Skip to main content

Anesthesia and analgesia for common research models of adult mice

Abstract

Anesthesia and analgesia are major components of many interventional studies on laboratory animals. However, various studies have shown improper reporting or use of anesthetics/analgesics in research proposals and published articles. In many cases, it seems “anesthesia” and “analgesia” are used interchangeably, while they are referring to two different concepts. Not only this is an unethical practice, but also it may be one of the reasons for the proven suboptimal quality of many animal researches. This is a widespread problem among investigations on various species of animals. However, it could be imagined that it may be more prevalent for the most common species of laboratory animals, such as the laboratory mice. In this review, proper anesthetic/analgesic methods for routine procedures on laboratory mice are discussed. We considered the available literature and critically reviewed their anesthetic/analgesic methods. Detailed dosing and pharmacological information for the relevant drugs are provided and some of the drugs’ side effects are discussed. This paper provides the necessary data for an informed choice of anesthetic/analgesic methods in some routine procedures on laboratory mice.

Background

Anesthesia and analgesia (A&A) are the main components of many procedures on laboratory animals. However, studies suggest that a considerable number of researches are based on improper use or reporting of A&A methods [1, 2]. In a study on 400 articles that included surgical models in mouse and other species, the authors concluded that the animal A&A are not reliably presented in the literature [3]. Another study also shows mouse research is essentially not reported well in the European literature [4].

Part of this problem may be related to insufficient training of investigators regarding A&A in laboratory animals [2]. For example, there is a common myth among some researchers assuming anesthesia is equivalent to analgesia. Accordingly, some papers may report using a hypnotic drug (pentobarbital) without analgesia for performing painful recovery surgeries on mice [5]. Navarro et al., discuss that the current method of peer-to-peer training in laboratory animal science, may causes misinterpretation of A&A concepts among researchers [6]. Also, the unfamiliarity of researchers with available anesthetic options in mice has led to limited range of anesthetic protocols in this animal [6].

Another part of this problem may be related to responses of A&A to specific anatomic and physiologic attributes of mice. For example, fast metabolism and excretion of A&A drugs in mice, reduces the half-life of these drugs and limits the duration of A&A. This along with reduced liver glycogen reserve in mice, promote hypoglycemia. When compared with larger species, the higher ratio of body surface area to body volume in mice leads to faster heat loss and hypothermia [7]. Small body size of mice also causes physical problmes. For example, endotracheal intubation and ventilation of this species are demanding tasks, and many veterinary anesthesia monitoring devices could not be used for mice anesthesia.

There may be other explanations for this large number of inappropriate A&A methods, such as fewer regulations in animal research (in comparison to veterinary clinical practice), lower availability of anesthetic monitoring devices in research settings, or high diversity of research procedures requiring anesthesia.

It is while, proper induction and maintenance of A&A are essential elements of maintaining animal welfare and preserving the scientific integrity of research [6, 8]. In one hand, distress and pain are components of many experimental procedures and substantially affect animal welfare. Therefore, proper use of sedatives, tranquilizers, analgesics and anesthetics are essential ethical requisites of many experiments [7]. On the other hand, using these drugs has two types of effects: (1) Alleviating the distress and pain of the animal which have important biochemical and physiologic effects on samples and data generated by the animal. For example, it is shown that pain causes an increase of epinephrine, cortisol, plasma glucose, endorphins, and many other chemicals in the body. Pain also increases heart rate, stroke volume, cardiac output, and leads to shallow fast breathing, etc. [9]. Proper use of analgesics can substantially prevent these effects and produce good data; (2) Direct physiologic effect of the drug on the animal and its corresponding samples and data [10]. For example, isoflurane better preserve cardiovascular functions than halothane. However, it is a potent suppressor of the respiratory system and its vasodilatory effects are more than halothane [11]. Therefore, proper choose of drugs in experiments can minimize the side effects of the drugs on research data.

The laboratory mouse is the most used species of animals in research, with an estimate of 61% of laboratory animals used in the European Union in 2017 [12]. Therefore, A&A in mice may constitute a major part of the problem with laboratory animal anesthesia. There are at least three invaluable papers [6, 7, 13] with a detailed discussion of A&A in mice. However, these papers have mainly discussed A&A methods according to the classes of medications rather than the actual procedures. Although this may be helpful in some circumstances, researchers are usually encountered with the question of “which anesthetic protocol should I use for procedure X?”. In this review, we would like to answer this type of question by discussing A&A for routine procedures on adult mice. We used appropriate references in this regard and critically appraised the quality of their anesthetic methods. The dose of anesthetic and perianesthetic medications are presented in Additional file 1. Analgesics doses are presented in Additional file 2. Important pharmacological data for most common anesthetic agents are presented in Additional file 3. An overview of the anesthetic modalities for various procedures on adult laboratory mice is provided in Table 1.

Table 1 Suggested anesthetics for common procedures on adult mice. For details, please refer to the text

Anesthesia

Anesthesia may be divided into three different concepts: local anesthesia, general anesthesia, and surgical anesthesia. Local anesthesia (aka local analgesia) refers to the loss of pain sensation in a specific body area [10]. General anesthesia is a drug-induced state of reversible unconsciousness, in which the animal could not be aroused by noxious stimuli [10] but it does not necessarily mean that the animal does not sense or perceive pain. Surgical anesthesia is a state of general anesthesia that has four components: unconsciousness, amnesia, muscular relaxation, and analgesia [10]. Currently, no anesthetic agent exists that could be solely used for achieving surgical anesthesia. Therefore, various agents are used to develop various components of surgical anesthesia and this is called ‘balanced anesthesia’. In a research setting, the method of anesthesia for mice may be selected according to the duration and type of the procedure, comfortability of the anesthetist with a method, and availability of required drugs and devices. Understanding the effects of anesthesia on research outcomes may determine the selection of the anesthetic method. When there is no alternative method available, this knowledge helps in inferring the achieved results. For example, some studies have reported that pentobarbital sodium or isoflurane may significantly alter tissue compositions [14,15,16,17,18,19]. Also, it is shown that short-term (2 min) and long-term (more than 10 min) anesthesia with either pentobarbital sodium or isoflurane before transcardial perfusion can make alterations in brain immunohistochemistry results [20].

Surgical anesthesia may be divided into five parts: (1) pre-anesthetic preparation including the selection of an anesthetic candidate and its preparation for anesthesia/surgery; (2) anesthetic induction, as the process by which a conscious animal is rendered unconscious; (3) anesthetic maintenance, that is maintaining an animal unconscious throughout the procedure; (4) anesthetic recovery, as the process by which an unconscious animal regains consciousness; and (5) post-anesthetic care, which includes animal care to gain full consciousness.

Analgesia

Surgical pain feeling includes nociception and nociperception. Nociception comprises the encoding of noxious stimuli to neural impulses (transduction), transmitting the encoded neural impulses to the CNS (transmission), and modulating neural impulses before delivering them to the brain (modulation). Nociception is independent of CNS activity [10]. In contrast, nociperception is the translation of neural impulses to conscious perception of pain in the brain [10].

Accordingly, analgesia refers to methods of inhibiting or interrupting nociception or nociperception, so that the patient does not feel pain in response to noxious stimuli that are otherwise painful [10]. Although general anesthesia may inhibit ‘nociperception’ to varying extents, it generally does not affect nociception. Therefore, if effective analgesic methods are not used, we may have an animal under general anesthesia but with ongoing nociception. Furthermore, anesthetic drugs may suppress motor functions and physical signs related to pain. At lower levels of unconsciousness (i.e. subanesthesic doses), the animal may perceive pain but may be unable to demonstrate associated physical signs. This may occur during the anesthetic recovery of an animal that has not received proper perioperative analgesia.

Pre-anesthetic preparation

Pre-anesthetic preparation of mice is best described elsewhere [6, 7]. Anticholinergics (atropine or glycopyrrolate) may be used to reduce salivary and bronchial secretions and avoid the substantial drop in heart rate due to anesthetic medications or surgical manipulations [7]. Anesthesia can be induced less than 10 min following IP administration of atropine.

Anesthetic monitoring and perianesthetic care

Anesthetic monitoring and perianesthetic care are best discussed in previous studies [6, 7, 13]. Briefly, the following parameters should be monitored and corrected if needed: depth of anesthesia, analgesia (hypoalgesia), body temperature, blood glucose level, body hydration, respiratory status (depth, rate, and pattern), heart and vascular functions (heart rate, capillary refill time, mucus membranes’ color, and pulse quality) and blood oxygen level (pulse-oximetry). The cornea should be kept moistened (e.g., by vitamin A + D ophthalmic ointment or wet pads). The animal should be protected from postural hypotension.

Induction

When using injectable anesthetics in mice, induction is usually indistinguishable from maintenance. This is in contrast to inhalational anesthesia, which usually includes a separate induction phase. We will discuss the induction along with maintenance, in the following sections.

Maintenance

Injections and catheterizations

Simple injections may induce a sudden sharp pain in animals. This may have stress-related consequences that may confound the outcomes of some studies. To reduce this effect, training of the animal may be considered before using them in projects [21], and this is a proven method of desensitizing the animal to simple injections. This is well demonstrated by the RISE institute (www.ri.se). Also, one may distract the animal by talking, giving treats, or gently touching its body while the injection is given.

To completely eliminate the pain, the topical anesthetic compound lidocaine-prilocaine (EMLA® cream) can be applied on the skin, 30–60 min before needle insertion [22, 23]. Flash anesthesia using an isoflurane chamber is suggested for inducing anesthesia for catheterization with minimal hemodynamic effects [24,25,26]. However, we think that the stress of anesthesia (even very quickly) is more than the stress induced by the insertion of a hypodermic needle.

Working with agitated animals

Performing procedures on agitated animals may require tranquilization with a lower dose of acepromazine. It should be noted that the animal can still perceive pain in this condition [27, 28].

Temporary immobilization for non-painful procedures

For some non-painful procedures, the animal may require a higher level of sedation. This may be useful for imaging techniques that require temporary (~ 5 min) immobilization. It may be accomplished in mice by using a higher dose of acepromazine. Flash anesthetic techniques using an anesthetic chamber (isoflurane or halothane), may also produce a sleep-like state in mice. To extend the duration of anesthesia, the nose cone method [29] can be used.

Longer immobilization for non-painful procedures

This may be used for long imaging processes or any non-painful procedure requiring complete immobilization. In this regard, hypnotics such as thiopental or pentobarbital may be used.

Imaging

In the majority of the imaging technologies, the mice need to be as calm as possible since movements could lead to disruption of signal acquisition and this would lead to false data generation [30]. The mice may be immobilized with restraining or anesthetizing. For some studies where anesthesia would interfere with the metabolic and biochemical processes of the study, the mouse should be restrained and imaged [31]. There are studies carried out on imaging devices where the animal would be free and imaged at the same time. This requires a device to be surgically attached to the animal so that the signals would be acquired [32].

Restraining a mouse during the imaging acquisition for a duration of seconds to hours is stressful and adds confounding factors to the imaging process. Therefore, if possible, the mouse should be anesthetized. Since most imaging procedures do not incur pain, analgesia may not be required [33]. However, for invasive imaging techniques, the type of analgesia is determined according to the type and extent of the invasive procedure.

In a rat study, it was shown that ketamine and chloral hydrate increase the binding of receptor ligands to dopamine D1 receptors in Positron emission tomography (PET) imaging [34], whereas pentobarbital decreased the binding. Similar findings are expected in mice. Ketamine and xylazine also were shown to increase the blood fluorodeoxyglucose (FDG) activity and reduce the tumor FDG uptake, causing a less contrast image to be acquired using PET imaging [35]. The same study also demonstrated the same effect using pentobarbital, but its effect was less prominent than ketamine, and xylazine. Isoflurane, sevoflurane, and desflurane inhibit the activity of the luciferase enzyme, which generates signals for detection by bioluminescence imaging [36]. Also, repetitive sevoflurane usage has an immunomodulatory effect in mice and this could affect the radiation imaging and the well-being of the animal [37]. Isoflurane was also shown to reduce the uptake of radioactively labeled FDG and subsequently PET imaging in mouse heart and brain, due to overall hypothermia caused by isoflurane [38]. Respiratory acidosis, as a complication of anesthesia, is concurrent with an increase in blood pCO2 level and this may affect PET imaging with FDG, in mouse models of cancer and arthritis [39]. More details on the anesthetic methods for imaging are presented elsewhere [13, 40].

Ophthalmic procedures

Heat, mechanical, and chemical stimuli could cause neurological reactions in most corneal nerves. Topical anesthetics may be used to reversibly block sodium channels of corneal nerves and avoid the transfer of impulses in the cornea, conjunctiva, and sclera [41]. Oxybuprocaine eye ointment may be used to provide local analgesia for ophthalmic procedures [7].

For enucleation, researchers have used intraperitoneal ketamine-medetomidine. At the end of the surgery, the effect of medetomidine was reversed using intraperitoneal atipamezole. Meloxicam was used as a postoperative analgesia [42]. It is shown that medetomidine could reduce tear production in a dose-dependent fashion [43]. It should be noted that administration of ketamine-medetomidine could not provide surgical anesthesia consistently. Therefore, anesthetic depth monitoring should be continuously performed before and during surgical procedures [44] and analgesics (such as buprenorphine) should be used when the anesthetic depth is insufficient. This combination is generally advised for minor procedures on mice [45].

For intravitreal implant injection, the mice were anesthetized using ketamine-xylazine [46]. This combination has also been used for developing an ocular hypertension model [47]. However, this combination is shown to induce corneal damage, irrespective of the treatments applied, to prevent corneal dehydration [48]. Ketamine-xylazine combination is believed to alter the ocular physiology and cause xylazine-mediated reversible cataracts [49, 50]. This combination may also cause retinal Müller cell reactivity [49].

For performing retro-orbital injection to the venous sinus, anesthesia was induced using an isoflurane induction chamber [51]. Then, the mouse was connected to a non-rebreathing anesthetic system via a funnel-shaped nose cone. A drop of 0.5% proparacaine hydrochloride ophthalmic solution was applied to the eye to provide sufficient analgesia for a painless procedure [51]. It should be noted that blood sampling from retro-orbital venous sinus is considered a terminal procedure in some countries (e.g., UK) and the animal should be euthanized after the procedure without gaining consciousness.

Intraperitoneal injections of a diluted solution of Hypnorm™ (fentanyl/fluanisone) and midazolam have been used for inducing conjunctival scarring models [52, 53]. In rats, this combination could also cause histologic corneal lesions with scores of 2–4. The severity and incidence rate of corneal lesions when using Hypnorm™ and midazolam in rats is less than ketamine-xylazine but more than pentobarbital or isoflurane [54]. Midazolam inhibits Vascular endothelial growth factor-mediated events in retinal cells, thus preventing hyperglycemia-induced vascular leakage in a mouse model of diabetes [55].

Various methods of anesthesia could affect the intraocular pressure (IOP) and this requires IOP monitoring before and during anesthesia. For example, anesthesia with ketamine/xylazine may result in an increased IOP [41, 56]. Ketamine/xylazine can also cause hyperopia and change the refraction of the eye. However, it is shown that pentobarbital has no effect on eye refraction [57]. To prevent oculocardiac reflex resulting from eyes manipulation, one may use atropine or glycopyrrolate [58].

Limbs

Limb procedures include limb ischemia model, model of gangrene, ischemia–reperfusion models, lymphedema, heat-induced limb length asymmetry, nerve transection, and limb transplantation. There are three types of pain involved in models involving femoral artery ligation (e.g. limb ischemia, gangrene, or ischemia–reperfusion models): (1) a somatic pain resulting from skin incision, subcutaneous tissue dissection, and skin suturing; and (2) a neurogenic pain caused by femoral nerve manipulation, and (3) ischemic pain consequent to tissue hypoxia caused by arterial ligation.

For the limb ischemia model, general anesthesia comprising 1.0 to 1.5% isoflurane, 60% dinitrogen monoxide, and 40% oxygen were used [59]. However, this anesthetic regimen seems to lack the potent analgesic component of balanced anesthesia. Therefore, we would add proper analgesics such as opioids (e.g., buprenorphine or morphine) and nonsteroidal anti-inflammatory drugs (NSAIDs; e.g., ketoprofen or flunixin meglumine) to this anesthetic regimen to provide surgical anesthesia. General anesthesia with an intraperitoneal injection of ketamine/xylazine is reported in the literature [60, 61] and seems to be more pertinent to this operation. Another study [62] has concluded that isoflurane is the most used inhalational anesthetic compound for modeling limb ischemia in mice. It has also ranked a combination of midazolam/medetomidine to be one of the most used injectable anesthetic compounds for this model [62]. Because of the early peripheral vasoconstriction induced by alpha-2 agonists, such as xylazine, the use of these agents in research concerning vessels with large amounts of vascular smooth muscles, is argued [62].

In a model of gangrene, intraperitoneal ketamine/xylazine accompanied by subcutaneous injection of sustained-release buprenorphine was used for performing ligation surgery on the femoral artery and vein [63].

For the ischemia–reperfusion model using the open and closed techniques, mice were anesthetized by inhalational isoflurane and subcutaneous buprenorphine [64].

Lymphedema models include irradiation with a single dose of 30 Gy ionizing radiation, using an X-ray machine. For this purpose, intraperitoneal injection of pentobarbital [65] or inhalational isoflurane [66] is used. After one week, a surgical operation was done under isoflurane anesthesia [65]. The 90-min surgical operation included a subcutaneous injection of a dye in the left paw, skin incision at the left inguinal region, ligature of the lymphatic vessels at three points, resection of the subiliac and popliteal lymph nodes, and suturing of the skin to its underlying muscles without apposing the skin edges [65]. Accordingly, reported isoflurane anesthesia lacks the analgesic component. We suggest adding an opioid analgesic (e.g., buprenorphine) to this protocol. As an alternative to isoflurane, another study has used subcutaneous fentanyl/midazolam for this surgery [66]. Postoperative analgesia included subcutaneous buprenorphine every 8 h [65].

In a model of heat-induced limb length asymmetry, the right sides of the body of the mice were positioned on a heating pad with a 40 °C constant temperature. For this purpose, isoflurane inhalation was used to hold the mice in lateral recumbency [67]. For Laser Doppler perfusion imaging of limbs, mice were lightly anesthetized by first inducing anesthesia in an induction chamber, and then by maintaining anesthesia by a nasal cannula delivering isoflurane/oxygen [63].Nerve transection on limbs may be performed on sciatic or femoral nerves. For this purpose, anesthesia was induced with 2–3% isoflurane. A single dose of buprenorphine was administered “prior to starting the surgery” [68]. In this regard, we suggest buprenorphine injection be performed about 1 h before starting the first surgical incision so that the preemptive analgesic requirement is met. We would also suggest a splash block with a drop of 1% lidocaine at the nerve resection site.

For limb transplantation research, limb resection is performed on a donor and recipient by incising the skin, muscles, nerves, and vascular structures and mid-shaft femoral transection [69]. The resected limb is then implanted in the recipient, by suturing appropriate structures together. For this purpose, analgesia was achieved using buprenorphine, administrated 1 h before the first skin incision. Anesthesia was induced in an isoflurane induction chamber and maintained by a nose cone delivering isoflurane and oxygen [69]. Since the procedure involves both soft tissues and hard tissue transections, we suggest using a lidocaine splash block on the nerves and also the femoral shaft before transecting these sensitive organs. The postoperative pain of the recipient was controlled with subcutaneous buprenorphine every 6 h, up to 72 h post-surgery [69].

Tracheostomy

Tracheostomy needs skin and fascia incisions, as well as dissection of the muscles and trachea, which all contribute to somatic pain. Considering the few nociceptors available in the trachea, it seems that visceral pain is not associated with tracheostomy-induced pain.

Tracheostomy is usually a preparation for a main surgical procedure. The choice of anesthetic agent for tracheostomy depends on the requirements of the main surgical model. As reported, intraperitoneal injection of pentobarbital sodium/ketamine [70], urethane/alpha-chloralose [71], and ketamine/xylazine [72], as well as inhaled isoflurane have been applied as anesthetic agents in this surgical procedure. The combination of pentobarbital/Ketamine/xylazine decreases the tracheal mucociliary clearance [73]. However, isoflurane, propofol, or fentanyl/dexmedetomidine/midazolam do not seem to change the tracheal mucociliary clearance values [73]. Opioid analgesics such as buprenorphine should be used as intra-operative analgesics. Due to the carcinogenic effects of urethane, we do not recommend using this agent for anesthesia.

Laparotomy

The abdominal cavity contains several viscera that are important in biomedical research. Laparotomy is the main approach to access these viscera. Laparotomy pain may originate from the soft tissues in and around the abdominal incision site including the skin, fascia, muscles, and ligaments. The superficial nerves may be compressed following surgery due to inflammation, scar tissue formation, muscle length unfitness, or hyperexcitability of the nerves, and may lead to neuralgia. Manipulation of viscera may cause acute (surgical) and chronic (postsurgical) pain and should be targeted appropriately [74]. For the first 24 h after laparoscopic procedures, it is believed that visceral pain constitutes the major part of overall pain [75, 76].

The most typical anesthetic agents used in mice models of laparotomy include inhaled anesthetics, such as isoflurane and sevoflurane, and injectable anesthetics, like ketamine/xylazine/acepromazine. Intraperitoneal injection of drugs for laparotomy has been a topic of debate. However, various reports have used this method [77, 78]. In general, we suggest avoiding abdominal lavage if the intraperitoneal route is used for anesthesia. Subcutaneous administration of lidocaine (10 mg/kg) with or without bupivacaine (3 mg/kg) at the intended site for the incision (line block) [79], provides multimodal analgesia and decreases the dose of the main anesthetics.

Liver and bile duct

For bile duct ligation, isoflurane or sevoflurane [80, 81] are used. In rats, it is shown that isoflurane induces liver injury, mainly due to a regulation of expression of insulin-like growth factor-1 [82]. However, isoflurane has lower hepatotoxicity, in comparison to other halogenated anesthetics (e.g., halothane) [83,84,85]. In horses, it is shown that anesthesia with isoflurane increases hepatic bilirubin excretion, but has a modest effect on hepatic bilirubin formation or clearance [86]. In humans’ serum samples taken 3–14 days after anesthesia, sevoflurane is shown to less frequently induce elevation of liver enzymes than isoflurane [87].

Besides using inhalation anesthetics, other systemic anesthetic agents such as a mixture of ketamine/xylazine/acepromazine have also been used [40, 88]. It is shown that ketamine/xylazine/acepromazine is a safe and effective protocol for liver transplantation surgery in mice [89]. Ketamine is shown to have hepatoprotective effects [90]. Postoperative pain of the bile duct ligation surgery could be alleviated by buprenorphine [91].

Partial hepatectomy is performed by isoflurane anesthesia along with meloxicam and buprenorphine analgesia. It is shown that flunixin meglumine could not provide enough analgesia in this surgery [92].

Kidneys

For renal models, pain may be induced by renal mechanosensory nerves (pressure-responsive) and renal chemosensory nerves (responsive to ischemia or renal interstitial fluid alterations) [93]. This is in addition to the general mechanisms of pain related to laparotomies. The proper anesthetic protocol should preserve renal functions during kidney studies. To induce renal ischemia–reperfusion injury (RIRI), a combination of ketamine/xylazine [94] or ketamine/medetomidine [95] is used. Although ketamine metabolites are eliminated by kidneys, it is generally safe to use it with models of compromised kidneys, such as RIRI. However, it is recommended to reduce the dose of xylazine in renal failure models [7]. Therefore, to provide required analgesia, buprenorphine should be used along with ketamine/xylazine or ketamine/medetomidine anesthesia [95].

For the renal ischemia model, the ketamine/xylazine combination is not recommended due to its short duration of surgical anesthesia (~ 20 min.), which is not adequate for a 1-h procedure. Isoflurane is also not a viable option due to its intrinsic renoprotective activity, which could counteract the effect of the surgical procedure [96]. Intraperitoneal injection of pentobarbital has been used for the renal ischemia model [96]. However, it should be combined with a potent analgesic (e.g., buprenorphine).

Buprenorphine is a recommended analgesia for mouse models of RIRI and ureteral obstruction [94, 97,98,99]. Since buprenorphine is metabolized by the liver, it can be used in models of compromised kidney functions [100]. Due to the renal side effects of NSAIDs, they should not be used as analgesic agents in RIRI models [58, 94].

For performing the renal micropuncture technique, thiobutabarbital sodium (Inactin®) has been used [101]. However, this short-acting barbiturate derivative does not possess analgesic effects and is not recommended to be used alone. Also, mice’s response to thiobutabarbital is not consistent [102]. Accordingly, ketamine has been used in combination with thiobutabarbital [101, 103]. Furthermore, barbiturates, like sodium pentobarbital are counter-indicated in RIRI models. In fact, they reduce blood pressure that would subsequently reduce kidney blood flow, glomerular filtration rate, and urine excretion [94]. Halothane and isoflurane may have fewer side effects on vascular activities and can be applied [101].

Due to better preservation of the renal function, α-chloralose seems to be an effective anesthetic agent for studying renal function in acute interventions [103]. However, α-chloralose lacks analgesic properties and is usually used for non-survival procedures [6, 104]. Therefore, we recommend considering other anesthetic protocols for this purpose.

For ureteral obstruction development, isoflurane [6, 105, 106] or pentobarbital [107] is used. Intra-operative analgesia is achieved using subcutaneous buprenorphine. Postoperative analgesia is maintained by local infiltration of bupivacaine at the incision site and continuation of buprenorphine injection for two days after surgery [106].

Goldblatt model of hypertension requires surgically induced renal arterial stenosis and may have three subtypes: two-kidney one-clip, two-kidney two-clip, and one-kidney one-clip. For these models, anesthetic agents with a minimum effect on renal or cardiovascular function are preferred. Isoflurane [108], pentobarbital [109], ketamine/xylazine [110,111,112], or ketamine/xylazine/acepromazine [113] are used for this model. Among these, ketamine/xylazine/acepromazine seems to better provide surgical anesthesia. Buprenorphine is used for post-surgical analgesia in this model [114].

Abdominal aorta

For abdominal aortic constriction, isoflurane [115, 116] and ketamine/xylazine [117] are used. Although using a ketamine/xylazine mixture is more frequent than that of isoflurane, this combination is not generally advised for major surgeries in mice [22]. Isoflurane should be used with a potent analgesic (e.g., buprenorphine).

Gastrointestinal tract

Gastrointestinal (GI) pain, as a form of visceral pain, may be induced by GI disorders or manipulation. For GI surgeries, anesthesia can be induced and maintained with isoflurane [118, 119] or sevoflurane. Buprenorphine is used along with these inhalational agents to provide analgesia [102, 119]. Isoflurane is shown to reduce GI propulsion in rats [120, 121]. Sevoflurane could change the composition of the intestinal microbiome in mice [122]. Breathing anesthetics are more convenient for both the operator and the animals [6, 102]. However, intraperitoneal injection of a ketamine/xylazine/acepromazine mixture or pentobarbital/buprenorphine is also used. Studies have shown no effect of ketamine on intestinal peristalsis in dogs, while they have shown shortened or prolonged peristalsis in pigs [123]. Ketamine is shown to protect the intestines from ischemic damage [123] and safeguard the stomach from lipopolysaccharide-induced injury [102]. Buprenorphine reduces GI motility and slows the passage of feed in the GI tract [124].

Dutton et al. [125] concluded that pentobarbital may induce local inflammation, and therefore it is not recommended for inflammatory bowel disease models. Dexmedetomidine is shown to significantly inhibit intestinal peristalsis in endotoxemic mice [126], suggesting that this drug should be used with caution in ileus models [126].

Buprenorphine is the routine analgesic for GI surgeries. It should be administered 1 h preceding the incision, with a second dose 6–12 h following the first dose. The next doses are prescribed within 6–12 h intervals, up to 72 h post-surgery [22].

Testes

Semen collection may be performed from the female reproductive tract after mating. This requires euthanasia of the female animal and resection of the whole reproductive tract. In this regard, an overdose of intraperitoneal thiopental or pentobarbital (200 mg/kg) with concentration of 60 mg/mL or intraperitoneal ketamine/xylazine with euthanasia dose (300–400 mg/kg + 30–40 mg/kg) can be used [127]. Another method for semen collection is through electroejaculation of the male mice, using a rectal probe. The mouse may be anesthetized with intraperitoneal ketamine/xylazine or intraperitoneal pentobarbitone [128]. It is shown that pentobarbitone anesthesia led to higher spermatozoa count and less urine contamination in the collected semen [128]. However, it should be noted that pentobarbitone induces severe respiratory depression in mice [129].

Ovaries

Oophorectomy or ovariectomy may be performed using isoflurane [130] or halothane [131, 132]. Intraperitoneal injection of ketamine/medetomidine has also been used for general anesthesia for this experimental procedure [132]. Intra-operative and postoperative analgesia may be achieved by opioids (e.g., buprenorphine) and NSAIDs (e.g., ketoprofen).

For oocyte collection, 2% tribromoethanol intraperitoneally or inhalation of isoflurane is used [133]. In oocyte retrieval surgery, any agent administered intraperitoneally may come in direct contact with the oviduct or cumulus-oocyte mass and could have toxic effects on oocytes. It is shown that mice anesthetized with intraperitoneal tribromoethanol have a higher proportion of dead oocytes, in comparison to mice euthanized just before the oocyte collection or mice anesthetized with isoflurane [133]. Therefore in these surgeries, it is recommended to use inhalation anesthesia, intravenous or intramuscular injectable anesthesia [134].

Skull

Surgeries involving the skull are categorized as brain surgeries or stroke induction.

Brain

Brain surgery is usually performed under stereotaxic frame guidance and involves injury to soft tissues (skin, muscles), and the osseous skull. Isoflurane [135,136,137] or ketamine/xylazine [136] are used for stereotaxic surgeries. Isoflurane anesthesia is combined with subcutaneous buprenorphine for intra-operative and postoperative analgesia for stereotaxic surgeries [135].

In one study [135], buprenorphine was injected just before making the surgical incision; though to achieve preemptive analgesia, we recommend the administration of buprenorphine 1 h before making the incision. In order to achieve multimodal analgesia, lidocaine (10 mg/kg) with or without bupivacaine (3 mg/kg) could be injected subcutaneously [79], just before making the skin incision. Also, before drilling or cutting the skull, its periosteum should be anesthetized with a drop of 1% lidocaine splash block [137]. It is also recommended to apply another lidocaine splash block to the periosteum after a few drillings [137]. It is shown that subcutaneous buprenorphine provides more effective analgesia, compared to commonly used NSAIDs (such as carprofen and meloxicam), following craniotomy [135]. Also, buprenorphine reduces the pain and distress in the subarachnoid hemorrhage model, but meloxicam or carprofen is unable to achieve this [138]. However, concurrent use of carprofen or meloxicam with buprenorphine reduces tissue inflammation after surgery. Subcutaneous dexamethasone (0.2 mg/kg) has been used to prevent brain swelling in craniotomy models [137, 139]. Carprofen is used for suppressing inflammation and immune response in a chronic model of the cranial window for cortical activity imaging [140].

For inducing seizures, stereotaxic intrahippocampal administration of kainic acid is performed under isoflurane anesthesia, combined with buprenorphine. Since isoflurane has no effect on ion channel function, this agent is recommended for the kainic acid-induced seizure model [136]. Ketamine/xylazine along with local anesthesia may also be used for this purpose, though ketamine may delay the onset of seizures due to its blocking effects on the NMDA channels [136].

Stroke

Ketamine and barbiturates are known to have a neuroprotective effect and may interfere with the effect of the test substance in stroke models. Due to this side effect, inhalation anesthesia is preferred for the induction of experimental stroke models [141]. Mechanical ventilation during isoflurane anesthesia seems to reduce the undesirable effects of anesthesia on the stroke model [141]. In a middle cerebral artery occlusion model, it was shown that postoperative meloxicam significantly reduced and confounded the stroke volume. Buprenorphine did not show this effect and was declared to be the proper analgesia for this stroke model [142, 143].

Spinal cord

Laminectomy is orthopedic surgery. Pain from spinal cord injury (SCI) is a type of neuropathic pain that is categorized as chronic pain [144]. Isoflurane has been used for developing the SCI model. Buprenorphine is used as postoperative analgesia for 2–3 days and after that, as needed per animal [145, 146]. In rats, buprenorphine could be used as an analgesic following SCI models, without behavioral, physiological, or anatomical effects [147]. It is shown that buprenorphine is more potent than morphine in alleviating neuropathic pain caused by SCI [147, 148].

Intraperitoneal injection of ketamine/xylazine is used to transplant neural stem cells into the spinal cord [149]. Chloral hydrate is an agent frequently used in studies of SCI or spared nerve injury [150, 151]. However, this agent is classified as a sedative without analgesic properties, and can only be used as a pre- or postoperative medication [152]. Furthermore, it is now considered an antiquated agent with serious animal welfare concerns [6]. Therefore, it should be avoided and replaced with other agents.

Tribromoethanol may rarely be used as an anesthetic in rodents [153]. This compound is very unstable and has specific storage conditions. Following intraperitoneal injection, it may cause severe inflammation and peritonitis in animals [154]. Due to the adverse effects of tribromoethanol, it is recommended to avoid using this agent [22].

Ketamine/xylazine are used despite the fact that they provide short-term surgical anesthesia (~ 20 min.), while laminectomy is a longer procedure. Therefore, we do not recommend this compound. To make a model of complete-crush transection SCI, mice were anesthetized using isoflurane [155]. However, this protocol lacks the analgesic component and we suggest a combined use of buprenorphine injection along with local analgesia at the site of laminectomy and spinal cord crushing.

It is shown that microinjection of lidocaine through a lumbar puncture following SCI in rats, prevents adverse cellular events related to the injury, and promotes behavioral recovery of the animal [156].

Thoracotomy

Thoracotomy may involve both somatic and visceral pains. When the intercostal approach is used, the pain has resulted from skin incision, dissection of the intercostal muscles that are heavily innervated, and manipulation of the thoracic visceral organs. When the thoracotomy involves hard tissues as in sternotomy or costotomy, disrupting the hard tissues of the sternum or ribs add to the overall pain of the procedure.

In many thoracotomy procedures, using anesthetic protocols with minimum cardiovascular side effects is of utmost importance. In this regard, urethane, sodium pentobarbital, and ketamine/xylazine have been shown to induce substantial depression in the cardiovascular system and are best to avoid when the (patho)physiology of the system is in question. It is suggested that isoflurane has minimum cardiovascular side effects and has a rapid recovery [157]. This is also shown that isoflurane has fewer effects on hemodynamics than ketamine/xylazine, urethane, or pentobarbital sodium [25].

Although anesthetic agents such as pentobarbital sodium [158], isoflurane [159], or ketamine/xylazine [160, 161] have been extensively used in literature, thoracotomy is considered one of the most painful procedures, and the major drawbacks of these agents are their poor, if any, analgesic effect [22, 162, 163].

To ensure a humane procedure, a potent analgesic regimen should be applied. In one study, anesthesia is induced by an intraperitoneal injection of a combination of midazolam/medetomidine/fentanyl. Afterward, the anesthesia is maintained by isoflurane and mechanical ventilation [164]. As an alternative, a mixture of ketamine and medetomidine may be used [164], in which we suggest adding buprenorphine 1 h before the first surgical incision.

In another study, anesthesia was induced in an anesthetic chamber containing isoflurane [165]. Afterward, intraperitoneal injection of a mixture of buprenorphine (0.1 mg/kg) and etomidate (10 mg/kg) was given. Anesthesia was continued initially by administrating 2% isoflurane with an oxygen flow rate of 80 ml/min [165], which was later reduced to 1–1.5% isoflurane to achieve a proper depth of anesthesia [166]. To connect the animal to the anesthetic system, endotracheal intubation was performed using a 3-cm shortened plastic cannula (G20). The tube was connected to a rodent ventilator with set values for the ventilation rate = 53.5 × M−0.26, and tidal volume = 6.2 × M1.01; in which M is the bodyweight of the animal in grams [165, 166]. Tidal volume was set to obtain a peak inspiratory pressure of 11 ± 1 cmH2O [165]. To avoid the interruption of normal respiration with ventilations, pancuronium can be injected intraperitoneally. Using neuromuscular blocking agents (e.g., pancuronium) complicates anesthetic monitoring. Therefore, at least continuous monitoring of the heart rate should be used as an indication of the depth of anesthesia [7, 165]. Positive end-expiratory pressure can be established before incising the diaphragm by placing the tip of the expiratory ventilation tube, 2 cm below the surface of the water [165]. For postoperative analgesia, buprenorphine is suggested for at least three days post-operatively [164]. However, other references recommend using pure opioid agonists (e.g., morphine, fentanyl) for at least the first 48 h post-surgery. Respiratory rate and quality should be monitored to avoid opioid-related respiratory depression. Systemic opioid therapy should be continued with partial agonists (e.g., buprenorphine) [167]. NSAIDs such as meloxicam, ketoprofen, or carprofen should also be used. Intercostal nerve block (e.g., lidocaine and/or bupivacaine) injection at the intercostal incision site and two intercostal spaces, cranial and caudal to the incision site, may also be used before closing the incision. This may be used in addition to opioids [58, 168]. Signs of pain should be monitored for at least 5 days after surgery.

Sepsis

Three main types of mouse models of sepsis are 1) administration of toxins of bacteria, 2) injection of an exact amount of a purified live bacterium or fecal-derived bacteria (polymicrobial infection), and 3) surgical ligation and puncture/destruction of the intestinal tract (mainly ileum-cecum portion). These models, especially the surgical type, can induce a high level of pain for the animals and require proper analgesia and anesthesia [169,170,171,172].

For surgical modeling, inhalational anesthesia with isoflurane is recommended. A combination of ketamine/xylazine is also suggested for anesthesia followed by at least two days of buprenorphine for postoperative analgesia [173].

Although analgesia is required for septic models [172], there are concerns related to the increased death rate due to the use of analgesics in these models. This has led to low consumption of analgesics in septic model studies. In fact, only 15% of studies that used the septic model reported using analgesics [174, 175]. This concern may be truly related to NSAIDs and morphine that can elevate the level of circulating IL-6. However, other opioid analgesics such as buprenorphine have low side effects and immunosuppressive activity and should be used to control the pain [174, 176, 177]. Buprenorphine can be used subcutaneously, immediately before induction of the model, and then every 12 h for at least two days [169, 174]. Besides, it is recommended to use 50–100 μL of lidocaine or bupivacaine (0.25%) in the surgical incision site to reduce the dosage of buprenorphine by half, for the first injection [177].

Wound models

Wound models include incisional or excisional cutaneous wounds and cause somatic pain. Ketamine/xylazine is used for excisional [178, 179] and incisional wound models. In rats, it is shown that ketamine/xylazine and thiopental could alter the expression of several genes related to wound healing [180].

Medetomidine/midazolam/butorphanol (0.3 mg/kg, 4 mg/kg, and 5 mg/kg) is shown to provide surgical anesthesia identical to ketamine/xylazine anesthesia [181]. Isoflurane is used for both types of wound models [182, 183]. Pentobarbital is suggested for excisional wounds [184], though it does not provide analgesia and should be accompanied by a local analgesic, such as bupivacaine or lidocaine [185].

Postoperative analgesia for wound healing research could be provided by short-term use of systemic or topical opioids, short-term use of NSAIDs (e.g., carprofen or meloxicam), immediate use of local anesthetics at the time of wound creation, or tramadol local infiltration at the injury site [185]. These modalities have not shown any adverse effect on wound healing [185]. Other analgesics, such as acetaminophen, gabapentin, or ketamine may also be considered adjunct therapies for wound pain management [185].

Conclusions

In conclusion, a proper method of surgical anesthesia includes both the anesthetic and analgesic components. Furthermore, post-surgical analgesia should be provided for almost all surgical procedures. Methods of anesthesia and analgesia for the below procedures on mice were presented in this paper: injections and catheterizations, working with agitated animals, temporary and long-term immobilization for non-painful procedures, imaging, ophthalmic procedures, limbs procedures, tracheostomy, laparotomy (liver, bile duct, kidneys, abdominal aorta, gastrointestinal tract, testes, and ovaries), skull (brain and stroke), spinal cord, thoracotomy, sepsis, and wound models. We saw that the induction and maintenance of anesthesia in mice could be achieved concurrently. In this regard, we suggested various injectable and inhalational drugs. Although ketamine and xylazine are common drugs in mice anesthesia, their use should be limited to minor surgeries. For major surgeries one should consider the addition of acepromazine to this combination or use potent analgesics. Isoflurane is a common inhalational drug in mice. It could be used as an open-drop method for non-surgical procedures or be accurately administered using a vaporizer and ventilator. We also critically examined some of the current literature according to the quality of their anesthesia/analgesia modalities. We found that many improper modalities lack the analgesic component and only provided general anesthesia, but not surgical anesthesia. Overall, this paper provides supported discussions on the choose of anesthesia/analgesia methods according to the type of the intended procedures.

Availability of data and materials

All relevant data are presented in the article.

Abbreviations

A&A:

Anesthesia and analgesia

FDG:

Blood fluorodeoxyglucose

GI:

Gastrointestinal

IM:

Intramuscular

IOP:

Intraocular pressure

NSAIDs:

Nonsteroidal anti-inflammatory drugs

PET:

Positron emission tomography

RIRI:

Renal ischemia-reperfusion injury

SCI:

Spinal cord injury

References

  1. Uhlig C, Krause H, Koch T, Gama de Abreu M, Spieth PM. Anesthesia and monitoring in small laboratory mammals used in anesthesiology, respiratory and critical care research: a systematic review on the current reporting in top-10 impact factor ranked journals. PLoS ONE. 2015;10(8):e0134205.

    Article  Google Scholar 

  2. Herrmann K, Flecknell P. Retrospective review of anesthetic and analgesic regimens used in animal research proposals. Altex. 2019;36:65–80.

    Article  Google Scholar 

  3. Carbone L, Austin J. Pain and laboratory animals: publication practices for better data reproducibility and better animal welfare. PLoS ONE. 2016;11:e0155001.

    Article  Google Scholar 

  4. Taylor K. Reporting the implementation of the Three Rs in European primate and mouse research papers: are we making progress? Altern Lab Anim. 2010;38:495–517.

    Article  CAS  Google Scholar 

  5. Okamura N, Hasegawa M, Nakoshi Y, Iino T, Sudo A, Imanaka-Yoshida K, et al. Deficiency of tenascin-C delays articular cartilage repair in mice. Osteoarthritis Cartilage. 2010;18:839–48.

    Article  CAS  Google Scholar 

  6. Navarro KL, Huss M, Smith JC, Sharp P, Marx JO, Pacharinsak C. Mouse anesthesia: the art and science. ILAR J. 2021;62(1–2):238–73.

    Article  CAS  Google Scholar 

  7. Gargiulo S, Greco A, Gramanzini M, Esposito S, Affuso A, Brunetti A, et al. Mice anesthesia, analgesia, and care, Part I: anesthetic considerations in preclinical research. ILAR J. 2012;53:E55-69.

    Article  Google Scholar 

  8. Anonymous. Recognition and alleviation of pain in laboratory animals. Washington: National Academies Press, Institute for Laboratory Animal Research (U. S.). Committee on Recognition and Alleviation of Pain in Laboratory Animals; 2009.

    Google Scholar 

  9. Kohn DF. Anesthesia and analgesia in laboratory animals. San Diego: Academic Press; 1997.

    Google Scholar 

  10. Grimm KA, Lamont LA, Tranquilli WJ, Greene SA, Robertson SA. Veterinary anesthesia and analgesia. 5th ed. Ames: Wiley Blackwell; 2015.

    Book  Google Scholar 

  11. Sarrafzadeh-Rezaei F, Ahmadi-Noorbakhsh S. Management, anesthesia, and surgery of laboratory animals. Urmia: Urmia University Press; 2010.

    Google Scholar 

  12. Busquet F, Kleensang A, Rovida C, Herrmann K, Leist M, Hartung T. New European Union statistics on laboratory animal use—what really counts! Altex. 2020;37:167–86.

    Article  Google Scholar 

  13. Gargiulo S, Greco A, Gramanzini M, Esposito S, Affuso A, Brunetti A, et al. Mice anesthesia, analgesia, and care, part II: anesthetic considerations in preclinical imaging studies. ILAR J. 2012;53:E70-81.

    Article  Google Scholar 

  14. Dallasen RM, Bowman JD, Xu Y. Isoflurane does not cause neuroapoptosis but reduces astroglial processes in young adult mice. Med Gas Res. 2011;1:27.

    Article  CAS  Google Scholar 

  15. Eöry ML, Zanuzzi CN, Fuentealba NA, Sguazza GH, Gimeno EJ, Galosi CM, et al. Effects of different anesthetics in the Murine model of EHV-1 infection. Vet Pathol. 2013;50:849–56.

    Article  Google Scholar 

  16. Lee HT, Kim M, Kim M, Kim N, Billings FT, D’Agati VD, et al. Isoflurane protects against renal ischemia and reperfusion injury and modulates leukocyte infiltration in mice. Am J Physiol Ren Physiol. 2007;293:F713–22.

    Article  CAS  Google Scholar 

  17. Mohamed AS, Hosney M, Bassiony H, Hassanein SS, Soliman AM, Fahmy SR, et al. Sodium pentobarbital dosages for exsanguination affect biochemical, molecular and histological measurements in rats. Sci Rep. 2020;10:378.

    Article  CAS  Google Scholar 

  18. Pi Z, Lin H, Yang J. Isoflurane reduces pain and inhibits apoptosis of myocardial cells through the phosphoinositide 3-kinase/protein kinase B signaling pathway in mice during cardiac surgery. Mol Med Rep. 2018. https://doi.org/10.3892/mmr.2018.8642.

    Article  Google Scholar 

  19. Ruxanda F, Gal AF, Raţiu C, Miclăuş V, Rus V, Oana LI. Comparative immunohistochemical assessment of the effect of repetitive anesthesia with isoflurane and sevoflurane on rat liver. Braz J Anesthesiol. 2016;66:465–9.

    Article  Google Scholar 

  20. Takamura H, Ichisaka S, Watanabe K, Toigawa M, Hata Y. Effects of anesthesia on immunohistochemical detection of phosphorylated extracellular signal-regulated kinase in cerebral cortex. J Neurosci Meth. 2008;170:300–4.

    Article  CAS  Google Scholar 

  21. Bengtsson C, Eriksson M. Handling and training of mice and rats results in calmer animals during experimental procedures. Animal Welfare Institute. 2020. https://awionline.org/awi-quarterly/summer-2020/handling-and-training-mice-and-rats-results-calmer-animals-during. Accessed 2 Feb 2022.

  22. Flecknell PA. Laboratory animal anaesthesia. 4th ed. Amsterdam: Elsevier; 2016.

    Google Scholar 

  23. Flecknell PA. Laboratory animal anaesthesia. An introduction for research workers and technicians. Academic Press; 1987.

    Google Scholar 

  24. Albrecht M, Henke J, Tacke S, Markert M, Guth B. Effects of isoflurane, ketamine-xylazine and a combination of medetomidine, midazolam and fentanyl on physiological variables continuously measured by telemetry in Wistar rats. BMC Vet Res. 2014;10:198.

    Article  Google Scholar 

  25. Janssen BJA, De Celle T, Debets JJM, Brouns AE, Callahan MF, Smith TL. Effects of anesthetics on systemic hemodynamics in mice. Am J Physiol Heart Circ Physiol. 2004;287:H1618–24.

    Article  CAS  Google Scholar 

  26. Hoff J. Methods of blood collection in the mouse. Lab Anim. 2000;29:7.

    Google Scholar 

  27. NCATS Inxight Drugs—Acepromazine Maleate. https://drugs.ncats.io/drug/37862HP2OM. Accessed 2 Sept 2022.

  28. Fox JG, Anderson LC, Otto GM, Pritchett-Corning KR, Whary MT, American College of Laboratory Animal Medicine. Laboratory animal medicine. 3rd ed. Amsterdam: Elsevier; 2015.

    Google Scholar 

  29. Anonymous. Open-drop or nose cone method of isoflurane anesthesia in mice and rats | University of Kentucky Research. https://www.research.uky.edu/division-laboratory-animal-resources/open-drop-or-nose-cone-method-isoflurane-anesthesia-mice-and. Accessed 28 Feb 2022.

  30. Sandhu GS, Solorio L, Broome A-M, Salem N, Kolthammer J, Shah T, et al. Whole animal imaging. Wiley Interdiscip Rev Syst Biol Med. 2010;2:398–421.

    Article  Google Scholar 

  31. Madularu D, Mathieu AP, Kumaragamage C, Reynolds LM, Near J, Flores C, et al. A non-invasive restraining system for awake mouse imaging. J Neurosci Methods. 2017;287:53–7.

    Article  Google Scholar 

  32. Woody C, Vaska P, Schlyer D, Pratte J-F, Junnarkar S, Park S-J, et al. Initial studies using the RatCAP conscious animal PET tomograph. Nucl Instrum Meth Phys Res A. 2007;571(1–2):14–7.

    Article  CAS  Google Scholar 

  33. Godbey T. Small animal handling, care, and anesthesia. Routledge Handbooks Online; 2016.

    Book  Google Scholar 

  34. Momosaki S, Hatano K, Kawasumi Y, Kato T, Hosoi R, Kobayashi K, et al. Rat-PET study without anesthesia: anesthetics modify the dopamine D1 receptor binding in rat brain. Synapse. 2004;54:207–13.

    Article  CAS  Google Scholar 

  35. Lee K-H, Ko B-H, Paik J-Y, Jung K-H, Choe YS, Choi Y, et al. Effects of anesthetic agents and fasting duration on 18F-FDG biodistribution and insulin levels in tumor-bearing mice. J Nucl Med. 2005;46:1531–6.

    CAS  Google Scholar 

  36. Keyaerts M, Remory I, Caveliers V, Breckpot K, Bos TJ, Poelaert J, et al. Inhibition of firefly luciferase by general anesthetics: effect on in vitro and in vivo bioluminescence imaging. PLoS ONE. 2012;7(1):e30061.

    Article  CAS  Google Scholar 

  37. Elena G, Amerio N, Ferrero P, Bay ML, Valenti J, Colucci D, et al. Effects of repetitive sevoflurane anaesthesia on immune response, select biochemical parameters and organ histology in mice. Lab Anim. 2003;37:193–203.

    Article  CAS  Google Scholar 

  38. Toyama H, Ichise M, Liow J-S, Vines DC, Seneca NM, Modell KJ, et al. Evaluation of anesthesia effects on [18F]FDG uptake in mouse brain and heart using small animal PET. Nucl Med Biol. 2004;31:251–6.

    Article  CAS  Google Scholar 

  39. Fuchs K, Kukuk D, Reischl G, Föller M, Eichner M, Reutershan J, et al. Oxygen breathing affects 3’-deoxy-3’-18F-fluorothymidine uptake in mouse models of arthritis and cancer. J Nucl Med. 2012;53:823–30.

    Article  CAS  Google Scholar 

  40. Hildebrandt IJ, Su H, Weber WA. Anesthesia and other considerations for in vivo imaging of small animals. ILAR J. 2008;49:17–26.

    Article  CAS  Google Scholar 

  41. Marr WG, Wood R, Senterfit L, Sigelman S. Effect of topical anesthetics*: on regeneration of corneal epithelium. Am J Ophthalmol. 1957;43:606–10.

    Article  CAS  Google Scholar 

  42. Aerts J, Nys J, Arckens L. A highly reproducible and straightforward method to perform in vivo ocular enucleation in the mouse after eye opening. J Vis Exp. 2014;92:e51936.

    Google Scholar 

  43. Kanda T, Mizoguchi Y, Furumoto K, Shimizu Y, Maeta N, Furukawa T. Effect of intramuscular medetomidine administration on tear flow in rats. Vet Sci. 2020;7:42.

    Article  Google Scholar 

  44. Burnside WM, Flecknell PA, Cameron AI, Thomas AA. A comparison of medetomidine and its active enantiomer dexmedetomidine when administered with ketamine in mice. BMC Vet Res. 2013;9:48.

    Article  Google Scholar 

  45. Cruz JI, Loste JM, Burzaco OH. Observations on the use of medetomidine/ketamine and its reversal with atipamezole for chemical restraint in the mouse. Lab Anim. 1998;32:18–22.

    Article  CAS  Google Scholar 

  46. Sun YJ, Lin C-H, Wu M-R, Lee SH, Yang J, Kunchur CR, et al. An intravitreal implant injection method for sustained drug delivery into mouse eyes. Cell Rep Methods. 2021;1:100125.

    Article  CAS  Google Scholar 

  47. Aihara M, Lindsey JD, Weinreb RN. Experimental mouse ocular hypertension: establishment of the model. Investig Ophthalmol Vis Sci. 2003;44:4314–20.

    Article  Google Scholar 

  48. Koehn D, Meyer KJ, Syed NA, Anderson MG. Ketamine/xylazine-induced corneal damage in mice. PLoS ONE. 2015;10:e0132804.

    Article  Google Scholar 

  49. Chatenay-Rivauday C, Bensaoula T, Ryckelynck H, Gérard C, Vidotto N, Ottlecz A, et al. Transient cataract in rodents: an isolated phenomenon induced by ketamine–xylazine? Investig Ophthalmol Vis Sci. 2004;45:354.

    Google Scholar 

  50. Calderone L, Grimes P, Shalev M. Acute reversible cataract induced by xylazine and by ketamine–xylazine anesthesia in rats and mice. Exp Eye Res. 1986;42:331–7.

    Article  CAS  Google Scholar 

  51. Yardeni T, Eckhaus M, Morris HD, Huizing M, Hoogstraten-Miller S. Retro-orbital injections in mice. Lab Anim (NY). 2011;40:155–60.

    Article  Google Scholar 

  52. Reichel MB, Cordeiro MF, Alexander RA, Cree IA, Bhattacharya SS, Khaw PT. New model of conjunctival scarring in the mouse eye. Br J Ophthalmol. 1998;82:1072–7.

    Article  CAS  Google Scholar 

  53. Flecknell PA, Mitchell M. Midazolam and fentanyl-fluanisone: assessment of anaesthetic effects in laboratory rodents and rabbits. Lab Anim. 1984;18:143–6.

    Article  CAS  Google Scholar 

  54. Turner PV, Albassam MA. Susceptibility of rats to corneal lesions after injectable anesthesia. Comp Med. 2005;55:175–82.

    CAS  Google Scholar 

  55. Lee Y-J, Kim M, Lee J-Y, Jung S-H, Jeon H-Y, Lee SA, et al. The benzodiazepine anesthetic midazolam prevents hyperglycemia-induced microvascular leakage in the retinas of diabetic mice. FASEB J. 2018;32:6089–99.

    Article  CAS  Google Scholar 

  56. Qiu Y, Yang H, Lei B. Effects of three commonly used anesthetics on intraocular pressure in mouse. Curr Eye Res. 2014;39:365–9.

    Article  CAS  Google Scholar 

  57. Tkatchenko TV, Tkatchenko AV. Ketamine–xylazine anesthesia causes hyperopic refractive shift in mice. J Neurosci Methods. 2010;193:67–71.

    Article  CAS  Google Scholar 

  58. Fish R, Danneman PJ, Brown M, Karas A. Anesthesia and analgesia in laboratory animals. Academic Press; 2011.

    Google Scholar 

  59. Goto T, Fukuyama N, Aki A, Kanabuchi K, Kimura K, Taira H, et al. Search for appropriate experimental methods to create stable hind-limb ischemia in mouse. Tokai J Exp Clin Med. 2006;31:128–32.

    Google Scholar 

  60. Lee JJ, Arpino JM, Yin H, Nong Z, Szpakowski A, Hashi AA, et al. Systematic interrogation of angiogenesis in the ischemic mouse hind limb. Arterioscler Thromb Vasc Biol. 2020;40:2454–67.

    Article  CAS  Google Scholar 

  61. Brenes RA, Jadlowiec CC, Bear M, Hashim P, Protack CD, Li X, et al. Toward a mouse model of hind limb ischemia to test therapeutic angiogenesis. J Vasc Surg. 2012;56:1669–79.

    Article  Google Scholar 

  62. Aref Z, de Vries MR, Quax PHA. Variations in surgical procedures for inducing hind limb ischemia in mice and the impact of these variations on neovascularization assessment. Int J Mol Sci. 2019;20(15):3704.

    Article  CAS  Google Scholar 

  63. Parikh PP, Castilla D, Lassance-Soares RM, Shao H, Regueiro M, Li Y, et al. A reliable mouse model of hind limb gangrene. Ann Vasc Surg. 2018;48:222–32.

    Article  Google Scholar 

  64. Drysch M, Wallner C, Schmidt SV, Reinkemeier F, Wagner JM, Lehnhardt M, et al. An optimized low-pressure tourniquet murine hind limb ischemia reperfusion model: Inducing acute ischemia reperfusion injury in C57BL/6 wild type mice. PLoS ONE. 2019;14:e0210961.

    Article  CAS  Google Scholar 

  65. Oyama A, Funayama E, Hayashi T, Saito A, Yamamoto Y. A new model of acquired lymphedema in the mouse hind limb: a preliminary report. Ann Plast Surg. 2012;69:565–8.

    Article  Google Scholar 

  66. Wiinholt A, Gerke O, Dalaei F, Bučan A, Madsen CB, Sørensen JA. Quantification of tissue volume in the hindlimb of mice using microcomputed tomography images and analysing software. Sci Rep. 2020;10:8297.

    Article  CAS  Google Scholar 

  67. Racine HL, Meadows CA, Ion G, Serrat MA. Heat-induced limb length asymmetry has functional impact on weight bearing in mouse hindlimbs. Front Endocrinol (Lausanne). 2018;9:289.

    Article  Google Scholar 

  68. Rinkevich Y, Montoro DT, Muhonen E, Lo D, Hasegawa M, Marshall CD, et al. Denervation of mouse lower hind limb by sciatic and femoral nerve transection. Bio-protoc. 2016;6:e1865.

    Article  Google Scholar 

  69. Furtmüller GJ, Oh B, Grahammer J, Lin C-H, Sucher R, Fryer ML, et al. Orthotopic hind limb transplantation in the mouse. J Vis Exp. 2016;108:e53483.

    Google Scholar 

  70. Hoffmeyer MR, Scalia R, Ross CR, Jones SP, Lefer DJ. PR-39, a potent neutrophil inhibitor, attenuates myocardial ischemia-reperfusion injury in mice. Am J Physiol Heart Circ Physiol. 2000;279:H2824–8.

    Article  CAS  Google Scholar 

  71. Oosterlinck W, Vanderper A, Flameng W, Herijgers P. Glucose tolerance and left ventricular pressure–volume relationships in frequently used mouse strains. J Biomed Biotechnol. 2011;2011:281312.

    Article  Google Scholar 

  72. Vandivort TC, An D, Parks WC. An improved method for rapid intubation of the trachea in mice. J Vis Exp. 2016;108:e53771.

    Google Scholar 

  73. Feldman KS, Kim E, Czachowski MJ, Wu Y, Lo CW, Zahid M. Differential effect of anesthetics on mucociliary clearance in vivo in mice. Sci Rep. 2021;11:4896.

    Article  CAS  Google Scholar 

  74. Blichfeldt-Eckhardt MR, Ording H, Andersen C, Licht PB, Toft P. Early visceral pain predicts chronic pain after laparoscopic cholecystectomy. Pain. 2014;155:2400–7.

    Article  Google Scholar 

  75. Joris J, Thiry E, Paris P, Weerts J, Lamy M. Pain after laparoscopic cholecystectomy: characteristics and effect of intraperitoneal bupivacaine. Anesth Analg. 1995;81:379–84.

    CAS  Google Scholar 

  76. Mouton WG, Bessell JR, Otten KT, Maddern GJ. Pain after laparoscopy. Surg Endosc. 1999;13:445–8.

    Article  CAS  Google Scholar 

  77. Liu J-Y, Cai Y-Y, Ding Z-Y, Zhou Z-Y, Lv M, Liu H, et al. Characterizing fibrosis and inflammation in a partial bile duct ligation mouse model by multiparametric magnetic resonance imaging. J Magn Reson Imaging. 2022;55(6):1864–74.

    Article  Google Scholar 

  78. Canbay A, Higuchi H, Bronk SF, Taniai M, Sebo TJ, Gores GJ. Fas enhances fibrogenesis in the bile duct ligated mouse: a link between apoptosis and fibrosis. Gastroenterology. 2002;123:1323–30.

    Article  CAS  Google Scholar 

  79. Durst MS, Arras M, Palme R, Talbot SR, Jirkof P. Lidocaine and bupivacaine as part of multimodal pain management in a C57BL/6J laparotomy mouse model. Sci Rep. 2021;11:10918.

    Article  CAS  Google Scholar 

  80. Tag CG, Weiskirchen S, Hittatiya K, Tacke F, Tolba RH, Weiskirchen R. Induction of experimental obstructive cholestasis in mice. Lab Anim. 2015;49(suppl-1):70–80.

    Article  CAS  Google Scholar 

  81. Van Campenhout S, Van Vlierberghe H, Devisscher L. Common bile duct ligation as model for secondary biliary cirrhosis. In: Vinken M, editor. Experimental cholestasis research. Springer; 2019. p. 237–47.

    Google Scholar 

  82. Zhu Y, Xiao X, Li G, Bu J, Zhou W, Zhou S. Isoflurane anesthesia induces liver injury by regulating the expression of insulin-like growth factor 1. Exp Ther Med. 2017;13:1608–13.

    Article  CAS  Google Scholar 

  83. Christ DD, Satoh H, Kenna JG, Pohl LR. Potential metabolic basis for enflurane hepatitis and the apparent cross-sensitization between enflurane and halothane. Drug Metab Dispos. 1988;16:135–40.

    CAS  Google Scholar 

  84. Njoku D, Laster MJ, Gong DH, Eger EI, Reed GF, Martin JL. Biotransformation of halothane, enflurane, isoflurane, and desflurane to trifluoroacetylated liver proteins: association between protein acylation and hepatic injury. Anesth Analg. 1997;84:173–8.

    Article  CAS  Google Scholar 

  85. Kharasch ED. Adverse drug reactions with halogenated anesthetics. Clin Pharmacol Ther. 2008;84:158–62.

    Article  CAS  Google Scholar 

  86. Engelking LR, Dodman NH, Hartman G, Valdez H. Effects of isoflurane anesthesia on equine liver function. Am J Vet Res. 1984;45:616–9.

    CAS  Google Scholar 

  87. Nishiyama T, Yokoyama T, Hanaoka K. Effects of sevoflurane and isoflurane anesthesia on arterial ketone body ratio and liver function. Acta Anaesthesiol Scand. 1999;43:347–51.

    Article  CAS  Google Scholar 

  88. Arras M, Autenried P, Rettich A, Spaeni D, Rülicke T. Optimization of intraperitoneal injection anesthesia in mice: drugs, dosages, adverse effects, and anesthesia depth. Comp Med. 2001;51(5):443–56.

    CAS  Google Scholar 

  89. He S, Atkinson C, Qiao F, Chen X, Tomlinson S. Ketamine–Xylazine–Acepromazine compared with isoflurane for anesthesia during liver transplantation in rodents. J Am Assoc Lab Anim Sci. 2010;49:45–51.

    CAS  Google Scholar 

  90. Suliburk JW, Helmer KS, Gonzalez EA, Robinson EK, Mercer DW. Ketamine attenuates liver injury attributed to endotoxemia: role of cyclooxygenase-2. Surgery. 2005;138:134–40.

    Article  Google Scholar 

  91. Liles JH, Flecknell PA. The influence of buprenorphine or bupivacaine on the post-operative effects of laparotomy and bile-duct ligation in rats. Lab Anim. 1993;27:374–80.

    Article  CAS  Google Scholar 

  92. Tubbs JT, Kissling GE, Travlos GS, Goulding DR, Clark JA, King-Herbert AP, et al. Effects of buprenorphine, meloxicam, and flunixin meglumine as postoperative analgesia in mice. J Am Assoc Lab Anim Sci. 2011;50:185–91.

    CAS  Google Scholar 

  93. Tellman MW, Bahler CD, Shumate AM, Bacallao RL, Sundaram CP. Management of pain in autosomal dominant polycystic kidney disease and anatomy of renal innervation. J Urol. 2015;193:1470–8.

    Article  Google Scholar 

  94. Le Clef N, Verhulst A, D’Haese PC, Vervaet BA. Unilateral renal ischemia-reperfusion as a robust model for acute to chronic kidney injury in mice. PLoS ONE. 2016;11:e0152153.

    Article  Google Scholar 

  95. Hesketh EE, Czopek A, Clay M, Borthwick G, Ferenbach D, Kluth D, et al. Renal ischaemia reperfusion injury: a mouse model of injury and regeneration. J Vis Exp. 2014;88:51816.

    Google Scholar 

  96. Wei Q, Dong Z. Mouse model of ischemic acute kidney injury: technical notes and tricks. Am J Physiol Ren Physiol. 2012;303:F1487–94.

    Article  CAS  Google Scholar 

  97. Hesketh EE, Vernon MA, Ding P, Clay S, Borthwick G, Conway B, et al. A murine model of irreversible and reversible unilateral ureteric obstruction. J Vis Exp. 2014;94:52559.

    Google Scholar 

  98. Deng J, Clair MS, Everett C, Reitman M, Star RA. Buprenorphine given after surgery does not alter renal ischemia/reperfusion injury. Comp Med. 2000;50(6):628–32.

    CAS  Google Scholar 

  99. Wang Z, Divanyan A, Jourd’heuil FL, Goldman RD, Ridge KM, Jourd’heuil D, et al. Vimentin expression is required for the development of EMT-related renal fibrosis following unilateral ureteral obstruction in mice. Am J Physiol Ren Physiol. 2018;315:F769–80.

    Article  CAS  Google Scholar 

  100. Guarnieri M, Brayton C, DeTolla L, Forbes-McBean N, Sarabia-Estrada R, Zadnik P. Safety and efficacy of buprenorphine for analgesia in laboratory mice and rats. Lab Anim (NY). 2012;41:337–43.

    Article  Google Scholar 

  101. Stockand JD, Vallon V, Ortiz P. In vivo and ex vivo analysis of tubule function. In: Terjung R, editor. Comprehensive physiology. Hoboken: Wiley; 2012.

    Google Scholar 

  102. Rhodes GJ. Surgical preparation of rats and mice for intravital microscopic imaging of abdominal organs. Methods. 2017;128:129–38.

    Article  CAS  Google Scholar 

  103. Rieg T, Richter K, Osswald H, Vallon V. Kidney function in mice: thiobutabarbital versus chloralose anesthesia. Naunyn-Schmiedeberg’s Arch Pharmacol. 2004;370:320–3.

    Article  CAS  Google Scholar 

  104. Vogler GA. Chapter 19—Anesthesia and analgesia. In: Suckow MA, Weisbroth SH, Franklin CL, editors. The laboratory rat. 2nd ed. Burlington: Academic Press; 2006. p. 627–64.

    Chapter  Google Scholar 

  105. Puri TS, Shakaib MI, Chang A, Mathew L, Olayinka O, Minto AWM, et al. Chronic kidney disease induced in mice by reversible unilateral ureteral obstruction is dependent on genetic background. Am J Physiol Ren Physiol. 2010;298:F1024–32.

    Article  CAS  Google Scholar 

  106. Thornhill BA, Chevalier RL. Variable partial unilateral ureteral obstruction and its release in the neonatal and adult mouse. In: Michos O, editor. Kidney development: methods and protocols. Totowa: Humana Press; 2012. p. 381–92.

    Chapter  Google Scholar 

  107. Achar E, Maciel TT, Collares CF, Teixeira VPC, Schor N. Amitriptyline attenuates interstitial inflammation and ameliorates the progression of renal fibrosis. Kidney Int. 2009;75:596–604.

    Article  CAS  Google Scholar 

  108. Lorenz JN, Lasko VM, Nieman ML, Damhoff T, Prasad V, Beierwaltes WH, et al. Renovascular hypertension using a modified two-kidney, one-clip approach in mice is not dependent on the α1 or α2 Na-K-ATPase ouabain-binding site. Am J Physiol Renal Physiol. 2011;301:F615–21.

    Article  CAS  Google Scholar 

  109. Johns C, Gavras I, Handy D, Salomao A, Gavras H. Models of experimental hypertension in mice. Hypertension. 1997;28:1064–9.

    Article  Google Scholar 

  110. Fujino T, Nakagawa N, Yuhki K, Hara A, Yamada T, Takayama K, et al. Decreased susceptibility to renovascular hypertension in mice lacking the prostaglandin I2 receptor IP. J Clin Investig. 2004;114:805–12.

    Article  CAS  Google Scholar 

  111. Cavallari LH, Fashingbauer LA, Camp JR, King ST, Geenen DL. Hypertension-induced renal fibrosis and spironolactone response vary by rat strain and mineralocorticoid receptor gene expression. J Renin Angiotensin Aldosterone Syst. 2008;9:146–53.

    Article  CAS  Google Scholar 

  112. Nekooeian AA, Khalili A, Khosravi MB. Effects of short-term renovascular hypertension and type 2 diabetes on cardiac functions in rats. Iran J Med Sci. 2014;39:51–9.

    Google Scholar 

  113. Kagiyama S, Varela A, Phillips MI, Galli SM. Antisense inhibition of brain renin-angiotensin system decreased blood pressure in chronic 2-kidney, 1 clip hypertensive rats. Hypertension. 2001;37(2):371–5.

    Article  CAS  Google Scholar 

  114. Del Bigio MR, Yan HJ, Kozlowski P, Sutherland GR, Peeling J. Serial magnetic resonance imaging of rat brain after induction of renal hypertension. Stroke. 1999;30:2440–7.

    Article  Google Scholar 

  115. Nicks AM, Kesteven SH, Li M, Wu J, Chan AY, Naqvi N, et al. Pressure overload by suprarenal aortic constriction in mice leads to left ventricular hypertrophy without c-Kit expression in cardiomyocytes. Sci Rep. 2020;10:15318.

    Article  CAS  Google Scholar 

  116. Li C, Zhang YY, Frieler RA, Zheng XJ, Zhang WC, Sun XN, et al. Myeloid mineralocorticoid receptor deficiency inhibits aortic constriction-induced cardiac hypertrophy in mice. PLoS ONE. 2014;9(10):e110950.

    Article  Google Scholar 

  117. Bosch L, de Haan JJ, Bastemeijer M, van der Burg J, van der Worp E, Wesseling M, et al. The transverse aortic constriction heart failure animal model: a systematic review and meta-analysis. Heart Fail Rev. 2021;26(6):1515–24.

    Article  Google Scholar 

  118. Hao Z, Zhao Z, Berthoud H-R, Ye J. Development and verification of a mouse model for Roux-en-Y gastric bypass surgery with a small gastric pouch. PLoS ONE. 2013;8:e52922.

    Article  CAS  Google Scholar 

  119. Ha SE, Wei L, Jorgensen BG, Lee MY, Park PJ, Poudrier SM, et al. A mouse model of intestinal partial obstruction. J Vis Exp. 2018. https://doi.org/10.3791/57381.

    Article  Google Scholar 

  120. Torjman MC, Joseph JI, Munsick C, Morishita M, Grunwald Z. Effects of isoflurane on gastrointestinal motility after brief exposure in rats. Int J Pharm. 2005;294:65–71.

    Article  CAS  Google Scholar 

  121. Ailiani AC, Neuberger T, Brasseur JG, Banco G, Wang Y, Smith NB, et al. Quantifying the effects of inactin vs Isoflurane anesthesia on gastrointestinal motility in rats using dynamic magnetic resonance imaging and spatio-temporal maps. Neurogastroenterol Motil. 2014;26:1477–86.

    Article  CAS  Google Scholar 

  122. Han C, Zhang Z, Guo N, Li X, Yang M, Peng Y, et al. Effects of sevoflurane inhalation anesthesia on the intestinal microbiome in mice. Front cell infect. 2021;11:633527.

    Article  CAS  Google Scholar 

  123. Wong YW, Lam LH, Tang HC, Liang Y, Tan S, Yew DT. Intestinal and liver changes after chronic ketamine and ketamine plus alcohol treatment. Microsc Res Tech. 2012;75:1170–5.

    Article  CAS  Google Scholar 

  124. Cowan A. Buprenorphine and gastrointestinal transit in rats: effect of naloxone on the biphasic dose-response curve. Clin Exp Pharmacol Physiol. 1992;19:47–9.

    Article  CAS  Google Scholar 

  125. Dutton JW, Artwohl JE, Huang X, Fortman JD. Assessment of pain associated with the injection of sodium pentobarbital in laboratory mice (Mus musculus). J Am Assoc Lab Anim Sci. 2019;58:373–9.

    Article  Google Scholar 

  126. Chang H, Li S, Li Y, Hu H, Cheng B, Miao J, et al. Effect of sedation with dexmedetomidine or propofol on gastrointestinal motility in lipopolysaccharide-induced endotoxemic mice. BMC Anesthesiol. 2020;20:227.

    Article  CAS  Google Scholar 

  127. AVMA Guidelines for the Euthanasia of Animals: 2020 edition. 1931 N. Schaumburg, IL: American Veterinary Medical Association; 2020.

  128. Tecirlioglu RT, Hayes ES, Trounson AO. Semen collection from mice: electroejaculation. Reprod Fertil Dev. 2002;14:363.

    Article  CAS  Google Scholar 

  129. Erhardt W, Hebestedt A, Aschenbrenner G, Pichotka B, Blümel G. A comparative study with various anesthetics in mice (pentobarbitone, ketamine-xylazine, carfentanyl-etomidate). Res Exp Med (Berl). 1984;184:159–69.

    Article  CAS  Google Scholar 

  130. Alshaikh AB, Padma AM, Dehlin M, Akouri R, Song MJ, Brännström M, et al. Decellularization of the mouse ovary: comparison of different scaffold generation protocols for future ovarian bioengineering. J Ovarian Res. 2019;12:58.

    Article  Google Scholar 

  131. Souza VR, Mendes E, Casaro M, Antiorio ATFB, Oliveira FA, Ferreira CM. Description of ovariectomy protocol in mice. In: Guest PC, editor. Pre-clinical models. New York: Springer New York; 2019. p. 303–9.

    Chapter  Google Scholar 

  132. Sophocleous A, Idris AI. Ovariectomy/orchiectomy in rodents. In: Idris AI, editor. Bone research protocols. New York: Springer New York; 2019. p. 261–7.

    Chapter  Google Scholar 

  133. Byers SL, Wiles MV, Taft RA. Surgical Oocyte Retrieval (SOR): a method for collecting mature mouse oocytes without euthanasia. J Am Assoc Lab Anim Sci. 2009;48:8.

    Google Scholar 

  134. Duselis AR, Vrana PB. Retrieval of mouse oocytes. J Vis Exp. 2007;3:185.

    Google Scholar 

  135. Cho C, Michailidis V, Lecker I, Collymore C, Hanwell D, Loka M, et al. Evaluating analgesic efficacy and administration route following craniotomy in mice using the grimace scale. Sci Rep. 2019;9:359.

    Article  Google Scholar 

  136. Bielefeld P, Sierra A, Encinas JM, Maletic-Savatic M, Anderson A, Fitzsimons CP. A standardized protocol for stereotaxic intrahippocampal administration of kainic acid combined with electroencephalographic seizure monitoring in mice. Front Neurosci. 2017;11:160.

    Article  Google Scholar 

  137. Mostany R, Portera-Cailliau C. A craniotomy surgery procedure for chronic brain imaging. J Vis Exp. 2008;12:680.

    Google Scholar 

  138. Staib-Lasarzik I, Nagel N, Sebastiani A, Griemert E-V, Thal SC. Analgesic treatment limits surrogate parameters for early stress and pain response after experimental subarachnoid hemorrhage. BMC Neurosci. 2019;20:49.

    Article  Google Scholar 

  139. Kyweriga M, Sun J, Wang S, Kline R, Mohajerani MH. A large lateral craniotomy procedure for mesoscale wide-field optical imaging of brain activity. J Vis Exp. 2017;123:52642.

    Google Scholar 

  140. Augustinaite S, Kuhn B. Chronic cranial window for imaging cortical activity in head-fixed mice. STAR Protoc. 2020;1:100194.

    Article  Google Scholar 

  141. Braeuninger S, Kleinschnitz C. Rodent models of focal cerebral ischemia: procedural pitfalls and translational problems. Exp Trans Stroke Med. 2009;1:8.

    Article  Google Scholar 

  142. Jacobsen KR, Fauerby N, Raida Z, Kalliokoski O, Hau J, Johansen FF, et al. Effects of buprenorphine and meloxicam analgesia on induced cerebral ischemia in C57BL/6 male mice. Comp Med. 2013;63:105–13.

    CAS  Google Scholar 

  143. Yulug B, Cam E, Yildiz A, Kilic E. Buprenorphine does not aggrevate ischemic neuronal injury in experimental focal cerebral ischemia. J Neuropsychiatry Clin Neurosci. 2007;19:331–4.

    Article  CAS  Google Scholar 

  144. Liu Z, Liang Y, Wang H, Lu Z, Chen J, Huang Q, et al. LncRNA expression in the spinal cord modulated by minocycline in a mouse model of spared nerve injury. J Pain Res. 2017;10:2503–14.

    Article  CAS  Google Scholar 

  145. Bhalala OG, Pan L, North H, McGuire T, Kessler JA. Generation of mouse spinal cord injury. Bio Protoc. 2013;3(17):e886.

    Article  Google Scholar 

  146. Thomas AM, Kubilius MB, Holland SJ, Seidlits SK, Boehler RM, Anderson AJ, et al. Channel density and porosity of degradable bridging scaffolds on axon growth after spinal injury. Biomaterials. 2013;34:2213–20.

    Article  CAS  Google Scholar 

  147. Santiago JM, Rosas O, Torrado AI, González MM, Kalyan-Masih PO, Miranda JD. Molecular, anatomical, physiological, and behavioral studies of rats treated with buprenorphine after spinal cord injury. J Neurotrauma. 2009;26(10):1783–93. https://doi.org/10.1089/neu.2007.0502.

    Article  Google Scholar 

  148. Christoph T, Kögel B, Schiene K, Méen M, De Vry J, Friderichs E. Broad analgesic profile of buprenorphine in rodent models of acute and chronic pain. Eur J Pharmacol. 2005;507:87–98.

    Article  CAS  Google Scholar 

  149. Carbajal KS, Weinger JG, Whitman LM, Schaumburg CS, Lane TE. Surgical transplantation of mouse neural stem cells into the spinal cords of mice infected with neurotropic mouse hepatitis virus. J Vis Exp. 2011;53:e2834.

    Google Scholar 

  150. Genovese T, Mazzon E, Crisafulli C, Esposito E, Di Paola R, Muià C, et al. Effects of combination of melatonin and dexamethasone on secondary injury in an experimental mice model of spinal cord trauma. J Pineal Res. 2007;43:140–53.

    Article  CAS  Google Scholar 

  151. Takasaki I, Kurihara T, Saegusa H, Zong S, Tanabe T. Effects of glucocorticoid receptor antagonists on allodynia and hyperalgesia in mouse model of neuropathic pain. Eur J Pharmacol. 2005;524:80–3.

    Article  CAS  Google Scholar 

  152. Erhorn S. xPharm: the comprehensive pharmacology reference. Amsterdam: Elsevier; 2008.

    Google Scholar 

  153. Kuhn PL, Wrathall JR. A mouse model of graded contusive spinal cord injury. J Neurotrauma. 1998;15:125–40.

    Article  CAS  Google Scholar 

  154. Meyer RE, Fish RE. Pharmacology of injectable anesthetics, sedatives, and tranquilizers. In: Anesthesia and analgesia in laboratory animals. Elsevier; 2008. p. 27–82.

  155. Li C, Zhu X, Lee C-M, Wu Z, Cheng L. A mouse model of complete-crush transection spinal cord injury made by two operations. Ann Transl Med. 2020;8(5):210.

    Article  Google Scholar 

  156. Turtle JD, Strain MM, Aceves M, Huang Y-J, Reynolds JA, Hook MA, et al. Pain input impairs recovery after spinal cord injury: treatment with lidocaine. J Neurotrauma. 2017;34:1200–8.

    Article  Google Scholar 

  157. Xu Q. A handbook of mouse models of cardiovascular disease. Hoboken: Wiley; 2006.

    Google Scholar 

  158. Giovanoli S, Notter T, Richetto J, Labouesse MA, Vuillermot S, Riva MA, et al. Late prenatal immune activation causes hippocampal deficits in the absence of persistent inflammation across aging. J Neuroinflam. 2015;12:221.

    Article  Google Scholar 

  159. Zhang Z, van Praag H. Maternal immune activation differentially impacts mature and adult-born hippocampal neurons in male mice. Brain Behav Immun. 2015;45:60–70.

    Article  Google Scholar 

  160. Gage GJ, Kipke DR, Shain W. Whole animal perfusion fixation for rodents. J Vis Exp. 2012;65:3564.

    Google Scholar 

  161. Mirabelli E, Ni L, Li L, Acioglu C, Heary RF, Elkabes S. Pathological pain processing in mouse models of multiple sclerosis and spinal cord injury: contribution of plasma membrane calcium ATPase 2 (PMCA2). J Neuroinflam. 2019;16(1):207.

    Article  Google Scholar 

  162. Hawk CT, Leary SL. Formulary for laboratory animals. 2nd ed. Ames: Iowa State University Press; 1999.

    Google Scholar 

  163. Maud P, Thavarak O, Cédrick L, Michèle B, Vincent B, Olivier P, et al. Evidence for the use of isoflurane as a replacement for chloral hydrate anesthesia in experimental stroke: an ethical issue. Biomed Res Int. 2014;2014:802539.

    Article  Google Scholar 

  164. Borst O, Ochmann C, Schönberger T, Jacoby C, Stellos K, Seizer P, et al. Methods employed for induction and analysis of experimental myocardial infarction in mice. Cell Physiol Biochem. 2011;28:1–12.

    Article  CAS  Google Scholar 

  165. Bacmeister L, Segin S, Medert R, Lindner D, Freichel M, Camacho Londoño JE. Assessment of PEEP-ventilation and the time point of parallel-conductance determination for pressure-volume analysis under β-adrenergic stimulation in mice. Front Cardiovasc Med. 2019;6:36.

    Article  Google Scholar 

  166. Pacher P, Nagayama T, Mukhopadhyay P, Bátkai S, Kass DA. Measurement of cardiac function using pressure–volume conductance catheter technique in mice and rats. Nat Protoc. 2008;3:1422–34.

    Article  CAS  Google Scholar 

  167. Faller KME, McAndrew DJ, Schneider JE, Lygate CA. Refinement of analgesia following thoracotomy and experimental myocardial infarction using the Mouse Grimace Scale. Exp Physiol. 2015;100:164–72.

    Article  CAS  Google Scholar 

  168. Orton EC, Monnet E. Small animal thoracic surgery. Wiley; 2017.

    Book  Google Scholar 

  169. Lilley E, Armstrong R, Clark N, Gray P, Hawkins P, Mason K, et al. Refinement of animal models of sepsis and septic shock. Shock. 2015;43:304–16.

    Article  CAS  Google Scholar 

  170. Marshall JC, Deitch E, Moldawer LL, Opal S, Redl H, van der Poll T. Preclinical models of shock and sepsis: what can they tell us? Shock. 2005;24:1–6.

    Article  Google Scholar 

  171. Mai S, Khan M, Liaw P, Fox-Robichaud A. Experimental sepsis models. In: Sepsis—an ongoing and significant challenge. IntechOpen; 2012.

  172. Korneev K. Mouse models of sepsis and septic shock. Mol Biol. 2019;53:704–17.

    Article  CAS  Google Scholar 

  173. Rittirsch D, Huber-Lang MS, Flierl MA, Ward PA. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc. 2009;4:31–6.

    Article  CAS  Google Scholar 

  174. Carpenter KC, Hakenjos JM, Fry CD, Nemzek JA. The influence of pain and analgesia in rodent models of sepsis. Comp med. 2019;69:546–54.

    Article  CAS  Google Scholar 

  175. Bara M, Joffe AR. The ethical dimension in published animal research in critical care: the public face of science. Crit care. 2014;18(1):R15.

    Article  Google Scholar 

  176. Cotroneo TM, Hugunin K, Shuster KA, Hwang HJ, Kakaraparthi BN, Nemzek-Hamlin JA. Effects of buprenorphine on a cecal ligation and puncture model in C57BL/6 mice. J Am Assoc Lab Anim Sci. 2012;51:357–65.

    CAS  Google Scholar 

  177. Chen W, Brenner M, Aziz M, Chavan SS, Deutschman CS, Diamond B, et al. Buprenorphine markedly elevates a panel of surrogate markers in a murine model of sepsis. Shock. 2019;52:550.

    Article  CAS  Google Scholar 

  178. Ahmadi-Noorbakhsh S, Sarrafzadeh-Rezaei F, Maham M, Dalir-Naghadeh B. Simple evaluation of the wound healing effect of topical ointments made from Senecio vernalis (Asterales: Asteraceae) plant extract in rats. Bull Vet Inst Pulawy. 2011;55:117–20.

    Google Scholar 

  179. Wosgrau ACC, da S Jeremias T, Leonardi DF, Pereima MJ, Giunta GD, Trentin AG. Comparative experimental study of wound Healing in Mice: Pelnac versus Integra. PLoS ONE. 2015;10:e0120322.

    Article  Google Scholar 

  180. Altun MA, Ozaydin A, Arkan H, Demiryas S, Akbas F, Bahtiyar N, et al. Anesthesia may alter mRNA expression of certain wound healing-associated genes in dermal wound environment of the rats. Mol Biol Rep. 2019;46:2819–27.

    Article  CAS  Google Scholar 

  181. Kawai S, Takagi Y, Kaneko S, Kurosawa T. Effect of three types of mixed anesthetic agents alternate to ketamine in mice. Exp Anim. 2011;60:481–7.

    Article  CAS  Google Scholar 

  182. Ansell DM, Campbell L, Thomason HA, Brass A, Hardman MJ. A statistical analysis of murine incisional and excisional acute wound models. Wound Repair Regen. 2014;22:281–7.

    Article  Google Scholar 

  183. Wu J, Landén NX. Investigation of skin wound healing using a mouse model. In: Botchkareva NV, Westgate GE, editors. Molecular dermatology: methods and protocols. New York: Springer; 2020. p. 239–47.

    Chapter  Google Scholar 

  184. Wang X, Ge J, Tredget EE, Wu Y. The mouse excisional wound splinting model, including applications for stem cell transplantation. Nat Protoc. 2013;8:302–9.

    Article  CAS  Google Scholar 

  185. Huss MK, Felt SA, Pacharinsak C. Influence of pain and analgesia on orthopedic and wound-healing models in rats and mice. Comp Med. 2019;69:535–45.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The authors received no financial support for the research, authorship, and/or publication of this article.

Author information

Authors and Affiliations

Authors

Contributions

SAN conceptualized the study, worked on the introduction, thoracotomy, and limbs, drafted the final manuscript and edited the works of the other coauthors. BK and MD worked on cutaneous procedures; ESP worked on imaging; GB worked on laparotomy and tracheostomy; MR, MAB, and GH worked on the pharmacology of drugs; HSA worked on the introduction and sepsis; MFA worked on ophthalmic procedures, tumors, spinal canal and tail; MG worked on the gastrointestinal system; MM worked on kidney; ND worked on thoracotomy and skull; SAT worked on the spinal canal and tail; SEK worked on skull; HKJ and SMP worked on procedures of the reproductive system and biopsy; and TS worked on catheterizations. All authors contributed to manuscript preparation, have read and agreed to the published version of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Siavash Ahmadi-Noorbakhsh.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1.

Anesthetic drugs used for laboratory mice. The doses of common anesthetic drugs used for laboratory mice are presented in this supplement.

Additional file 2.

Analgesic drugs for laboratory mouse. The doses of common analgesic drugs used for laboratory mice are presented in this supplement.

Additional file 3.

Pharmacology of common anesthetic drugs in adult mice. The pharmacology of common anesthetic drugs in adult mice are discussed in this supplement.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ahmadi-Noorbakhsh, S., Farajli Abbasi, M., Ghasemi, M. et al. Anesthesia and analgesia for common research models of adult mice. Lab Anim Res 38, 40 (2022). https://doi.org/10.1186/s42826-022-00150-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s42826-022-00150-3

Keywords