abnormal parathyroid glands. A

study of 114 cases Arch Pathol Lab Med 105 421-427.

Alvarez-Hernandez D, Naves M, Santamaria I, Menarguez J, Torregrosa V, et al. 2003

Response of parathyroid glands to calcitriol in culture: Is this response mediated

by the genetic polymorphisms in vitamin D receptor? Kidney Int Suppl S19-22.

Ambudkar IS, Ong HL, Liu X, Bandyopadhyay BC & Cheng KT 2007 TRPC1: the

link between functionally distinct store-operated calcium channels Cell Calcium

42 213-223.

Ashcroft SJ 2000 The beta-cell K(ATP) channel J Membr Biol 176 187-206.

Bacskai BJ & Friedman PA 1990 Activation of latent Ca2+ channels in renal epithelial

cells by parathyroid hormone Nature 347 388-391.

Bedetti CD, Dekker A & Watson CG 1984 Functioning oxyphil cell adenoma of the

parathyroid gland: a clinicopathologic study of ten patients with

hyperparathyroidism Hum Pathol 15 1121-1126.

Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, Goetz R, Kuro-o M, et al. 2007 The

parathyroid is a target organ for FGF23 in rats J Clin Invest 117 4003-4008.

Berson SA, Yalow RS, Aurbach GD & Potts JT 1963 IMMUNOASSAY OF BOVINE

AND HUMAN PARATHYROID HORMONE Proc Natl Acad Sci U S A 49

613-617.

Birnbaum J, Klandorf H, Giuliano A & Van Herle A 1988 Lithium stimulates the

release of human parathyroid hormone in vitro J Clin Endocrinol Metab 66

1187-1191.

Bjorklund P, Krajisnik T, Akerstrom G, Westin G & Larsson TE 2008 Type I

membrane klotho expression is decreased and inversely correlated to serum

calcium in primary hyperparathyroidism J Clin Endocrinol Metab 93 4152-

4157.

Boucher A, D'Amour P, Hamel L, Fugere P, Gascon-Barre M, et al. 1989 Estrogen

replacement decreases the set point of parathyroid hormone stimulation by

calcium in normal postmenopausal women J Clin Endocrinol Metab 68 831-

836.

Page 52: New insights in parathyroid hormone secretion

52

Bourdeau A, Souberbielle JC, Bonnet P, Herviaux P, Sachs C, et al. 1992

Phospholipase-A2 action and arachidonic acid metabolism in calcium-mediated

parathyroid hormone secretion Endocrinology 130 1339-1344.

Branham JH 1908 I. Tetany following Thyroidectomy Cured by the Subcutaneous

Injection of Parathyroid Emulsion Ann Surg 48 161-164.

Brown AJ, Zhong M, Finch J, Ritter C & Slatopolsky E 1995 The roles of calcium and

1,25-dihydroxyvitamin D3 in the regulation of vitamin D receptor expression

by rat parathyroid glands Endocrinology 136 1419-1425.

Brown EM 1981 Lithium induces abnormal calcium-regulated PTH release in

dispersed bovine parathyroid cells J Clin Endocrinol Metab 52 1046-1048.

Brown EM 1991 Extracellular Ca2+ sensing, regulation of parathyroid cell function,

and role of Ca2+ and other ions as extracellular (first) messengers Physiol Rev

71 371-411.

Brown EM 2007 The calcium-sensing receptor: physiology, pathophysiology and CaR-

based therapeutics Subcell Biochem 45 139-167.

Brown EM, Dawson-Hughes BF, Wilson RE & Adragna N 1981 Calmodulin in

dispersed human parathyroid cells J Clin Endocrinol Metab 53 1064-1071.

Brown EM, Fuleihan Ge-H, Chen CJ & Kifor O 1990 A comparison of the effects of

divalent and trivalent cations on parathyroid hormone release, 3',5'-cyclic-

adenosine monophosphate accumulation, and the levels of inositol phosphates

in bovine parathyroid cells Endocrinology 127 1064-1071.

Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, et al. 1993 Cloning and

characterization of an extracellular Ca(2+)-sensing receptor from bovine

parathyroid Nature 366 575-580.

Brown EM, Gardner DG, Windeck RA & Aurbach GD 1978 Relationship of

intracellular 3',5'-adenosine monophosphate accumulation to parathyroid

hormone release from dispersed bovine parathyroid cells Endocrinology 103

2323-2333.

Carney JA 1996 The glandulae parathyroideae of Ivar Sandstrom. Contributions from

two continents Am J Surg Pathol 20 1123-1144.

Cetani F, Picone A, Cerrai P, Vignali E, Borsari S, et al. 2000 Parathyroid expression

of calcium-sensing receptor protein and in vivo parathyroid hormone-Ca(2+)

set-point in patients with primary hyperparathyroidism J Clin Endocrinol Metab

85 4789-4794.

Page 53: New insights in parathyroid hormone secretion

53

Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ, et al. 2005 The beta-

glucuronidase klotho hydrolyzes and activates the TRPV5 channel Science 310

490-493.

Chang W, Pratt SA, Chen TH, Tu CL, Mikala G, et al. 2001 Parathyroid cells express

dihydropyridine-sensitive cation currents and L-type calcium channel subunits

Am J Physiol Endocrinol Metab 281 E180-189.

Chen YA & Scheller RH 2001 SNARE-mediated membrane fusion Nat Rev Mol Cell

Biol 2 98-106.

Cheng KT, Liu X, Ong HL & Ambudkar IS 2008 Functional requirement for Orai1 in

store-operated TRPC1-STIM1 channels J Biol Chem 283 12935-12940.

Chicka MC, Hui E, Liu H & Chapman ER 2008 Synaptotagmin arrests the SNARE

complex before triggering fast, efficient membrane fusion in response to Ca2+

Nat Struct Mol Biol 15 827-835.

Chieregatti E, Chicka MC, Chapman ER & Baldini G 2004 SNAP-23 functions in

docking/fusion of granules at low Ca2+ Mol Biol Cell 15 1918-1930.

Clapham DE 1995 Calcium signaling Cell 80 259-268.

Clapham DE 2007 Calcium signaling Cell 131 1047-1058.

Collip JB 1925 The Internal Secretion of the Parathyroid Glands Proc Natl Acad Sci U

S A 11 484-485.

Corbetta S, Mantovani G, Lania A, Borgato S, Vicentini L, et al. 2000 Calcium-sensing

receptor expression and signalling in human parathyroid adenomas and primary

hyperplasia Clin Endocrinol (Oxf) 52 339-348.

DeLellis RA, Lloyd RV, Heitz PU & C E 2004. World Health Organization

classification of tumours: Pathology and genetics of tumours of endocrine

organs. IARC Press, Lyon,

Demay MB, Kiernan MS, DeLuca HF & Kronenberg HM 1992 Sequences in the

human parathyroid hormone gene that bind the 1,25-dihydroxyvitamin D3

receptor and mediate transcriptional repression in response to 1,25-

dihydroxyvitamin D3 Proc Natl Acad Sci U S A 89 8097-8101.

Dempster DW, Tobler PH, Olles P, Born W & Fischer JA 1982 Potassium stimulates

parathyroid hormone release from perifused parathyroid cells Endocrinology

111 191-195.

Duarte B, Hargis GK & Kukreja SC 1988 Effects of estradiol and progesterone on

parathyroid hormone secretion from human parathyroid tissue J Clin

Endocrinol Metab 66 584-587.

Page 54: New insights in parathyroid hormone secretion

54

Eknoyan G 1995 A history of the parathyroid glands Am J Kidney Dis 26 801-807.

Ellsworth R 1932 STUDIES ON THE PHYSIOLOGY OF THE PARATHYROID

GLANDS: V. Action of Parathyroid Extract on the Renal Threshold for

Phosphorus J Clin Invest 11 1011-1017.

Farnebo F, Enberg U, Grimelius L, Backdahl M, Schalling M, et al. 1997 Tumor-

specific decreased expression of calcium sensing receptor messenger

ribonucleic acid in sporadic primary hyperparathyroidism J Clin Endocrinol

Metab 82 3481-3486.

Farnebo F, Hoog A, Sandelin K, Larsson C & Farnebo LO 1998 Decreased expression

of calcium-sensing receptor messenger ribonucleic acids in parathyroid

adenomas Surgery 124 1094-1098; discussion 1098-1099.

Ferzandi TR & MacGregor RR 1997 Thapsigargin shifts the Ca set point of parathyroid

cells to lower extracellular [Ca] Endocrine 7 361-366.

Frischauf I, Schindl R, Derler I, Bergsmann J, Fahrner M, et al. 2008 The STIM/Orai

coupling machinery Channels (Austin) 2 261-268.

Gardner DG & Shoback D 2007. Greenspan's Basic & Clinical Endocrinology.

McGraw-Hill Medical Publishing, The 8th edition.

Garfinkel PE, Ezrin C & Stancer HC 1973 Hypothyroidism and hyperparathyroidism

associated with lithium Lancet 2 331-332.

Gerst JE 1999 SNAREs and SNARE regulators in membrane fusion and exocytosis

Cell Mol Life Sci 55 707-734.

Goswami R, Brown EM, Kochupillai N, Gupta N, Rani R, et al. 2004 Prevalence of

calcium sensing receptor autoantibodies in patients with sporadic idiopathic

hypoparathyroidism Eur J Endocrinol 150 9-18.

Grabowski P, Schonfelder J, Ahnert-Hilger G, Foss HD, Stein H, et al. 2004

Heterogeneous expression of neuroendocrine marker proteins in human

undifferentiated carcinoma of the colon and rectum Ann N Y Acad Sci 1014

270-274.

Graff L, Castrop F, Bauer M, Hofler H & Gratzl M 2001 Expression of vesicular

monoamine transporters, synaptosomal-associated protein 25 and syntaxin1: a

signature of human small cell lung carcinoma Cancer Res 61 2138-2144.

Graze KK 1981 Hyperparathyroidism in association with lithium therapy J Clin

Psychiatry 42 38-39.

Page 55: New insights in parathyroid hormone secretion

55

Greenberg C, Kukreja SC, Bowser EN, Hargis GK, Henderson WJ, et al. 1987

Parathyroid hormone secretion: effect of estradiol and progesterone Metabolism

36 151-154.

Habener JF 1981 Regulation of parathyroid hormone secretion and biosynthesis Annu

Rev Physiol 43 211-223.

Hayashi H & Miyata H 1994 Fluorescence imaging of intracellular Ca2+ J Pharmacol

Toxicol Methods 31 1-10.

Heath DA 1989 Primary hyperparathyroidism. Clinical presentation and factors

influencing clinical management Endocrinol Metab Clin North Am 18 631-646.

Henry HL, Midgett RJ & Norman AW 1974 Regulation of 25-hydroxyvitamin D3-1-

hydroxylase in vivo J Biol Chem 249 7584-7592.

Hewavitharana T, Deng X, Soboloff J & Gill DL 2007 Role of STIM and Orai proteins

in the store-operated calcium signaling pathway Cell Calcium 42 173-182.

Imura A, Tsuji Y, Murata M, Maeda R, Kubota K, et al. 2007 alpha-Klotho as a

regulator of calcium homeostasis Science 316 1615-1618.

Inoue R 2005 TRP channels as a newly emerging non-voltage-gated CA2+ entry

channel superfamily Curr Pharm Des 11 1899-1914.

Inoue R, Hanano T, Shi J, Mori Y & Ito Y 2003 Transient receptor potential protein as

a novel non-voltage-gated Ca2+ entry channel involved in diverse

pathophysiological functions J Pharmacol Sci 91 271-276.

Johansson H, Larsson R, Wallfelt C, Rastad J, Akerstrom G, et al. 1988 Calcium-

agonistic action of Mn2+ in the parathyroid cell Mol Cell Endocrinol 59 77-82.

Juppner H, Abou-Samra AB, Freeman M, Kong XF, Schipani E, et al. 1991 A G

protein-linked receptor for parathyroid hormone and parathyroid hormone-

related peptide Science 254 1024-1026.

Kanai G, Kakuta T, Sawada K, Yokoyama TA, Tanaka R, et al. 2009 Suppression of

parathyroid hormone production in vitro and in vivo by RNA interference

Kidney Int 75 490-498.

Kaneko C, Mizunashi K, Tanaka M, Uzuki M, Kikuchi M, et al. 1999 Relationship

between Ca-dependent change of serum PTH and extracellular Ca2+-sensing

receptor expression in parathyroid adenoma Calcif Tissue Int 64 271-272.

Kato M, Hagiwara M, Nimura Y, Shionoya S & Hidaka H 1991 Purification and

characterization of calcium-calmodulin kinase II from human parathyroid

glands J Endocrinol 131 155-162.

Page 56: New insights in parathyroid hormone secretion

56

Kawata T, Imanishi Y, Kobayashi K, Miki T, Arnold A, et al. 2007 Parathyroid

hormone regulates fibroblast growth factor-23 in a mouse model of primary

hyperparathyroidism J Am Soc Nephrol 18 2683-2688.

Kempson SA, Lotscher M, Kaissling B, Biber J, Murer H, et al. 1995 Parathyroid

hormone action on phosphate transporter mRNA and protein in rat renal

proximal tubules Am J Physiol 268 F784-791.

Kifor O, Diaz R, Butters R & Brown EM 1997 The Ca2+-sensing receptor (CaR)

activates phospholipases C, A2, and D in bovine parathyroid and CaR-

transfected, human embryonic kidney (HEK293) cells J Bone Miner Res 12

715-725.

Kifor O, Moore FD, Jr., Wang P, Goldstein M, Vassilev P, et al. 1996 Reduced

immunostaining for the extracellular Ca2+-sensing receptor in primary and

uremic secondary hyperparathyroidism J Clin Endocrinol Metab 81 1598-1606.

Kinder BK, Delahunt NG, Jamieson JD & Gorelick FS 1987 Calcium-calmodulin-

dependent protein kinase in hyperplastic human parathyroid glands

Endocrinology 120 170-177.

Kobayashi K, Imanishi Y, Miyauchi A, Onoda N, Kawata T, et al. 2006 Regulation of

plasma fibroblast growth factor 23 by calcium in primary hyperparathyroidism

Eur J Endocrinol 154 93-99.

Krajisnik T, Bjorklund P, Marsell R, Ljunggren O, Akerstrom G, et al. 2007 Fibroblast

growth factor-23 regulates parathyroid hormone and 1alpha-hydroxylase

expression in cultured bovine parathyroid cells J Endocrinol 195 125-131.

Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, et al. 1997 Mutation of the

mouse klotho gene leads to a syndrome resembling ageing Nature 390 45-51.

Kytola S, Farnebo F, Obara T, Isola J, Grimelius L, et al. 2000 Patterns of

chromosomal imbalances in parathyroid carcinomas Am J Pathol 157 579-586.

Lau K & Bourdeau JE 1995 Parathyroid hormone action in calcium transport in the

distal nephron Curr Opin Nephrol Hypertens 4 55-63.

LeBoff MS, Shoback D, Brown EM, Thatcher J, Leombruno R, et al. 1985 Regulation

of parathyroid hormone release and cytosolic calcium by extracellular calcium

in dispersed and cultured bovine and pathological human parathyroid cells J

Clin Invest 75 49-57.

Levi R, Ben-Dov IZ, Lavi-Moshayoff V, Dinur M, Martin D, et al. 2006 Increased

parathyroid hormone gene expression in secondary hyperparathyroidism of

experimental uremia is reversed by calcimimetics: correlation with

Page 57: New insights in parathyroid hormone secretion

57

posttranslational modification of the trans acting factor AUF1 J Am Soc

Nephrol 17 107-112.

Lindberg JS, Culleton B, Wong G, Borah MF, Clark RV, et al. 2005 Cinacalcet HCl, an

oral calcimimetic agent for the treatment of secondary hyperparathyroidism in

hemodialysis and peritoneal dialysis: a randomized, double-blind, multicenter

study J Am Soc Nephrol 16 800-807.

Lundgren E, Hagstrom EG, Lundin J, Winnerback K, Roos J, et al. 2002 Primary

hyperparathyroidism revisited in menopausal women with serum calcium in the

upper normal range at population-based screening 8 years ago World J Surg 26

931-936.

Lundgren E, Rastad J, Thrufjell E, Akerstrom G & Ljunghall S 1997 Population-based

screening for primary hyperparathyroidism with serum calcium and parathyroid

hormone values in menopausal women Surgery 121 287-294.

Magliola L & Forte LR 1984 Prolactin stimulation of parathyroid hormone secretion in

bovine parathyroid cells Am J Physiol 247 E675-680.

McHenry CR, Racke F, Meister M, Warnaka P, Sarasua M, et al. 1991 Lithium effects

on dispersed bovine parathyroid cells grown in tissue culture Surgery 110 1061-

1066.

Miyamoto K, Ito M, Segawa H & Kuwahata M 2000 Secondary hyperparathyroidism

and phosphate sensing in parathyroid glands J Med Invest 47 118-122.

Miyamoto K, Tatsumi S, Segawa H, Morita K, Nii T, et al. 1999 Regulation of PiT-1, a

sodium-dependent phosphate co-transporter in rat parathyroid glands Nephrol

Dial Transplant 14 Suppl 1 73-75.

Murer H, Lotscher M, Kaissling B, Levi M, Kempson SA, et al. 1996 Renal brush

border membrane Na/Pi-cotransport: molecular aspects in PTH-dependent and

dietary regulation Kidney Int 49 1769-1773.

Nakatani T, Sarraj B, Ohnishi M, Densmore MJ, Taguchi T, et al. 2009 In vivo genetic

evidence for klotho-dependent, fibroblast growth factor 23 (Fgf23) -mediated

regulation of systemic phosphate homeostasis FASEB J 23 433-441.

Nechama M, Ben-Dov IZ, Briata P, Gherzi R & Naveh-Many T 2008 The mRNA

decay promoting factor K-homology splicing regulator protein post-

transcriptionally determines parathyroid hormone mRNA levels FASEB J 22

3458-3468.

Neher E & Sakmann B 1976 Single-channel currents recorded from membrane of

denervated frog muscle fibres Nature 260 799-802.

Page 58: New insights in parathyroid hormone secretion

58

Neher E, Sakmann B & Steinbach JH 1978 The extracellular patch clamp: a method for

resolving currents through individual open channels in biological membranes

Pflugers Arch 375 219-228.

Nemere I & Larsson D 2002 Does PTH have a direct effect on intestine? J Cell

Biochem 86 29-34.

Nemeth EF & Kosz LM 1989 Adenine nucleotides mobilize cellular Ca2+ and inhibit

parathyroid hormone secretion Am J Physiol 257 E505-513.

Nemeth EF & Scarpa A 1987 Rapid mobilization of cellular Ca2+ in bovine

parathyroid cells evoked by extracellular divalent cations. Evidence for a cell

surface calcium receptor J Biol Chem 262 5188-5196.

Niki I, Yokokura H, Sudo T, Kato M & Hidaka H 1996 Ca2+ signaling and

intracellular Ca2+ binding proteins J Biochem 120 685-698.

Nilsson IL, Yin L, Lundgren E, Rastad J & Ekbom A 2002 Clinical presentation of

primary hyperparathyroidism in Europe--nationwide cohort analysis on

mortality from nonmalignant causes J Bone Miner Res 17 Suppl 2 N68-74.

Norman AW 1979 Vitamin D metabolism and calcium absorption Am J Med 67 989-

998.

Oetting M, LeBoff MS, Levy S, Swiston L, Preston J, et al. 1987 Permeabilization

reveals classical stimulus-secretion coupling in bovine parathyroid cells

Endocrinology 121 1571-1576.

Oldham SB, Lipson LG & Tietjen GE 1982 Presence of calmodulin in parathyroid

adenomas Miner Electrolyte Metab 7 273-280.

Ong HL, Cheng KT, Liu X, Bandyopadhyay BC, Paria BC, et al. 2007 Dynamic

assembly of TRPC1-STIM1-Orai1 ternary complex is involved in store-

operated calcium influx. Evidence for similarities in store-operated and calcium

release-activated calcium channel components J Biol Chem 282 9105-9116.

Palanisamy N, Imanishi Y, Rao PH, Tahara H, Chaganti RS, et al. 1998 Novel

chromosomal abnormalities identified by comparative genomic hybridization in

parathyroid adenomas J Clin Endocrinol Metab 83 1766-1770.

Palmer M, Jakobsson S, Akerstrom G & Ljunghall S 1988 Prevalence of

hypercalcaemia in a health survey: a 14-year follow-up study of serum calcium

values Eur J Clin Invest 18 39-46.

Peacock M, Bilezikian JP, Klassen PS, Guo MD, Turner SA, et al. 2005 Cinacalcet

hydrochloride maintains long-term normocalcemia in patients with primary

hyperparathyroidism J Clin Endocrinol Metab 90 135-141.

Page 59: New insights in parathyroid hormone secretion

59

Pedersen SF, Owsianik G & Nilius B 2005 TRP channels: an overview Cell Calcium

38 233-252.

Pool EH 1907 II. Tetany Parathyreopriva: A Case Report, with a Brief Discussion of

the Disease and of the Parathyroid Glands Ann Surg 46 507-540.

Potier M & Trebak M 2008 New developments in the signaling mechanisms of the

store-operated calcium entry pathway Pflugers Arch 457 405-415.

Potts JT 2005 Parathyroid hormone: past and present J Endocrinol 187 311-325.

Qin L, Raggatt LJ & Partridge NC 2004 Parathyroid hormone: a double-edged sword

for bone metabolism Trends Endocrinol Metab 15 60-65.

Rhys Evans PH, Montgomer PQ & Gullane PJ 2004. Principles and practice of head

and neck oncology. London, Martin Dunitz, First edition.

Ritter CS, Armbrecht HJ, Slatopolsky E & Brown AJ 2006 25-Hydroxyvitamin D(3)

suppresses PTH synthesis and secretion by bovine parathyroid cells Kidney Int

70 654-659.

Rudberg C, Grimelius L, Johansson H, Ljunghall S, Odselius R, et al. 1986 Alteration

in density, morphology and parathyroid hormone release of dispersed

parathyroid cells from patients with hyperparathyroidism Acta Pathol Microbiol

Immunol Scand A 94 253-261.

Salido GM, Sage SO & Rosado JA 2009 TRPC channels and store-operated Ca(2+)

entry Biochim Biophys Acta 1793 223-230.

Saxe A, Gibson G & Silveira E 1995 Effects of long-term lithium infusion on normal

parathyroid tissue Surgery 117 577-580.

Schipani E, Karga H, Karaplis AC, Potts JT, Jr., Kronenberg HM, et al. 1993 Identical

complementary deoxyribonucleic acids encode a human renal and bone

parathyroid hormone (PTH)/PTH-related peptide receptor Endocrinology 132

2157-2165.

Schluter KD 1999 PTH and PTHrP: Similar Structures but Different Functions News

Physiol Sci 14 243-249.

Sela-Brown A, Silver J, Brewer G & Naveh-Many T 2000 Identification of AUF1 as a

parathyroid hormone mRNA 3'-untranslated region-binding protein that

determines parathyroid hormone mRNA stability J Biol Chem 275 7424-7429.

Seriwatanachai D, Thongchote K, Charoenphandhu N, Pandaranandaka J, Tudpor K, et

al. 2008 Prolactin directly enhances bone turnover by raising osteoblast-

expressed receptor activator of nuclear factor kappaB ligand/osteoprotegerin

ratio Bone 42 535-546.

Page 60: New insights in parathyroid hormone secretion

60

Setoguti T, Inoue Y & Kato K 1981 Electron-microscopic studies on the relationship

between the frequency of parathyroid storage granules and serum calcium

levels in the rat Cell Tissue Res 219 457-467.

Setoguti T, Inoue Y & Shin M 1988 Electron-microscopic studies on the threshold

value of calcium concentration for the release of storage granules and the

acceleration of their degradation in the rat parathyroid gland Cell Tissue Res

251 531-539.

Shepardson HC 1927 Endocrinology: DO All Forms of Tetany Depend on a

Parathyroid-Calcium Disturbance? Cal West Med 26 364.

Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, et al. 2004 Targeted

ablation of Fgf23 demonstrates an essential physiological role of FGF23 in

phosphate and vitamin D metabolism J Clin Invest 113 561-568.

Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, et al. 2001 Cloning and

characterization of FGF23 as a causative factor of tumor-induced osteomalacia

Proc Natl Acad Sci U S A 98 6500-6505.

Shimada T, Muto T, Urakawa I, Yoneya T, Yamazaki Y, et al. 2002 Mutant FGF-23

responsible for autosomal dominant hypophosphatemic rickets is resistant to

proteolytic cleavage and causes hypophosphatemia in vivo Endocrinology 143

3179-3182.

Shoback D, Chen TH, Pratt S & Lattyak B 1995 Thapsigargin stimulates intracellular

calcium mobilization and inhibits parathyroid hormone release J Bone Miner

Res 10 743-750.

Shoback D, Thatcher J, Leombruno R & Brown E 1983 Effects of extracellular Ca++

and Mg++ on cytosolic Ca++ and PTH release in dispersed bovine parathyroid

cells Endocrinology 113 424-426.

Shoback DM, Bilezikian JP, Turner SA, McCary LC, Guo MD, et al. 2003 The

calcimimetic cinacalcet normalizes serum calcium in subjects with primary

hyperparathyroidism J Clin Endocrinol Metab 88 5644-5649.

Shoback DM & Brown EM 1984a Forskolin increases cellular cyclic adenosine

monophosphate content and parathyroid hormone release in dispersed bovine

parathyroid cells Metabolism 33 509-514.

Shoback DM & Brown EM 1984b PTH release stimulated by high extracellular

potassium is associated with a decrease in cytosolic calcium in bovine

parathyroid cells Biochem Biophys Res Commun 123 684-690.

Page 61: New insights in parathyroid hormone secretion

61

Shoback DM, Membreno LA & McGhee JG 1988 High calcium and other divalent

cations increase inositol trisphosphate in bovine parathyroid cells

Endocrinology 123 382-389.

Shoback DM, Thatcher J, Leombruno R & Brown EM 1984 Relationship between

parathyroid hormone secretion and cytosolic calcium concentration in dispersed

bovine parathyroid cells Proc Natl Acad Sci U S A 81 3113-3117.

Sloand JA & Shelly MA 2006 Normalization of lithium-induced hypercalcemia and

hyperparathyroidism with cinacalcet hydrochloride Am J Kidney Dis 48 832-

837.

Soboloff J, Spassova MA, Tang XD, Hewavitharana T, Xu W, et al. 2006 Orai1 and

STIM reconstitute store-operated calcium channel function J Biol Chem 281

20661-20665.

Stewart CP & Percival GH 1927 Studies on Calcium Metabolism: The Action of the

Parathyroid Hormone on the Calcium Content of the Serum and on the

Absorption and Excretion of Calcium Biochem J 21 301-313.

Takahashi A, Camacho P, Lechleiter JD & Herman B 1999 Measurement of

intracellular calcium Physiol Rev 79 1089-1125.

Takatsuki K, Hanley DA & Sherwood LM 1980 Effects of magnesium ion on

parathyroid hormone secretion in vitro Calcif Tissue Int 32 201-206.

Tanaka Y, Funahashi H, Imai T, Seo H, Tominaga Y, et al. 1996 Oxyphil cell function

in secondary parathyroid hyperplasia Nephron 73 580-586.

Tang J, Maximov A, Shin OH, Dai H, Rizo J, et al. 2006 A complexin/synaptotagmin 1

switch controls fast synaptic vesicle exocytosis Cell 126 1175-1187.

Tatsumi S, Segawa H, Morita K, Haga H, Kouda T, et al. 1998 Molecular cloning and

hormonal regulation of PiT-1, a sodium-dependent phosphate cotransporter

from rat parathyroid glands Endocrinology 139 1692-1699.

Tebben PJ, Singh RJ, Clarke BL & Kumar R 2004 Fibroblast growth factor 23,

parathyroid hormone, and 1alpha,25-dihydroxyvitamin D in surgically treated

primary hyperparathyroidism Mayo Clin Proc 79 1508-1513.

Tokumoto M, Taniguchi M, Matsuo D, Tsuruya K, Hirakata H, et al. 2005 Parathyroid

cell growth in patients with advanced secondary hyperparathyroidism: vitamin

D receptor, calcium sensing receptor, and cell cycle regulating factors Ther

Apher Dial 9 Suppl 1 S27-34.

Torres PU 2006 Cinacalcet HCl: a novel treatment for secondary hyperparathyroidism

caused by chronic kidney disease J Ren Nutr 16 253-258.

Page 62: New insights in parathyroid hormone secretion

62

Tsien RY, Rink TJ & Poenie M 1985 Measurement of cytosolic free Ca2+ in individual

small cells using fluorescence microscopy with dual excitation wavelengths

Cell Calcium 6 145-157.

Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, et al. 2006 Klotho

converts canonical FGF receptor into a specific receptor for FGF23 Nature 444

770-774.

Valimaki S, Hoog A, Larsson C, Farnebo LO & Branstrom R 2003 High extracellular

Ca2+ hyperpolarizes human parathyroid cells via Ca(2+)-activated K+ channels

J Biol Chem 278 49685-49690.

Vincent S 1904 Some observations upon the functions of the thyroid and parathyroid

glands J Physiol 32 65-86.

Wallace J & Scarpa A 1982 Regulation of parathyroid hormone secretion in vitro by

divalent cations and cellular metabolism J Biol Chem 257 10613-10616.

Wallace J & Scarpa A 1983 Similarities of Li+ and low Ca2+ in the modulation of

secretion by parathyroid cells in vitro J Biol Chem 258 6288-6292.

Wallfelt C, Larsson R, Johansson H, Rastad J, Akerstrom G, et al. 1985 Stimulus-

secretion coupling of parathyroid hormone release: studies of 45Ca and 86Rb

fluxes Acta Physiol Scand 124 239-245.

Wang ZW 2008 Regulation of synaptic transmission by presynaptic CaMKII and BK

channels Mol Neurobiol 38 153-166.

Willard DP 1935 RELATIONSHIP OF THE PARATHYROID GLAND TO

CALCIUM METABOLISM Ann Surg 102 351-356.

Wong C, Lai T, Hilly JM, Stewart CE & Farndon JR 2002 Selective estrogen receptor

modulators inhibit the effects of insulin-like growth factors in

hyperparathyroidism Surgery 132 998-1006; discussion 1006-1007.

Yano S, Sugimoto T, Tsukamoto T, Chihara K, Kobayashi A, et al. 2000 Association

of decreased calcium-sensing receptor expression with proliferation of

parathyroid cells in secondary hyperparathyroidism Kidney Int 58 1980-1986.

Yokoyama K, Matsuba D, Adachi-Akahane S, Takeyama H, Tabei I, et al. 2009

Dihydropyridine- and voltage-sensitive Ca2+ entry in human parathyroid cells

Exp Physiol

Zhang SL, Yu Y, Roos J, Kozak JA, Deerinck TJ, et al. 2005 STIM1 is a Ca2+ sensor

that activates CRAC channels and migrates from the Ca2+ store to the plasma

membrane Nature 437 902-905.

Page 63: New insights in parathyroid hormone secretion

63

Zhou W, Katz MH, Deftos LJ, Snyder CS, Baird S, et al. 2003 Metachronous double

parathyroid adenomas involving two different cell types: chief cell and oxyphil

cell Endocr Pract 9 522-525.

Zofkova I, Rojdmark S & Kancheva RL 1993 Does estrogen replacement therapy

influence parathyroid hormone responsiveness to exogenous hypercalcemia in

postmenopausal women? J Endocrinol Invest 16 323-327.


Recommended