JPS6118722A - Stimulator for defensive ability of living body - Google Patents

Stimulator for defensive ability of living body

Info

Publication number
JPS6118722A
JPS6118722A JP59138023A JP13802384A JPS6118722A JP S6118722 A JPS6118722 A JP S6118722A JP 59138023 A JP59138023 A JP 59138023A JP 13802384 A JP13802384 A JP 13802384A JP S6118722 A JPS6118722 A JP S6118722A
Authority
JP
Japan
Prior art keywords
ability
substance
biological defense
ginseng
polysaccharide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
JP59138023A
Other languages
Japanese (ja)
Inventor
Shunzo Hatono
鳩野 俊三
Toru Fuwa
不破 亨
Kenji Mizutani
健二 水谷
Ryoji Kasai
笠井 良次
Osamu Tanaka
治 田中
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wakunaga Pharmaceutical Co Ltd
Original Assignee
Wakunaga Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wakunaga Pharmaceutical Co Ltd filed Critical Wakunaga Pharmaceutical Co Ltd
Priority to JP59138023A priority Critical patent/JPS6118722A/en
Publication of JPS6118722A publication Critical patent/JPS6118722A/en
Pending legal-status Critical Current

Links

Abstract

PURPOSE:The titled preparation that contains, as an active ingredient, polysaccharides isolated from ginseng. CONSTITUTION:Polysaccharides, preferably containing at least one monosaccharide selected from arabinose, galactose, and glucose and having more than 100,000mol.wt., which have been isolated from ginseng, are used to give the objective preparation stimulating biological defensive ability such as functional stimulation of reticuloendothelial system, resistance to microbism and antitumor ability. The preparation is in the form of dust, granules, tablets, capsules, syrup or injection solution and is given orally or parenterally. The dose is 0.1-1,000 mg/day/man based on the polysaccharide in oral administration once a day or in portions. The polysaccharide is extracted from genseng with water and the extract is precipitated by addition of an organic solvent. Then, the precipitate is subjected to chromatography with an ion-exchange resin or gel filtration agent.

Description

【発明の詳細な説明】 発明の背景 本発明は、薬用ニンジンから単離されたその多糖画分を
有効成分とする生体防御能亢進剤に関する。
DETAILED DESCRIPTION OF THE INVENTION BACKGROUND OF THE INVENTION The present invention relates to a biodefense enhancer containing a polysaccharide fraction isolated from medicinal ginseng as an active ingredient.

薬用ニンジンの中でもとりわけオタネニンジン(Pan
ax ginseng C,A、 MEYER)は朝鮮
二ノジンとも呼ばれ、古来よシ強壮、強精、消炎、利尿
、抗糖尿病用の薬剤として用いられてきたことは周知の
ことである。同じく薬用ニンジンの一種である人参三七
(Panax notoglnseng F、 H,C
HEN )は、その乾燥した塊根を田七と称し、田七は
古くから止血、消腫、鎮痛、消炎系として用いられ、近
年特に中国などで滋養強壮薬として供されているもので
ある。
Among medicinal ginseng, Panax ginseng (Pan ginseng)
Ax ginseng C, A, MEYER) is also called Korean Ninojin, and it is well known that it has been used since ancient times as a tonic, tonic, anti-inflammatory, diuretic, and antidiabetic drug. Panax notoglnseng (Panax notoglnseng F, H, C) is also a type of medicinal ginseng.
The dried tuberous root of HEN) is called Tashichi, and Tashichi has been used since ancient times as a hemostatic, antitumor, analgesic, and anti-inflammatory agent, and in recent years has been served as a tonic and tonic, especially in China.

また、薬用ニンジン中の成分についての薬理作用の報告
については、主にサホこン成分について行なわれ、例え
ば抗糖尿病作用C特開5!;−1、2.272/号公報
など)、抗腫瘍作用(特開外−ざり01、2号公報など
)が報告されている。しかしながら糖タンパク成分、多
糖成分なとにつ謁ての報告はほとんど見当たらず、址だ
葦だ研究の余地を残して−るのが現状である。
In addition, reports on the pharmacological effects of ingredients in medicinal ginseng have mainly been made on the ingredients of ginseng, such as anti-diabetic activity C JP-A-5! ;-1, 2.272/2004, etc.), and antitumor effects (Japanese Patent Application Laid-open No. 01, 2, etc.). However, there are almost no reports on glycoprotein components or polysaccharide components, and the current situation is that there is still room for further research.

ところで、人聞は一般に老令になると諸疾患に対する抵
抗能が低下して柚りの病気に罹患しゃすくなる。原因と
しては、体内諸器官や種々の代謝作用が衰えるとともに
異物や老廃物の処理作用のある食細胞の機能が低下し、
生体防御能が衰えることが考えられる。
By the way, as people grow older, their ability to resist various diseases generally decreases, making them more susceptible to yuzu diseases. The cause is a decline in various internal organs and metabolic processes, as well as a decline in the function of phagocytes, which process foreign substances and waste.
It is thought that the body's defense ability declines.

本発明者は薬用ニンジンの成分について研究を重ねた結
果、薬用ニンジン中の多糖画分に生体防御能があること
を発見して本発明を完成するに到った。
As a result of repeated research on the components of medicinal ginseng, the present inventor discovered that the polysaccharide fraction in medicinal ginseng has a biological defense ability, thereby completing the present invention.

発明の概要 本発明は新規な生体防御能亢進剤の提供を目的とし、薬
用ニンジン中の多糖成分がこの作用を有すると込う当業
者にとっても思わぬ発見に基づくものである。
SUMMARY OF THE INVENTION The present invention aims to provide a novel biological defense ability enhancing agent, and is based on the unexpected discovery even for those skilled in the art that the polysaccharide component in medicinal ginseng has this effect.

したがって、本発明による生体防御能亢進剤は、単離さ
れた薬用ニンジンの多糖成分を有効成分とすること、を
特徴とするものである。
Therefore, the biological defense ability enhancer according to the present invention is characterized in that the active ingredient is a polysaccharide component of isolated medicinal ginseng.

そして、本発明による生体防御能亢進剤の提供は言うま
でもなく諸疾患対策に有意義な貢献をなすものである。
Needless to say, the provision of the biological defense enhancer according to the present invention will make a significant contribution to measures against various diseases.

発明の詳細な説明 薬用ニンジン 本発明でいう薬用ニンジンとは、ウコギ科(Arall
aceae )、バナ・ソクス属(Panax )に属
する植物を指し、オタネニンジン、人参三七、アメリカ
ニンジン(Panax qulnquafollum 
LINNE )、チクセツ=yシフ (Panax j
aponlcua C,A、 MEYER)などがこれ
にあたる。多糖成分を取得すべき部分はとりわけ根部が
好壕しく、取得効率を考えれば乾燥したものを用いるの
が好都合である。また、これらのパナ・・クス属植物を
常法によって組織培養に付し、その培養物を用いること
もできる。
DETAILED DESCRIPTION OF THE INVENTION Medicinal ginseng The medicinal ginseng as used in the present invention refers to Aralliaceae (Arallaceae).
aceae), refers to plants belonging to the genus Panax, including Panax ginseng, Panax ginseng, and American ginseng (Panax qulnquafollum).
LINNE), Chikusetsu=y Schiff (Panax j
Examples include aponlcua C, A, MEYER). The root part is particularly favorable for obtaining the polysaccharide component, and in terms of obtaining efficiency, it is convenient to use the dry part. It is also possible to subject these plants of the genus Panacus to tissue culture by a conventional method and use the resulting culture.

多糖成分およびその取得方法 本発明で用層る多糖成分け、分子量的に不均一な多糖そ
のものあるいは実質的に多糖成分から成るものである。
Polysaccharide component and method for obtaining the same The polysaccharide component used in the present invention is a polysaccharide itself or substantially composed of a polysaccharide component having a nonuniform molecular weight.

したがって構成糖残基の種類や組成比、平均分子量、構
造などは、薬用ニンジン由来の多糖であれば任意のもの
を用いることができるが、好ましくは、アラビノース(
arablnoae )、ガラクトース(galact
ose )およびグルコース(gluaoa@ )  
から選ばれる少なくとも7種の糖を単位構成成分として
含む、分子量lO万以上の多糖成分がよい。
Therefore, any type of constituent sugar residue, composition ratio, average molecular weight, structure, etc. can be used as long as it is a polysaccharide derived from medicinal ginseng, but preferably arabinose (
arablnoae), galactose
ose ) and glucose (gluaoa@)
A polysaccharide component having a molecular weight of 10,000 or more and containing at least seven types of sugars selected from the following as unit components is preferable.

なお、本発明でいう多糖とは通常の概念にしたがうもの
であり、すなわち、分子量的に不均一で平均重合度が1
00をこえる、直鎖状または分枝状の巨大分子をさす。
Note that the polysaccharide used in the present invention follows the usual concept, that is, it has a nonuniform molecular weight and an average degree of polymerization of 1.
Refers to linear or branched macromolecules exceeding 000.

本発明で用いる多糖成分を取得するためにはニンジンか
ら該成分を単離することのできる任意の方法を用−八れ
ばよい。すなわち薬用ニンジンを好ましくは破砕後、水
もしくは水溶性有機溶媒に浸漬して抽出を行ない、有機
溶媒を加えて沈殿物を得ろ。そして沈殿物をイオン交換
樹脂もしくはゲルf過剤を用いたクロマトグラフィーに
付し、目的物を得ることができる。原料ニンジンは脱脂
操作に付してもよく、筐た上記沈殿物を透析に付しても
目的物を得ることができよう。
In order to obtain the polysaccharide component used in the present invention, any method capable of isolating the component from carrots may be used. That is, medicinal carrots are preferably crushed, then extracted by immersing them in water or a water-soluble organic solvent, and the organic solvent is added to obtain a precipitate. The desired product can then be obtained by subjecting the precipitate to chromatography using an ion exchange resin or gel filter. The raw material carrot may be subjected to a defatting operation, and the desired product may also be obtained by subjecting the precipitate in a casing to dialysis.

生体防御能亢進剤 本発明の生体防御能亢進剤は薬用ニンジンの多糖成分そ
れ自体または適宜製剤用の賦形剤、結合剤、希釈剤と混
合して成るものであシ、粉末、顆粒、錠剤、カプセル剤
、シロ・ツブ剤、注射剤などの形態で経口的または非経
口的に投与することができる。また、必要に応じて他の
薬剤を調合させてもよい。投与量は、年令、体重、症状
によシ適宜増減するが、経口的には通常成人、7日、多
糖成分として0./〜1ooott5tが望筐し10好
まし論具体例は、薬用ニンジンの多糖成分と製剤上の補
助成分とからなるものである。筐だ、本発明の他の好ま
しい具体例は、上記1日当たりの投与量を7回な−し数
回に分けて服用させるための単位投与形態のものである
Biodefense Enhancing Agent The biodefense enhancing agent of the present invention may be composed of the polysaccharide component of medicinal ginseng itself or mixed with an appropriate excipient, binder, or diluent for formulation, such as powder, granules, or tablets. It can be administered orally or parenterally in the form of capsules, tablets, injections, etc. Further, other drugs may be mixed as necessary. The dosage may be adjusted depending on age, body weight, and symptoms, but for oral administration, it is usually administered to an adult for 7 days, at a dosage of 0.00% as a polysaccharide component. /~1ooott5t is desired and 10 A preferred example is one consisting of a polysaccharide component of medicinal ginseng and an auxiliary component in the formulation. Another preferred embodiment of the present invention is a unit dosage form for administering the above-mentioned daily dose in seven or more divided doses.

なお、本発明における多糖成分の毒性は、例えば、後記
実験例における物質Bの場合、ICR系雄性マウス(!
匹)に対する。2 o o tru)/Kpの静脈投与
で死亡例が認められなかったこと、および物質Bと構造
類似であるレンチナン(木村郁部ほか、最新医学、3乙
、70ど1.t(/りと/))およびPS−に(クレス
チン(三共))等が周知の通り低毒性であることによシ
、一般に低いものと考えることができる。
In addition, the toxicity of the polysaccharide component in the present invention is, for example, in the case of substance B in the experimental example described later, in ICR male mice (!
against). No deaths were observed with intravenous administration of 2 o o tru)/Kp, and lentinan, which is structurally similar to substance B (Kimura Ikube et al., Modern Igaku, 3 otsu, 70 do 1.t (/rito /)) and PS- (Krestin (Sankyo)), etc., are generally considered to be low in toxicity, as they are known to have low toxicity.

生体防御能亢進作用 生体に異物(細菌、ウィルス、等)が侵入すると、これ
に対してマクロファージの賞食作用、リンパ球の分化に
よる抗体産生作用等の生体防御反応が現われ、外敵を排
除する巧妙な機構が生じてくる。しかし、免疫低下また
け防御因子低下時には、攻撃因子(異物)優勢となって
、日和見感染等の各種疾患に罹シやすくなる。
Enhancement of biological defense ability When foreign substances (bacteria, viruses, etc.) invade the living body, biological defense reactions such as phagocytosis by macrophages and antibody production by differentiation of lymphocytes appear to eliminate foreign invaders. A mechanism emerges. However, when the immune system is weakened and protective factors are decreased, aggressive factors (foreign substances) become dominant, making the body susceptible to various diseases such as opportunistic infections.

本発明でいう生体防御能亢進作用は、上記に示した様な
生体防御反応を活性化して、異物を生体から排除する作
用をいい、二次的な抗ウィルス、抗腫瘍、抗寄生虫、抗
細菌作用およびその他の外来性抗原の排除に関するもの
である。
In the present invention, the action of enhancing biological defense function refers to the action of activating the biological defense reactions shown above to eliminate foreign substances from the living body, and has secondary antiviral, antitumor, antiparasitic, and antiviral effects. It concerns the elimination of bacterial action and other foreign antigens.

実験例 1)多糖成分の取得 三七人参、2に2に水jす・ソトルを加え、温浸して得
られる抽出物を減圧下で濃縮し、得られた残渣(収率/
と1件1を適量の水に溶かしたのちとれに≠倍量のエタ
ノールを加え、得られる沈殿物を73、gチの収率で取
得した。
Experimental Example 1) Obtaining polysaccharide components Panax ginseng was added to 2 with water and sotol, and the extract obtained by digestion was concentrated under reduced pressure, and the resulting residue (yield/
After dissolving 1 and 1 in an appropriate amount of water, ≠ times the amount of ethanol was added to the solution, and the resulting precipitate was obtained at a yield of 73.g.

そのエタノール沈殿物をセファデーIクスG−50(フ
ァルマシア)を充填したカラム(21rga X Io
otrrm)に付して分離し、水によシ溶出される画分
を得た(収率1、/%)。
The ethanol precipitate was transferred to a column (21rga
otrrm) to obtain a fraction eluted with water (yield: 1/%).

次にその画分をDEAE−TOYOPEARL  l、
10M(東洋曹達)を充填したカラム(、z、rmmx
♂oomri)に付し、0.02M−NH11HCO5
により溶出させて、物質Aを得た(収率θ、≠jチ)。
Then, the fraction was transferred to DEAE-TOYOPEARL,
Column packed with 10M (Toyo Soda) (, z, rmmx
0.02M-NH11HCO5
By elution, substance A was obtained (yield θ, ≠j).

物質Aは、更に分取用TSKゲルtaooOPWG(東
洋曹達)を充填したカラA (,2Jtmax6oor
n )に付し、水により溶出させて、単一な物質Bを得
た(収率O1,2♂%)。
Substance A was further filled with preparative TSK gel taooOPWG (Toyo Soda).
n) and eluted with water to obtain a single substance B (yield: O1.2%).

λ)物質Bの性状及び構造 物質Bけ、高速液体クロマトグラフィー(HLC)によ
る(分析用)TSKゲル≠ooopwおよび6θ00P
W(東洋曹達)を用いたゲル沢過分析にお−て単一であ
り、紫外線(UV)吸収はなく、白色粉末であシ、比旋
光度(〔α〕君0=+/j3.00(C= o、りj、
H2O)である。1だ、光散乱法による分子量の測定に
おいて1.≠≠×706  を示した物質Bの/N−H
2S0I加水分解物のトリメチルシリルエステル(TM
S)化体のガスクロマトグラフィー(GLC)において
アラビノース、ガラクトースおよびグルコースが検出さ
れ、ピーク面積比より、その生成比はそれぞれo、t 
: r、o : yti、zであった。
λ) Properties and structure of substance B, TSK gel (for analysis) by high performance liquid chromatography (HLC)≠ooopw and 6θ00P
Gel permeability analysis using W (Toyo Soda) showed that it was single, had no ultraviolet (UV) absorption, was a white powder, and had a specific optical rotation ([α]kun0=+/j3.00). (C= o, rij,
H2O). 1. In the measurement of molecular weight by light scattering method. /NH of substance B showing ≠≠×706
Trimethylsilyl ester of 2S0I hydrolyzate (TM
Arabinose, galactose, and glucose were detected in gas chromatography (GLC) of the S) compound, and from the peak area ratio, the production ratio was o and t, respectively.
: r, o : yti, z.

/3 C−NMR(A7.、!’ M出、洋:p2o)
にお込では第5図に示したように、α−1、り グリカ
ンを主鎖とし、α−1、乙に分枝したグリコーゲン様の
シグナル(Photls Daia 、 etal、、
  100. /QJ(/りr2))の他、末端α−ア
ラビノフラノースに帰属されるシグナル(C1:δ=/
/θ、1、C2:δ=ざλ、、z、c5:δ= 77、
G 、 C11:δ=ざ弘、7.C5:δ= g1、z
 )などが認められた。
/3 C-NMR (A7.,!'M output, Western: p2o)
As shown in Figure 5, in Nioki, a glycogen-like signal with α-1, glycan as the main chain and branches to α-1, Otsu (Photols Daiia et al.
100. /QJ (/ri r2)), a signal assigned to terminal α-arabinofuranose (C1:δ=/
/θ,1,C2:δ=zaλ,,z,c5:δ=77,
G, C11: δ=Zahiro, 7. C5: δ= g1, z
) etc. were recognized.

3)物質Bのカーボンクリアランス(Carbon c
le−11ranee )能活性効果 (1)  実験動物 ICR系雄性マウスを一定期間飼育後、健康と思われる
j週齢のものを/群/θ匹として本実験に使用した。
3) Carbon clearance of substance B (Carbon c
le-11ranee) Functional activity effect (1) Experimental animals ICR male mice were raised for a certain period of time, and then healthy J-week-old mice/group/θ mice were used in this experiment.

(2)実験方法 ハルパーン(Halpern )  らのカーボン・ク
リアランス法(Halpern 、 B、 H,、et
al 、、 J、 Retjculo−endothe
l、Soc 、 / 、 77 (/り6≠))に従い
、上記のマウスに下記に示す濃度の被検液を腹腔内投与
し1時間後にカーボン懸濁液(ペリカン・インク(ペリ
カン社製)を生理食塩水で≠倍希釈したもの)をr m
l / KPの割合で、尾静脈投与した。2分および7
0分後にマウスの眼窩からマイクロピベーノトを用いて
一、20μノの血液を採取した。
(2) Experimental method Carbon clearance method of Halpern et al.
al., J. Retjculo-endothe
1, Soc, /, 77 (/ri6≠)), the test solution at the concentration shown below was intraperitoneally administered to the above mice, and 1 hour later, a carbon suspension (Pelican Inc. (manufactured by Pelican)) was administered to the above mice. diluted with physiological saline) to r m
It was administered via the tail vein at a ratio of l/KP. 2 minutes and 7
After 0 minutes, 1.20 μm of blood was collected from the eye socket of the mouse using a micropive tube.

得られた血液にo、i%Na2CO3溶液rmlを加え
て溶血させた後、分光光度計(高車製作所)を用いて6
7よnmにおける吸光度(OD6r5)を測定して、カ
ーボン懸濁液の血中消失宋速度を求めた。
After hemolyzing the obtained blood by adding o, i% Na2CO3 solution rml, it was measured using a spectrophotometer (Takashuma Seisakusho).
The absorbance at 7 nm (OD6r5) was measured to determine the rate at which the carbon suspension disappeared from the blood.

(被検液の投与量) げ)生理食塩水(対照)    j属/ KP(ロ)物
質B   10 try / Ky−+ 、2 fRy
/ml (jml/に2)e→  ・   ユ0ツ/今
→tグ/ml (・  )←二)     〜    
   弘O竺り/KP−+ざ 竺り/mJ  (・  
   )なお、結果の統計的処理はステー−プント(S
tudent’ a )  を検定にて行なった。
(Dosage of test solution) G) Physiological saline (control) Genus J/KP (B) Substance B 10 try/Ky-+, 2 fRy
/ml (jml/に2)e→・yu0tsu/now→tgu/ml (・)←2)~
Hiroo silk/KP-+Za silk/mJ (・
) The statistical processing of the results was performed using Sta-Punt (S
student'a) was performed in the assay.

(3)  実験結果 物質Bの投与量とカーボン懸濁液の消失速度との関係は
、第1図に示す通りであった。なお、図面中の棒グラフ
の上部に記しである値は、カーボン懸濁液の消失速度(
X10’ rnin−” )を示す。
(3) Experimental results The relationship between the dose of substance B and the rate of disappearance of the carbon suspension was as shown in FIG. In addition, the value written above the bar graph in the drawing is the disappearance rate of the carbon suspension (
X10'rnin-'').

物質Bの投与により、用量依存的にカーボンクリアラン
ス能の増強が認められ、中でもqo my / Kp投
与群においては顕著な効果が得られた。
By administering substance B, enhancement of carbon clearance ability was observed in a dose-dependent manner, and particularly remarkable effects were obtained in the qomy/Kp administration group.

≠)物質Aの感染抵抗性の増強効果 (1)実験動物 ICR系雄性マウスを一定期間動物舎内で飼育後、健康
と思われる夕週齢のものを7群6匹として本実験に使用
した。
≠) Enhancement effect of substance A on infection resistance (1) Experimental animals ICR male mice were kept in an animal house for a certain period of time, and then 7 groups of 6 mice, which appeared to be healthy, were used in this experiment. .

(2)  実験方法 上記マウスに下記に示す濃度の被検液を腹腔内投与し、
評時間後に尿路感染症患者よシ単離したE、 coil
  0U−00り株(r X 10Bcell /ml
 ) 0. / rttlをマウス背部皮下に接種して
3時間毎に!日間生死を判定した。
(2) Experimental method A test solution with the concentration shown below was administered intraperitoneally to the above mice.
E, coil isolated from patients with urinary tract infection after hours of treatment.
0U-00 strain (r x 10Bcell/ml
) 0. / Inoculate rttl subcutaneously on the back of mice every 3 hours! It was determined whether the animal was alive or dead.

(被検液の投与量) ←)生理食塩水(対照)   夕m/ / Kp(ロ)
物質A、2、夕15’/KP−+0. !My/ml 
(rrrtl/に9 )ヒ”)   ”    ’O”
i’/Kp−+、21ny/d(tm//Kp)←) 
  −tpotq/Kp−+rmy/ml (rml/
Kp )(3)実験結果 に、co11感染マウスに対する物質Aの経時的な生存
率は、第2図に示す通シであった。なお、グラフ中の(
)内の値はマウス6匹中の生存数を示す。
(Dosage of test solution) ←) Physiological saline (control) m/ / Kp (b)
Substance A, 2, 15'/KP-+0. ! My/ml
(rrrtl/ni9)hi") "'O"
i'/Kp-+, 21ny/d(tm//Kp)←)
-tpotq/Kp-+rmy/ml (rml/
Kp) (3) As for the experimental results, the survival rate of Substance A over time for co11-infected mice was consistent with that shown in FIG. In addition, in the graph (
Values in ) indicate the number of survivors out of 6 mice.

物質Aの投与によシ用量依存的に延線効果が認められ、
このうち弘oq/Kp投与群では、評時間後に7例死亡
したのみで、残り5例は、j日後においても生存して−
だ。
Administration of substance A caused a dose-dependent effect on line extension,
Of these, in the Hiroq/Kp administration group, only 7 cases died after the evaluation time, and the remaining 5 cases remained alive even after j days.
is.

り物質Bの抗体産生能に及ぼす影響 (1)実験動物 ICR系雄性マウスを一定期間動物舎内で飼育後、健康
と思われるj週齢のものを/群J〜7匹として本実験に
使用した。
Effect of Substance B on Antibody Production Ability (1) Experimental Animals After raising ICR male mice in an animal house for a certain period of time, J-week-old mice that appeared to be healthy were used in this experiment as 7 mice per group J. did.

(2)実験方法 シャーン(Jerne )らのプラーク法(Jerne
、 N。
(2) Experimental method Jerne et al.'s plaque method (Jerne et al.
,N.

K、、 etal、、 5cience 、 /IA0
.4t03(7Fl+3) )に従込、上記のマウスに
、/ X 10Bcell/mA’のヒツジ赤血球(以
下5RBCと記す)浮遊液O3/mlを尾静脈投与し感
作させた後、3時間毎に3日間にわたって下記に示す濃
度の被検液を腹腔内投与した。感作弘ローに胸腺を摘出
して、5RBCに対する抗体産生細胞を測定した。
K,, etal,, 5science, /IA0
.. 4t03(7Fl+3)), the above mice were sensitized by administering O3/ml of a sheep red blood cell (hereinafter referred to as 5RBC) suspension at /X 10Bcell/mA' through the tail vein, and then sensitized at 3 hours every 3 hours. A test solution at the concentration shown below was administered intraperitoneally over a period of days. The thymus of the sensitized Hiro was removed and the number of antibody-producing cells against 5RBC was measured.

(被検液の投与量) げ)生理食塩水(対照)   夕ml / )P(ロ)
物質B  O,,2mj//に?−+0−0ググ/rn
l (、r ml /に9 )ヒ→  I  、2グ/
今→Q、≠1・l/ゴ(!ゴ/Kf)に)物質E   
1、oInf?/Kp−+2m9/ml (j ml/
Kp )(ホ)  〃   リ■/ Kp→ざダ/m/
(”   )なお、結果の統計的処理はスチーーデント
(Student’s ) を検定にて行なった。
(Dosage of test solution) G) Physiological saline (control) mL / ) P (B)
Substance B O,,2mj//? -+0-0gugu/rn
l (,r ml /9)hi → I,2g/
Now → Q, ≠1・l/go (!go/Kf)) substance E
1.oInf? /Kp-+2m9/ml (j ml/
Kp ) (e) 〃 ri■/ Kp→zada/m/
('') The results were statistically processed using Student's test.

(3)実験結果 物質Bの投与量と5RBCに対する抗体産生細胞数の関
係は、下表に示す通りであ−た。
(3) Experimental Results The relationship between the dose of substance B and the number of antibody-producing cells against 5RBC was as shown in the table below.

5RBCに対する抗体産生細胞数は、物質Bの投与量と
は逆相関的に増加が認められ、特に0.2m97 Kg
投与群においては、コントロール群の約3倍と顕著な増
加が得られた。
The number of antibody-producing cells against 5RBC was observed to increase inversely with the dose of substance B, especially at 0.2 m97 Kg.
In the treated group, a remarkable increase of about 3 times that in the control group was obtained.

6)物質Bの試験管内マクロファージ活性効果(11実
験方法 BALB/C系雄性マウスの腹腔に、チオグリコンート
培地3−を投与し、3日後にマウスを脱血化させ、腹腔
内に、イーグルMEM培地!Mを注入し、腹腔滲出細胞
(以下PECと記す)を採取した。その後、リン酸緩衝
生理食塩水(以下PBS”と記す)にて洗浄し、toi
牛脂児血清を添加したダルベツコ変法イーグルMEM培
地にて10’Ce1ls / !rLlに調整した。細
胞浮遊液をりtウェルの平底ブv−)に0.2 ml/
we 11ずつ分注しブこ後、3時間1、?7℃/ !
r% CO2の栄件下でも培養し、浮遊細胞を除去した
。更に、PBS−で洗浄し、10チ牛脂児血清添加ダル
ベツコ変法イーグルMEM培地にて、10,30および
100μI/−の濃度に調整した物質Bの被検液を0.
2 mlZwellずつ分注した・φg時間培養後、培
地を20μ!採取し、グルコースC−テストキット(和
光)によ)グルコース含量を測定した。
6) In vitro macrophage activation effect of substance B (11 Experimental method) Thioglyconate medium 3- was administered into the peritoneal cavity of a BALB/C male mouse, and after 3 days, the mouse was exsanguinated, and Eagle's MEM medium was intraperitoneally administered! M was injected, and peritoneal exudate cells (hereinafter referred to as PEC) were collected. Thereafter, they were washed with phosphate buffered saline (hereinafter referred to as "PBS"), and the toi
10'Ce1ls/! in Dulbecco's modified Eagle MEM medium supplemented with tallow serum. Adjusted to rLl. Pour 0.2 ml/cell suspension into the flat-bottom tube of the well.
We dispensed 11 each and 3 hours after dispensing, 1? 7℃/!
The cells were also cultured under r% CO2 to remove floating cells. Furthermore, test solutions of substance B, washed with PBS- and adjusted to concentrations of 10, 30 and 100 μI/- in Dulbecco's modified Eagle MEM medium supplemented with 10 T tallow serum, were added at 0.
After culturing for φg hours, 20μ of the medium was dispensed into each well. The glucose content was measured using a glucose C-test kit (Wako).

なお、結果の統計的処理はステー−プント(Stude
nt’s ) を−検定にて行な、9た。
In addition, statistical processing of the results is performed using Stude
nt's) was carried out using the -test, and 9 was obtained.

(2)実験結果 物質Bの添加量とPECのグルコース消費量との関係は
、下表に示す通りであった。
(2) Experimental Results The relationship between the amount of substance B added and the amount of glucose consumed by PEC was as shown in the table below.

物質Bの添加によシ用量依存的にグルコース消費の促進
が認められ、特に5Op9/rru5、/ 0 (71
6!/lnl添加群では有意な促進が認められた。
Addition of substance B promoted glucose consumption in a dose-dependent manner, especially 5Op9/rru5,/0 (71
6! Significant promotion was observed in the /lnl addition group.

を−検定 *二p<o、oi **:p(0,001 7)物質への抗腫瘍効果 [11実験動物 ICR系雄性マウスを一定期間動物舎内で飼育後、健康
と思われるj退的のものを7群を匹として本実験に使用
した。
- Test *2 p < o, oi **: p (0,001 7) Antitumor effect on substance Seven groups of animals were used in this experiment.

(2)実験方法 上記のマウスの腋窩部皮下に/ X 10BCell/
mlのサルコーマ%lIO細胞浮遊液0./mlを投与
して移植した。移植後、下記に示す濃度の被検液をJ時
間毎に10日間にわたーて腹腔内に投与し、一方では/
週間毎に!週間にわたーてM瘍係数(長径(酎)×短径
(鵡))を測定した。さらに、!週間後に、腫瘍を摘出
し、腫瘍重量を測定した。
(2) Experimental method Subcutaneously in the axillary region of the above mouse/X 10BCell/
ml Sarcoma %lIO cell suspension 0. /ml was administered and transplanted. After transplantation, the test solution with the concentration shown below was administered intraperitoneally every J hours for 10 days, while /
Every week! The M tumor coefficient (major axis (chu) x short axis (mamo)) was measured over a week. moreover,! After a week, the tumor was removed and the tumor weight was measured.

(被検液の投与量) (イ)生理食塩水(対照)    3ml/に?(ロ)
物質A   2.!m9/Kg−*0.!m9/ml(
夕ml/Ky)G”3   ’    10m9/に9
−+、2m9/ml (tt   )に)  //  
  m Tv/に9→gダ/麻(〃)(3)実験結果 物質Aのザルコーマ/10細胞に対する抗腫瘍係数の減
少効果は第3図に示す通シであり、また!週間後の腫瘍
重量は下表に示す通シであった。
(Dosage of test solution) (a) Physiological saline (control) 3ml/? (B)
Substance A 2. ! m9/Kg-*0. ! m9/ml (
Evening ml/Ky) G"3' 10m9/Ni9
-+, 2m9/ml (tt) //
m Tv/ni9→g da/hemp (〃) (3) Experimental results The effect of substance A on reducing the antitumor coefficient on Sarcoma/10 cells is consistent with that shown in Figure 3, and! The tumor weight after a week was as shown in the table below.

物質AをJjm9/Kp投与した群において、λ週目よ
シ腫瘍係数の減少が認められはじめ、≠週目でほぼ退縮
した。更に夕週目には金側完治退縮した。
In the group to which Substance A was administered to Jjm9/Kp, a decrease in the tumor index began to be observed at week λ, and almost regressed at week ≠. Furthermore, in the evening, the gold side was completely healed and regressed.

♂)物質Bの抗腫瘍性マクロファージの誘導効果(1)
実験方法 DBA/2系雄性マウスを一定期間動物舎内で飼育後、
健康と思われる7〜10週令の7975匹に物質BをP
BS−を用いて、!Fm97m1に調製した被検液10
m1/に9(対照にはPBS−のみを用いた)を腹腔内
投与し1、3および3日後にPECを採取し、熊谷らの
方法に従ってマクロファージを分離した(免疫実験操作
法、211.73〜24L7り(/り7り))。
♂) Effect of substance B on inducing antitumor macrophages (1)
Experimental method After raising DBA/2 male mice in an animal house for a certain period of time,
Substance B was administered to 7,975 apparently healthy animals aged 7 to 10 weeks.
Using BS-! Test solution 10 prepared to Fm97ml
9 (PBS-only was used as a control) was administered intraperitoneally to m1/, PEC were collected 1, 3, and 3 days later, and macrophages were isolated according to the method of Kumagai et al. (Immunology Experimental Procedures, 211.73). ~24L7ri (/ri7ri)).

分離したマクロファージは、to%牛脂児血清添加RP
MI #l)培地にて10 Cs1l/−に調製した。
Isolated macrophages were treated with RP supplemented with to% tallow serum.
MI #l) medium was prepared to 10 Cs1l/-.

その細胞浮遊液をりtウェル平底ブv−)に0.2ml
/wel1分注し、7時間培養後、浮遊細胞を除去し、
PBS−にて洗浄した。次に、10%牛脂児血清添加R
PMI /1.I10培地を用いて105Cal is
 /m’に調製した標的細胞(マストサイトーマPに1
5細胞)をO,コ―/we I L分注し、り時間培養
した。培養終了3時間前に3H−チミジンをコμCi/
W(Ill添加し、培養終了後、グラスフィルターにて
、細胞を採取し、細胞内に取シ込まれた3H−チミジ/
の量を液体シンチン−シロンカウンターにて測定し、下
式によ多細胞障害指数を求めた。
Pour 0.2 ml of the cell suspension into the T-well flat-bottom tube (v-).
/well 1, and after culturing for 7 hours, remove floating cells,
Washed with PBS-. Next, 10% beef fat serum added R
PMI /1. 105 Cal is using I10 medium
Target cells prepared at /m' (mastocytoma P at 1
5 cells) were aliquoted in O, Co/we IL and cultured for an hour. 3 hours before the end of culture, add μCi/3H-thymidine.
After adding W (Ill) and completing the culture, the cells were collected using a glass filter, and the 3H-thymidine/
The amount was measured using a liquid syntin-silon counter, and the multicellular damage index was calculated using the following formula.

対照(cpm ) :未刺激マクロファージと共に培養
した際の3H−チミジンの取り 込み量 試料(cpm ) :試料で活性化したマクロファージ
と共に培養した際の3H−チミ ジンの取り込み量 (2)実験結果 物質Bの添加によるマクロファージの細胞障害指数の経
時的変化は、下表に゛示す通υであ−た。
Control (cpm): Amount of 3H-thymidine uptake when cultured with unstimulated macrophages Sample (cpm): Amount of 3H-thymidine uptake when cultured with macrophages activated with the sample (2) Experimental results Addition of substance B The changes over time in the macrophage cytotoxicity index were as shown in the table below.

物質Bを添加して1日後に得られたマクロファージはマ
ストサイトーマp tit細胞に対して強い細胞障害性
を示すことが認められ、3日および!日後に得られたマ
クロファージでは徐々に活性は減少したものの30%程
度の細胞障害指数が得られ。
Macrophages obtained 1 day after addition of substance B were found to exhibit strong cytotoxicity against mastocytoma ptit cells, and after 3 days and ! Although the activity of macrophages obtained after a day gradually decreased, a cytotoxicity index of about 30% was obtained.

したがって化合物Bに強いマクロファージ活性化作用が
あることが明らかになった。
Therefore, it was revealed that compound B has a strong macrophage activating effect.

り)物質Bの培養細胞に対する直接作用(1)実験方法 マウス正常細胞として3T3−スイスアルピノ株化細胞
CG、 Todara、、 etal、 J、Ce11
.Biol、、 /7゜2タタ(lヂt3))およびマ
ウス腫瘍細胞としてサルコーーr/10株化細胞(J、
 Nut、Cancer In5t、。
(1) Direct action of substance B on cultured cells (1) Experimental method As mouse normal cells, 3T3-Swiss Alpino cell line CG, Todara, etal, J, Ce11
.. Biol, /7°2 Tata (ldit3)) and Sarco r/10 cell line (J,
Nut, Cancer In5t.

/3.  /2タタ(lり!3))の二種の細胞をjチ
FBS(GIBCO)添加D’MEM培地(日水)でよ
×l0IlCo 11/ 0. j rul/vre 
11となるよ5にJウェル平底ブV−ト(−Z Cm”
 、 Bun(5)に分注し、37℃/ j % CO
2/湿潤条件下で夕時間培養した。その後、培地を除去
し、あらかじめ物質Bを!チFBS (GIBCO)添
加D’MEM培地(日水)で16.soおよび2よo)
tg/ mlの濃度に調製した被検液(なお対照には、
物質Bの代わりにjチFBS(GIBCO)添加D′M
EM培地(日水)を加えた)をそれぞれ!濃度につき2
ウエルずつ加えtg時間培養した。その後、o、in)
トリパン・プル(Trypan 、 blue )で処
理し、生細胞数を測定した( Mcllmang、W、
F、、 J。
/3. /2 types of cells (l0!3)) were grown in D'MEM medium (Nissui) supplemented with FBS (GIBCO). j rul/vre
11. 5. J well flat bottom V-to (-Z Cm"
, aliquoted into Buns (5) and heated at 37°C/j% CO
2/ Cultured in the evening under humid conditions. After that, remove the medium and add substance B in advance! 16. with D'MEM medium (Nichisui) supplemented with Chi FBS (GIBCO). so and 2yo)
Test solution prepared to a concentration of tg/ml (for control,
Addition of FBS (GIBCO) D'M instead of substance B
EM medium (Nissui) added) respectively! 2 per concentration
The cells were added to each well and cultured for tg. then o, in)
The cells were treated with Trypan blue and the number of viable cells was measured (Mcllman, W.
F., J.

Immunology、、 79.4Ur (/9’j
7 ) )(2)実験結果 物質Bの添加量と弘ざ時間培養後の細胞の増加率(チ)
との関係は、第μ図に示す通)であつた。
Immunology,, 79.4Ur (/9'j
7) ) (2) Experimental results Addition amount of substance B and increase rate of cells after culture for a long time (chi)
The relationship was as shown in Figure μ).

JTJ−スイスアルピノ株化細胞およびサルコーマir
o株化細胞に対して、物質Bは、いずれの濃度において
も細胞の増殖に影響を与えなかった。
JTJ-Swiss Alpino cell line and sarcoma ir
Regarding cell lines, substance B had no effect on cell proliferation at any concentration.

従−て、培養細胞に対する直接作用がないことが確認で
きた。
Therefore, it was confirmed that there was no direct effect on cultured cells.

to)実験結果の評価 以上の実験結果から、薬用ニンジンから単離された多糖
成分は、生体防御能として細網内皮系機能促進能、細菌
感染抵抗能および抗腫瘍能について九進剤として有効で
あるということができる。
to) Evaluation of experimental results From the above experimental results, the polysaccharide component isolated from medicinal ginseng is effective as a nonalcoholic agent in terms of biodefense ability to promote reticuloendothelial system function, bacterial infection resistance, and antitumor ability. It can be said that there is.

【図面の簡単な説明】[Brief explanation of the drawing]

第1図は、物質Bのカーボンクリアランス能増強作用を
示すグラフである。 第1図は、物質AのE、coli  感染に対する抵抗
性増強作用を示したグラフである。 第3図は、物質Aのサルコーマiro細胞に対する抗腫
瘍作用を示したグラフである。 第≠図は、物質Bの培養細胞に対する直接作用を示した
グラフである。 第5図は、物質Bの13C−NMRスペクトル図を模写
したものである。
FIG. 1 is a graph showing the effect of substance B on enhancing carbon clearance ability. FIG. 1 is a graph showing the effect of substance A on enhancing resistance to E. coli infection. FIG. 3 is a graph showing the antitumor effect of substance A on sarcoma iro cells. Figure ≠ is a graph showing the direct effect of substance B on cultured cells. FIG. 5 is a reproduction of the 13C-NMR spectrum of substance B.

Claims (1)

【特許請求の範囲】 1、単離された薬用ニンジンの多糖成分を有効成分とす
る生体防御能亢進剤。 2、薬用ニンジンが人参三七(Panax notog
inseng F.H.CHEN)である、特許請求の
範囲第1項記載の生体防御能亢進剤。 3、多糖成分がアラビノース、ガラクトースおよびグル
コースから選ばれる少なくとも一種の糖を単位構成成分
として含む、特許請求の範囲第1項または第2項記載の
生体防御能亢進剤。 4、多糖成分が平均分子量100,000以上のもので
ある、特許請求の範囲第1項〜3項記載の生体防御能亢
進剤。 5、生体防御能が細網内皮系機能促進能である、特許請
求の範囲第1〜4項記載の生体防御能亢進剤。 6、生体防御能が細菌感染抵抗能である、特許請求の範
囲第1〜4項記載の生体防御能亢進剤。 7、生体防御能が抗腫瘍能である、特許請求の範囲第1
〜4項記載の生体防御能亢進剤。
[Scope of Claims] 1. A biological defense enhancer containing an isolated polysaccharide component of medicinal ginseng as an active ingredient. 2. Medicinal ginseng is Panax notog
inseng F. H. The biological defense ability enhancer according to claim 1, which is CHEN). 3. The biodefense enhancer according to claim 1 or 2, wherein the polysaccharide component contains at least one type of sugar selected from arabinose, galactose, and glucose as a unit component. 4. The biological defense ability enhancer according to claims 1 to 3, wherein the polysaccharide component has an average molecular weight of 100,000 or more. 5. The biological defense ability enhancing agent according to claims 1 to 4, wherein the biological defense ability is an ability to promote reticuloendothelial system function. 6. The biological defense ability enhancer according to claims 1 to 4, wherein the biological defense ability is bacterial infection resistance. 7. Claim 1, wherein the biological defense ability is antitumor ability
The biological defense ability enhancer according to item 4.
JP59138023A 1984-07-05 1984-07-05 Stimulator for defensive ability of living body Pending JPS6118722A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP59138023A JPS6118722A (en) 1984-07-05 1984-07-05 Stimulator for defensive ability of living body

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP59138023A JPS6118722A (en) 1984-07-05 1984-07-05 Stimulator for defensive ability of living body

Publications (1)

Publication Number Publication Date
JPS6118722A true JPS6118722A (en) 1986-01-27

Family

ID=15212244

Family Applications (1)

Application Number Title Priority Date Filing Date
JP59138023A Pending JPS6118722A (en) 1984-07-05 1984-07-05 Stimulator for defensive ability of living body

Country Status (1)

Country Link
JP (1) JPS6118722A (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000050054A1 (en) * 1999-02-25 2000-08-31 Cv Technologies Inc. Treatment of autoimmune diseases with american ginseng extract
US6555527B1 (en) 1999-08-27 2003-04-29 Korea Atomic Energy Research Institute Hematopoietic, myeloprotecting, antitumor immune cells generating and radiosensitizing polysaccharide isolated from Panax ginseng
JP2005531576A (en) * 2002-05-28 2005-10-20 エムディー バイオアルファ カンパニー リミテッド Fractions with anticancer and antimetastatic activity from carrot leaves and stems
US7075038B2 (en) 2004-04-06 2006-07-11 Jcs/Thg, Llc Cooking apparatus with cooking chamber support
JP2009256387A (en) * 1997-12-12 2009-11-05 Fx Life Sciences Ag Process of making north american ginseng fraction, product containing the same and use as immunomodulator
US9050313B2 (en) 2008-02-29 2015-06-09 Valeant Canada Lp Activation of innate and adaptive immune responses by a ginseng extract
CN106727824A (en) * 2016-12-30 2017-05-31 天津市中升挑战生物科技有限公司 The preparation method and application of chrysanthemum leaf Notogineng Extract and its compound preparation

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009256387A (en) * 1997-12-12 2009-11-05 Fx Life Sciences Ag Process of making north american ginseng fraction, product containing the same and use as immunomodulator
WO2000050054A1 (en) * 1999-02-25 2000-08-31 Cv Technologies Inc. Treatment of autoimmune diseases with american ginseng extract
US8039027B1 (en) 1999-02-25 2011-10-18 Fx Life Sciences Ag Treatment of autoimmune diseases with American ginseng extract
US6555527B1 (en) 1999-08-27 2003-04-29 Korea Atomic Energy Research Institute Hematopoietic, myeloprotecting, antitumor immune cells generating and radiosensitizing polysaccharide isolated from Panax ginseng
JP2005531576A (en) * 2002-05-28 2005-10-20 エムディー バイオアルファ カンパニー リミテッド Fractions with anticancer and antimetastatic activity from carrot leaves and stems
US7075038B2 (en) 2004-04-06 2006-07-11 Jcs/Thg, Llc Cooking apparatus with cooking chamber support
US9050313B2 (en) 2008-02-29 2015-06-09 Valeant Canada Lp Activation of innate and adaptive immune responses by a ginseng extract
CN106727824A (en) * 2016-12-30 2017-05-31 天津市中升挑战生物科技有限公司 The preparation method and application of chrysanthemum leaf Notogineng Extract and its compound preparation

Similar Documents

Publication Publication Date Title
Nie et al. Bioactive polysaccharides from Cordyceps sinensis: Isolation, structure features and bioactivities
JP4194839B2 (en) Method for producing pectin hydrolyzate
Gao et al. Characteristic anti-inflammatory and antioxidative effects of enzymatic-and acidic-hydrolysed mycelium polysaccharides by Oudemansiella radicata on LPS-induced lung injury
JPH05502018A (en) Immune system activation method
Bai et al. Antitumor and immunomodulating activity of a polysaccharide from Sophora flavescens Ait.
US6555527B1 (en) Hematopoietic, myeloprotecting, antitumor immune cells generating and radiosensitizing polysaccharide isolated from Panax ginseng
JPH08500589A (en) Astragalus Polysaccharide Immunomodulator
Pitchaipillai et al. In vitro antidiabetic activity of ethanolic leaf extract of bruguiera Cylindrica L.–glucose uptake by yeast cells method
KR20140091674A (en) Use of foti to enhance stem cell mobilization and proliferation
CN113480676B (en) Oligogalacturonic acid polysaccharide, compound, preparation method and application thereof
JPS6118722A (en) Stimulator for defensive ability of living body
KR101829637B1 (en) A composition for improving, preventing and treating digestion dysfunction, leukocyte reduce, bone marrow suppression by side effects after anti-cancer therapy comprising Rhus verniciflua stoke extract
JP6681466B2 (en) Homogeneous polysaccharide having immunomodulatory activity and method for preparing the same
KR20180090198A (en) Composition comprising the extract of Molokia leaf for immune activity
JPH0245499A (en) Glycoprotein of panax ginseng and use thereof
Qu et al. Characterization and macrophages immunomodulatory activity of two water-soluble polysaccharides from Abrus cantoniensis
US20160227829A1 (en) Dietary composition containing cistanche deserticola polysaccharide with inhibitory effects on colon cancer
US6838096B2 (en) Black soybean polysaccharides
Son et al. Effects of intravenous administration of polysaccharide purified from fermented barley on tumor metastasis inhibition via immunostimulating activities
CN109294984B (en) Lentinan capsule for efficiently amplifying NK cells in vivo and preparation method thereof
EP1398036B1 (en) Ganoderma lucidum spores for treatment of systemic lupus erytrhomatosus (SLE)
JPS61109732A (en) Agent for inducing tumoricidal substance
CN101053588A (en) Preparation method for compound arabinogalactan film coated tablets
JPH0245501A (en) Chikusetsu ginseng polysaccharide and use thereof
WO2014054421A1 (en) Peyer&#39;s patch activator